1
|
Kong X, Lei L, Ma Z. LINC00704 facilitates cell proliferation, migration, and invasion via miR-323a-3p/SLC44A1 axis in epithelial ovarian cancer. Discov Oncol 2025; 16:640. [PMID: 40301147 DOI: 10.1007/s12672-025-01866-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 02/03/2025] [Indexed: 05/01/2025] Open
Abstract
Epithelial ovarian cancer (EOC) is the most common subtype of ovarian cancer and is a highly recurrent and lethal malignancy of the female reproductive system. Accumulating evidence has indicated that long non-coding RNAs (lncRNAs) play pivotal roles in tumorigenesis. Long intergenic non-protein coding RNA 704 (LINC00704) has been recognized as an oncogenic lncRNA in several malignancies. However, its specific role in EOC remains unclear. In this study, RT-qPCR revealed the upregulation of LINC00704 in EOC tissues and cell lines. CCK-8 and EdU assays showed that LINC00704 knockdown inhibited EOC cell proliferation. Flow cytometry analysis demonstrated that LINC00704 silencing induced EOC cell cycle arrest and apoptosis. Transwell assays indicated the inhibitory effects of LINC00704 silencing on EOC cell migration and invasion. Mechanistically, bioinformatics analysis, RNA pull-down, luciferase reporter and RNA immunoprecipitation assays revealed that LINC00704 upregulated SLC44A1 expression by competitively binding to miR-323a-3p. Moreover, rescue experiments showed that SLC44A1 overexpression could reverse LINC00704 silencing-mediated effects on the malignant behaviors of EOC cells. In conclusion, LINC00704 promotes EOC cell aggressiveness in vitro by regulating the miR-323a-3p/SLC44A1 axis.
Collapse
Affiliation(s)
- Xiaojuan Kong
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Hunan University of Chinese Medicine, 95 Shaoshan Middle Road, Yuhua District, Changsha City, Hunan Province, China
| | - Lei Lei
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Hunan University of Chinese Medicine, 95 Shaoshan Middle Road, Yuhua District, Changsha City, Hunan Province, China.
| | - Zhengjiao Ma
- Department of Obstetrics and Gynecology, Wancheng District Traditional Chinese Medicine Hospital in Nanyang City, Nanyang, 473000, China
| |
Collapse
|
2
|
Chen M, Cheng M, Shao C, Liang W, Tang Y, Ding F. Fulvic acid exhibits antitumor effects in ovarian cancer cells by upregulating cytochrome P450 family 1 subfamily A member 1 expression. Discov Oncol 2025; 16:523. [PMID: 40221570 PMCID: PMC11993516 DOI: 10.1007/s12672-025-02236-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 03/25/2025] [Indexed: 04/14/2025] Open
Abstract
OBJECTIVE Fulvic acid (FA), a humic substance, has various applications in agricultural (animal husbandry) and pharmaceutical industries. However, to the best of our knowledge, its antitumor effects remain unclear. This study aimed to elucidate the effects and underlying mechanisms of FA in ovarian cancer cells. METHODS To determine the half-maximal inhibitory concentration (IC50) of FA, SK-OV-3 and OVCAR3 ovarian cancer cells were exposed to various concentrations of FA. The effects of FA and expression of cytochrome P450 family 1 subfamily A member 1 (CYP1A1) on cellular proliferation, migration, and invasion were evaluated using the Cell Counting Kit-8 and transwell assays for migration and invasion. Differentially expressed messenger ribonucleic acids (mRNAs) were identified via Illumina ribonucleic acid (RNA) sequencing and verified using fluorescent quantitative reverse transcription polymerase chain reaction (qRT-PCR). CYP1A1 protein levels were measured by western blotting. RESULTS The IC50 values of FA for OVCAR3 and SK-OV-3 cells were 689.9 and 752.0 µg/ml, respectively. FA treatment suppressed cell proliferation, invasion, and migration. In FA-treated SK-OV-3 cells, 117 mRNAs were upregulated, and 342 mRNAs were downregulated, as identified by Illumina RNA sequencing. The qRT-PCR results revealed that FA upregulated CYP1A1 expression in both cell lines. CYP1A1 overexpression mimicked the effects of FA treatment on cell proliferation, invasion, and migration. Furthermore, CYP1A1 knockdown alleviated these effects induced by FA treatments. CONCLUSION FA suppressed cell proliferation, invasion, and migration and upregulated CYP1A1 expression in SK-OV-3 and OVCAR3 cells. Our results suggest that FA demonstrates antitumor effects in ovarian cancer cells through CYP1A1 regulation.
Collapse
Affiliation(s)
- Min Chen
- Shandong Agricultural University Fertilizer Science & Technology Co., Ltd, No. 249 Chuangye Road, Feicheng City, Tai'an, 271608, China
- Department of Obstetrics and Gynecology, The Affiliated Tai'an City Central Hospital of Qingdao University, Tai'an, 271000, China
- Shandong Agricultural University, Tai'an, 271018, China
| | - Ming Cheng
- Department of Gastrointestinal Surgery, The Affiliated Tai'an City Central Hospital of Qingdao University, Tai'an, 271000, China
| | - Chenchen Shao
- Department of Obstetrics and Gynecology, The Affiliated Tai'an City Central Hospital of Qingdao University, Tai'an, 271000, China
| | - Wenwen Liang
- Department of Obstetrics and Gynecology, The Affiliated Tai'an City Central Hospital of Qingdao University, Tai'an, 271000, China
| | - Yi Tang
- College of Animal Science and Technology, Shandong Agricultural University, No. 61 Daizong Street, Taishan District, Tai'an, 271018, China.
| | - Fangjun Ding
- Shandong Agricultural University Fertilizer Science & Technology Co., Ltd, No. 249 Chuangye Road, Feicheng City, Tai'an, 271608, China.
| |
Collapse
|
3
|
Alvarez Secord A, Lewin SN, Murphy CG, Cecere SC, Barquín A, Gálvez-Montosa F, Mathews CA, Konecny GE, Ray-Coquard I, Oaknin A, Rubio Pérez MJ, Bonaventura A, Diver EJ, Ayuk SM, Wang Y, Corr BR, Salutari V. The efficacy and safety of mirvetuximab soravtansine in FRα-positive, third-line and later, recurrent platinum-sensitive ovarian cancer: the single-arm phase II PICCOLO trial. Ann Oncol 2025; 36:321-330. [PMID: 39617145 DOI: 10.1016/j.annonc.2024.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 11/13/2024] [Accepted: 11/23/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND Mirvetuximab soravtansine-gynx (MIRV) is a first-in-class, folate receptor alpha (FRα)-targeting antibody-drug conjugate with United States Food and Drug Administration approval for FRα-positive platinum-resistant ovarian cancer. PICCOLO is a phase II, global, open-label, single-arm trial of MIRV as third-line or greater (≥3L) treatment in patients with FRα-positive (≥75% of cells with ≥2+ staining intensity) recurrent platinum-sensitive ovarian cancer (PSOC). PATIENTS AND METHODS Participants received MIRV (6 mg/kg adjusted ideal body weight every 3 weeks) until progressive disease (PD), unacceptable toxicity, withdrawal of consent, or death. Primary endpoint was investigator-assessed objective response rate (ORR). Key secondary endpoint was investigator-assessed duration of response (DOR). Additional endpoints included investigator-assessed progression-free survival (PFS), overall survival (OS), and safety. Analyses of subgroups by disease characteristics (e.g. platinum-free interval) and treatment history [e.g. prior bevacizumab and poly (adenosine diphosphate [ADP]-ribose) polymerase inhibitor (PARPi) treatment] were exploratory. RESULTS Seventy-nine participants were enrolled and efficacy assessable. The primary endpoint was met; ORR was 51.9% [95% confidence interval (CI) 40.4% to 63.3%]. Median DOR was 8.25 months (95% CI 5.55-10.78 months) and median PFS was 6.93 months (95% CI 5.85-9.59 months). OS was not mature at data cut-off. ORR was 45.8% (95% CI 32.7% to 59.2%) in participants with PD while on/within 30 days of prior PARPi (n = 59) and 60.0% (95% CI 14.7% to 94.7%) in those without PD with prior PARPi (n = 5). No new safety signals occurred; most common treatment-emergent adverse events (TEAEs) were gastrointestinal, neurosensory, and resolvable ocular events. TEAEs led to discontinuation in 13 participants (16%) and death in 2 participants (3%). CONCLUSIONS MIRV as ≥3L treatment in heavily pretreated recurrent FRα-positive PSOC demonstrated notable efficacy and tolerable safety, including among those with prior PD on or within 30 days of PARPi (NCT05041257).
Collapse
MESH Headings
- Humans
- Female
- Middle Aged
- Ovarian Neoplasms/drug therapy
- Ovarian Neoplasms/pathology
- Ovarian Neoplasms/mortality
- Ovarian Neoplasms/metabolism
- Folate Receptor 1/metabolism
- Folate Receptor 1/antagonists & inhibitors
- Aged
- Neoplasm Recurrence, Local/drug therapy
- Neoplasm Recurrence, Local/pathology
- Immunoconjugates/adverse effects
- Immunoconjugates/therapeutic use
- Immunoconjugates/administration & dosage
- Maytansine/analogs & derivatives
- Maytansine/adverse effects
- Maytansine/therapeutic use
- Maytansine/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/administration & dosage
- Adult
- Drug Resistance, Neoplasm/drug effects
- Progression-Free Survival
- Aged, 80 and over
Collapse
Affiliation(s)
- A Alvarez Secord
- Duke Cancer Institute, Duke University School of Medicine, Durham, USA.
| | - S N Lewin
- Holy Name Medical Center Regional Cancer Center, Teaneck, USA
| | - C G Murphy
- Bon Secours Hospital Cork, Cork, Ireland; Cancer Trials Ireland, Dublin, Ireland
| | - S C Cecere
- Department of Urology and Gynecology, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Naples, Italy; Multicenter Italian Trials in Ovarian Cancer and Gynecologic Malignancies (MITO), Naples, Italy
| | - A Barquín
- Gynecological, Genitourinary, and Skin Cancer Unit, Hospital Universitario HM Sanchinarro, Madrid, Spain
| | - F Gálvez-Montosa
- Medical Oncology Department, Hospital Universitario de Jaén, Jaén, Spain
| | - C A Mathews
- Women & Infants Hospital, Legorreta Cancer Center, The Warren Alpert Medical School of Brown University, Providence, USA
| | - G E Konecny
- Department of Medical Oncology, University of California Los Angeles Medical Center, Santa Monica, USA
| | - I Ray-Coquard
- Leon Berard Center, Lyon, France; GINECO Group, Lyon, France
| | - A Oaknin
- Medical Oncology Service, Vall d'Hebron Institute of Oncology, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - M J Rubio Pérez
- Grupo Español de Investigación en Cáncer de Ovario (GEICO), Madrid, Spain; Department of Medical Oncology, Hospital Universitario Reina Sofía, Córdoba, Spain
| | - A Bonaventura
- Newcastle Private Hospital, New Lambton Heights, Australia
| | | | | | - Y Wang
- ImmunoGen, Inc, Waltham, USA
| | - B R Corr
- Division of Gynecologic Oncology, University of Colorado Cancer Center, Aurora, USA
| | - V Salutari
- Policlinico Universitario Fondazione Agostino Gemelli IRCCS, Rome, Italy
| |
Collapse
|
4
|
Li M, Han D, Jin H. Comprehensive bioinformatics analysis of co-mutation of FLG2 and TP53 reveals prognostic effect and influences on the immune infiltration in ovarian serous cystadenocarcinoma. Transl Cancer Res 2025; 14:1282-1296. [PMID: 40104745 PMCID: PMC11912057 DOI: 10.21037/tcr-24-1596] [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: 09/03/2024] [Accepted: 12/17/2024] [Indexed: 03/20/2025]
Abstract
Background Ovarian cancer remains one of the most lethal gynecological malignancies, characterized by late-stage diagnosis and high rates of recurrence. The present study aims to explore the prognostic and immunological implications of FLG2 and TP53, the two genes exhibiting a high mutation frequency across various cancer types, in the context of ovarian serous cystadenocarcinoma (OV). Methods The study systematically analyzed and discussed the potential implications of co-mutation of FLG2 and TP53 on prognosis and immune response using a cohort of 585 ovarian cancer samples. The differentially expressed genes (DEGs) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis were performed on 300 ovarian cancer samples with RNA sequencing (RNA-seq) data. Results The co-mutation of FLG2 and TP53 was identified in the 585 ovarian cancer cohort, and the group with co-mutation exhibited improved outcomes in terms of overall survival (OS), progression-free survival (PFS), and disease-specific survival (DSS). Additionally, the co-mutation (FLG2 +/TP53 +) group demonstrated higher scores in tumor mutation burden (TMB) comparing to that of the other three groups. The score of microsatellite instability (MSI) in the co-mutant group was only higher than that of the co-wild-type (FLG2 -/TP53 -). A total of 327 DEGs were identified in both the co-mutation and non-co-mutation (NCM) groups using limma analysis in the subgroup of 300 patients with RNA-seq data. Subsequent KEGG analysis revealed that these DEGs were implicated in various biological processes, including thermogenesis, Parkinson's disease (PD), and oxidative phosphorylation signaling pathways. Additionally, the co-mutation group exhibited elevated levels of various immune cells. Furthermore, a nomogram with high predictive accuracy was developed by integrating co-mutation status with clinical characteristics. Conclusions In the context of OV, the concurrent mutation of FLG2 and TP53 not only induces immune activation, but also helps identify a subset of patients with a more favorable prognosis.
Collapse
Affiliation(s)
- Meng Li
- Department of Radiotherapy, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
| | - Dongmei Han
- Center for Precision Cancer Medicine and Translational Research, Tianjin Cancer Hospital Airport Hospital, Tianjin, China
| | - Hao Jin
- Center for Precision Cancer Medicine and Translational Research, Tianjin Cancer Hospital Airport Hospital, Tianjin, China
- Clinical Research Management Department, Tianjin Cancer Hospital Airport Hospital, Tianjin, China
| |
Collapse
|
5
|
Philips TJ, Erickson BK, Thomas SN. Opportunities for predictive proteogenomic biomarkers of drug treatment sensitivity in epithelial ovarian cancer. Front Oncol 2025; 14:1503107. [PMID: 39839766 PMCID: PMC11746003 DOI: 10.3389/fonc.2024.1503107] [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: 09/28/2024] [Accepted: 12/12/2024] [Indexed: 01/23/2025] Open
Abstract
Genomic analysis has played a significant role in the identification of driver mutations that are linked to disease progression and response to drug treatment in ovarian cancer. A prominent example is the stratification of epithelial ovarian cancer (EOC) patients with homologous recombination deficiency (HRD) characterized by mutations in DNA damage repair genes such as BRCA1/2 for treatment with PARP inhibitors. However, recent studies have shown that some epithelial ovarian tumors respond to PARP inhibitors irrespective of their HRD or BRCA mutation status. An exclusive focus on the genome overlooks the significant insight that can be gained from other biological analytes, including proteins, which carry out cellular functions. Proteogenomics is the integration of genomics, transcriptomics, epigenomics and proteomics data. This review paper provides novel insight into the role of proteogenomics as an analytical approach to identify predictive biomarkers of drug treatment response in epithelial ovarian cancer. Proteogenomic analysis can facilitate the identification of predictive biomarkers of drug treatment response, consequently greatly improving the stratification of patients with EOC for treatment towards a goal of personalized medicine.
Collapse
Affiliation(s)
- Trudy J. Philips
- Molecular Pharmacology and Therapeutics Graduate Program, University of Minnesota School of Medicine, Minneapolis, MN, United States
| | - Britt K. Erickson
- Department of Obstetrics, Gynecology and Women’s Health, University of Minnesota School of Medicine, Minneapolis, MN, United States
| | - Stefani N. Thomas
- Department of Laboratory Medicine and Pathology, University of Minnesota School of Medicine, Minneapolis, MN, United States
| |
Collapse
|
6
|
Alsheikh B, Attar R, Güleç Yilmaz S, Barham SY, Bakırezer SD. The Role of GPX1 (rs1050450) Variants in Ovarian Cancer Susceptibility Within a Turkish Population. Cancer Control 2025; 32:10732748251332441. [PMID: 40229954 PMCID: PMC12033509 DOI: 10.1177/10732748251332441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 03/05/2025] [Accepted: 03/18/2025] [Indexed: 04/16/2025] Open
Abstract
IntroductionOvarian cancer (OC) remains one of the most lethal gynecological malignancies, primarily due to challenges in early detection and the consequent poor prognosis. Genetic predisposition plays a critical role in OC development, with the Glutathione Peroxidase 1 (GPX1) gene receiving increasing attention. The GPX1 gene polymorphism rs1050450 has been implicated in various cancers, potentially through its impact on oxidative stress mechanisms.ObjectiveThis study aimed to investigate the association between the GPX1 (rs1050450) polymorphism and the risk of developing OC in a Turkish population.MethodsA retrospective case-control study was conducted involving 90 women diagnosed with OC and 90 healthy controls. Genotyping of the GPX1 (rs1050450) polymorphism was performed using real-time PCR (RT-PCR). Statistical analyses were conducted using the SPSS software, with chi-square and t-tests applied where appropriate.ResultsThe CC genotype of the GPX1 (rs1050450) polymorphism was significantly associated with a reduced risk of OC (P = 0.002; OR = 0.304; 95% CI = 0.161-0.577), whereas the TT genotype was linked to an increased risk, demonstrating a threefold elevation in susceptibility (P = 0.036; OR = 3.308; 95% CI = 1.024-10.682). Additionally, the T allele was associated with an approximately threefold increased risk of developing OC (P = 0.0002).ConclusionThese findings suggest that the GPX1 (rs1050450) polymorphism may play a significant role in OC susceptibility, with the CC genotype offering potential protective effects and the TT genotype indicating increased risk. This genetic variant may serve as a useful marker for assessing OC risk; however, further studies involving larger and more diverse populations are needed to validate these results.
Collapse
Affiliation(s)
- Baker Alsheikh
- Department of Molecular Medicine, Faculty of Medicine, Yeditepe University, Istanbul, Turkey
| | - Rukset Attar
- Department of Obstetrics and Gynecology, Faculty of Medicine, Yeditepe University Hospital, Yeditepe University, Istanbul, Turkey
| | - Seda Güleç Yilmaz
- Department of Medical Biology, Faculty of Medicine, Yeditepe University Hospital, Yeditepe University, Istanbul, Turkey
| | | | - Selvi Duman Bakırezer
- Faculty of Medicine, Yeditepe University Hospital, Yeditepe University, Istanbul, Turkey
| |
Collapse
|
7
|
Efthymiou E, Kelekis NL. Editorial for "Peritumoral MRI Radiomics Features Increase the Evaluation Efficiency for Response to Chemotherapy in Patients With Epithelial Ovarian Cancer". J Magn Reson Imaging 2024; 60:2728-2729. [PMID: 38599797 DOI: 10.1002/jmri.29360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 01/29/2024] [Indexed: 04/12/2024] Open
Affiliation(s)
- Evgenia Efthymiou
- Second Department of Radiology, General University Hospital Attikon, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos L Kelekis
- Second Department of Radiology, General University Hospital Attikon, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
8
|
Chen H, Zhou Y, Tang Y, Lan J, Lin C, Chen Q, Kuang H. Neutrophil extracellular traps in tumor progression of gynecologic cancers. Front Immunol 2024; 15:1421889. [PMID: 39555072 PMCID: PMC11563837 DOI: 10.3389/fimmu.2024.1421889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 09/30/2024] [Indexed: 11/19/2024] Open
Abstract
This article delves into the intricate interplay between tumors, particularly gynecologic malignancies, and neutrophil extracellular traps (NETs). The relationship between tumors, specifically gynecologic malignancies, and NETs is a multifaceted and pivotal area of study. Neutrophils, pivotal components of the immune system, are tasked with combating foreign invaders. NETs, intricate structures released by neutrophils, play a vital role in combating systemic infections but also play a role in non-infectious conditions such as inflammation, autoimmune diseases, and cancer. Cancer cells have the ability to attract neutrophils, creating tumor-associated neutrophils, which then stimulate the release of NETs into the tumor microenvironment. The impact of NETs within the tumor microenvironment is profound and intricate. They play a significant role in influencing cancer development and metastasis, as well as modulating tumor immune responses. Through the release of proteases and pro-inflammatory cytokines, NETs directly alter the behavior of tumor cells, increasing invasiveness and metastatic potential. Additionally, NETs can trigger epithelial-mesenchymal transition in tumor cells, a process associated with increased invasion and metastasis. The interaction between tumors and NETs is particularly critical in gynecologic malignancies such as ovarian, cervical, and endometrial cancer. Understanding the mechanisms through which NETs operate in these tumors can offer valuable insights for the development of targeted therapeutic interventions. Researchers are actively working towards harnessing this interaction to impede tumor progression and metastasis, opening up new avenues for future treatment modalities. As our understanding of the interplay between tumors and NETs deepens, it is anticipated that novel treatment strategies will emerge, potentially leading to improved outcomes for patients with gynecologic malignancies. This article provides a comprehensive overview of the latest research findings on the interaction between NETs and cancer, particularly in gynecologic tumors, serving as a valuable resource for future exploration in this field.
Collapse
Affiliation(s)
- Hong Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Ying Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Yaling Tang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Jianfa Lan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Chao Lin
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Qionghua Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Hongying Kuang
- The Second Department of Gynecology, The First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
9
|
Xu Y, Han X, You S, Zhu W, Zhang M, Lu C, He J, Yao Q. SLC45A4 is involved in malignant progression of ovarian cancer through glycolytic metabolic reprogramming. Sci Rep 2024; 14:23031. [PMID: 39363015 PMCID: PMC11450204 DOI: 10.1038/s41598-024-74249-z] [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: 06/06/2024] [Accepted: 09/24/2024] [Indexed: 10/05/2024] Open
Abstract
Tumor cells promote malignant behaviors such as proliferation, invasion, and metastasis of cancer cells through glucose metabolic reprogramming, but the role of the H-dependent sugar cotransporter SLC45A4 in regulating metabolic reprogramming in ovarian cancer (OC) remains largely unknown. This study aimed to investigate the effects of SLC45A4 silencing on the transcriptome spectrum of ovarian cancer cells (OCC), glucose uptake, lactic acid production, intracellular ATP levels, and the expression and activity of HIF-α glycolysis signaling pathway. The results showed that SLC45A4 is overexpressed in OC and its elevated expression correlates with adverse clinical outcomes in OC patients. Silencing of SLC45A4 significantly inhibited the proliferation, invasion, and metastasis of OCC by suppressing glucose uptake and glycolysis, and it also reduced the expression of HIF-α glycolysis signaling pathway in OC tissues. In vivo experiments using shRNA to knock down SLC45A4 in xenograft models in nude mice demonstrated a significant inhibition of tumor growth. These findings suggest that SLC45A4 silencing can restrain the malignant progression of OC by inhibiting glucose uptake in OCC and affecting the reprogramming of glycolytic energy metabolism, indicating that SLC45A4 may serve as a potential therapeutic target for OC intervention.
Collapse
Affiliation(s)
- Yuance Xu
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Xiahui Han
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Shijing You
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Wei Zhu
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Mingyun Zhang
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Changyu Lu
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Junqi He
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Qin Yao
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China.
| |
Collapse
|
10
|
Lin L, Zou X, Nong W, Ge Y, Li F, Luo B, Zhang Q, Xie X. The potential value of cancer-testis antigens in ovarian cancer: Prognostic markers and targets for immunotherapy. Immun Inflamm Dis 2024; 12:e1284. [PMID: 38896069 PMCID: PMC11186301 DOI: 10.1002/iid3.1284] [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: 01/23/2024] [Revised: 05/11/2024] [Accepted: 05/15/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND Tumor immunotherapy has become an important adjuvant therapy after surgery, radiotherapy, and chemotherapy. In recent years, the role of tumor-associated antigen (TAA) in tumor immunotherapy has become increasingly prominent. Cancer-testis antigen (CTA) is a kind of TAA that is highly restricted in a variety of tumors and can induce an immune response. AIMS This review article aimed to evaluate the role of CTA on the progression of ovarian cancer, its diagnostic efficacy, and the potential for immunotherapy. METHODS We analyzed publications and outlined a comprehensive of overview the regulatory mechanism, immunogenicity, clinical expression significance, tumorigenesis, and application prospects of CTA in ovarian cancer, with a particular focus on recent progress in CTA-based immunotherapy. RESULTS The expression of CTA affects the occurrence, development, and prognosis of ovarian cancer and is closely related to tumor immunity. CONCLUSION CTA can be used as a biomarker for the diagnosis and prognosis evaluation of ovarian cancer and is an ideal target for antitumor immunotherapy. These findings provide novel insights on CTA in the improvement of diagnosis and treatment for ovarian cancer. The successes, current challenges and future prospects were also discussed to portray its significant potential.
Collapse
Affiliation(s)
- Lina Lin
- Department of Histology and Embryology, School of Basic Medicine ScienceGuangxi Medical UniversityNanningGuangxiPeople's Republic of China
- Department of Obstetrics and GynecologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiPeople's Republic of China
| | - Xiaoqiong Zou
- Department of Histology and Embryology, School of Basic Medicine ScienceGuangxi Medical UniversityNanningGuangxiPeople's Republic of China
| | - Weixia Nong
- Department of Histology and Embryology, School of Basic Medicine ScienceGuangxi Medical UniversityNanningGuangxiPeople's Republic of China
| | - Yingying Ge
- Department of Histology and Embryology, School of Basic Medicine ScienceGuangxi Medical UniversityNanningGuangxiPeople's Republic of China
| | - Feng Li
- Department of Histology and Embryology, School of Basic Medicine ScienceGuangxi Medical UniversityNanningGuangxiPeople's Republic of China
| | - Bin Luo
- Department of Histology and Embryology, School of Basic Medicine ScienceGuangxi Medical UniversityNanningGuangxiPeople's Republic of China
- Education Department of Guangxi Zhuang Autonomous RegionKey Laboratory of Basic Research on Regional Diseases (Guangxi Medical University)NanningGuangxiPeople's Republic of China
| | - Qingmei Zhang
- Department of Histology and Embryology, School of Basic Medicine ScienceGuangxi Medical UniversityNanningGuangxiPeople's Republic of China
- Education Department of Guangxi Zhuang Autonomous RegionKey Laboratory of Basic Research on Regional Diseases (Guangxi Medical University)NanningGuangxiPeople's Republic of China
| | - Xiaoxun Xie
- Department of Histology and Embryology, School of Basic Medicine ScienceGuangxi Medical UniversityNanningGuangxiPeople's Republic of China
- Education Department of Guangxi Zhuang Autonomous RegionKey Laboratory of Basic Research on Regional Diseases (Guangxi Medical University)NanningGuangxiPeople's Republic of China
- Ministry of Education, Key Laboratory of Early Prevention and Treatment of Regional High Frequency Tumor (Guangxi Medical University)NanningGuangxiPeople's Republic of China
| |
Collapse
|
11
|
Lliberos C, Richardson G, Papa A. Oncogenic Pathways and Targeted Therapies in Ovarian Cancer. Biomolecules 2024; 14:585. [PMID: 38785992 PMCID: PMC11118117 DOI: 10.3390/biom14050585] [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: 04/04/2024] [Revised: 05/06/2024] [Accepted: 05/11/2024] [Indexed: 05/25/2024] Open
Abstract
Epithelial ovarian cancer (EOC) is one of the most aggressive forms of gynaecological malignancies. Survival rates for women diagnosed with OC remain poor as most patients are diagnosed with advanced disease. Debulking surgery and platinum-based therapies are the current mainstay for OC treatment. However, and despite achieving initial remission, a significant portion of patients will relapse because of innate and acquired resistance, at which point the disease is considered incurable. In view of this, novel detection strategies and therapeutic approaches are needed to improve outcomes and survival of OC patients. In this review, we summarize our current knowledge of the genetic landscape and molecular pathways underpinning OC and its many subtypes. By examining therapeutic strategies explored in preclinical and clinical settings, we highlight the importance of decoding how single and convergent genetic alterations co-exist and drive OC progression and resistance to current treatments. We also propose that core signalling pathways such as the PI3K and MAPK pathways play critical roles in the origin of diverse OC subtypes and can become new targets in combination with known DNA damage repair pathways for the development of tailored and more effective anti-cancer treatments.
Collapse
Affiliation(s)
- Carolina Lliberos
- Cancer Program, Monash Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia;
- Neil Beauglehall Department of Medical Oncology Research, Cabrini Health, Malvern, VIC 3144, Australia
| | - Gary Richardson
- Neil Beauglehall Department of Medical Oncology Research, Cabrini Health, Malvern, VIC 3144, Australia
| | - Antonella Papa
- Cancer Program, Monash Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia;
| |
Collapse
|
12
|
Thuy Lu Vo T, Hoang VH, Thi Phuong Dung P, Anh Chi N, Minh Huy V, Tung Ngo S, Thi Kim Nguyen Y, Thi Thu Hien T, Hoang TH, Thi Do Y, Hae Seo J, Tran PT. Design, synthesis and biological evaluation of novel quinazoline derivatives as immune checkpoint inhibitors. Bioorg Med Chem Lett 2024; 108:129796. [PMID: 38754563 DOI: 10.1016/j.bmcl.2024.129796] [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: 12/18/2023] [Revised: 04/30/2024] [Accepted: 05/12/2024] [Indexed: 05/18/2024]
Abstract
In this work, we report 14 novel quinazoline derivatives as immune checkpoint inhibitors, IDO1 and PD-L1. The antitumor screening of synthesized compounds on ovarian cancer cells indicated that compound V-d and V-l showed the most activity with IC50 values of about 5 μM. Intriguingly, compound V-d emerges as a stand out, triggering cell death through caspase-dependent and caspase-independent manners. More importantly, V-d presents its ability to hinder tumor sphere formation and re-sensitized cisplatin-resistant A2780 cells to cisplatin treatment. These findings suggest that compound V-d emerges as a promising lead candidate for the future development of immuno anticancer agents.
Collapse
Affiliation(s)
- Tam Thuy Lu Vo
- Department of Biochemistry, Keimyung University School of Medicine, Daegu 42601, South Korea; Faculty of Pharmacy and Nursing, Tay Do University, 68 Lo Hau Thanh My, Can Tho 900000, Viet Nam
| | - Van-Hai Hoang
- Faculty of Pharmacy, PHENIKAA University, Hanoi 12116, Viet Nam; PHENIKAA Institute for Advanced Study (PIAS), PHENIKAA University, Yen Nghia, Hadong, Hanoi 12116, Viet Nam
| | | | - Nguyen Anh Chi
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi 100000, Viet Nam
| | - Vu Minh Huy
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi 100000, Viet Nam
| | - Son Tung Ngo
- Laboratory of Biophysics, Institute for Advanced Study in Technology, Ton Duc Thang University, Ho Chi Minh City 72915, Viet Nam; Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City 72915, Viet Nam
| | - Yen Thi Kim Nguyen
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, South Korea
| | - Tran Thi Thu Hien
- Vietnam University of Traditional Medicine, 2 Tran Phu, Ha Dong, Hanoi 100000, Viet Nam
| | - Tham H Hoang
- Center for Biomedical Informatics, Vingroup Big Data Institute, 458 Minh Khai Street, Hai Ba Trung, Hanoi 100000, Viet Nam; College of Engineering and Computer Science, VinUniversity, Vinhomes Ocean Park, Gia Lam, Hanoi 100000, Viet Nam
| | - Yen Thi Do
- Department of Biochemistry, Keimyung University School of Medicine, Daegu 42601, South Korea
| | - Ji Hae Seo
- Department of Biochemistry, Keimyung University School of Medicine, Daegu 42601, South Korea.
| | - Phuong-Thao Tran
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi 100000, Viet Nam.
| |
Collapse
|
13
|
Fedorova MV, Voznesensky VI, Sosnova EA, Proskurnina EV. Activity of NAD(P)H-Oxidoreductases in Ovarian Cancer. Biomedicines 2024; 12:1052. [PMID: 38791014 PMCID: PMC11117946 DOI: 10.3390/biomedicines12051052] [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] [Revised: 05/07/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
Reactive oxygen species (ROS) play an important and controversial role in carcinogenesis. Microsomal redox chains containing NADH- and NADPH-dependent oxidoreductases are among the main sites of intracellular ROS synthesis, but their role in the oxidative balance has not been fully studied. Here, we studied the activity of cytochrome b5 reductase (CYB5R) and cytochrome P450 reductase (CYPOR) in ovarian cancer tissues and cells isolated from peritoneal fluid, along with the antioxidant capacity of peritoneal fluid. We used the developed a chemiluminescence assay based on stimulation with NADH and NADPH, which reflects the activity of CYB5R and CYPOR, respectively. The activity of CYB5R and CYPOR was significantly higher in moderately and poorly differentiated ovarian adenocarcinomas compared with well-differentiated adenocarcinomas and cystadenomas. For the chemotherapy-resistant tumors, the activity of tissue CYB5R and CYPOR was lower compared to the non-resistant tumors. In the peritoneal fluid, the antioxidant capacity significantly increased in this series, benign tumors < well-differentiated < moderately and poorly differentiated adenocarcinomas, so the antioxidant excess was observed for moderately and poorly differentiated adenocarcinomas. The antioxidant capacity of peritoneal fluid and the activity of CYB5R and CYPOR of cells isolated from peritoneal fluid were characterized by a direct moderate correlation for moderately and poorly differentiated adenocarcinomas. These results indicate the significant role of NAD(P)H oxidoreductases and the antioxidant potential of peritoneal fluid in cancer biochemistry. The parameters studied are useful for diagnostics and prognostics. The developed assay can be used to analyze CYB5R and CYPOR activity in other tissues and cells.
Collapse
Affiliation(s)
- Maria V. Fedorova
- Central Research Institute of Epidemiology, Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing, 111123 Moscow, Russia;
| | | | - Elena A. Sosnova
- Department of Obstetrics and Gynecology No. 1, Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 119048 Moscow, Russia;
| | | |
Collapse
|
14
|
Tan H, Han X, Li C, Liu W, Li K, Sheng X, Qi S. Myelopreservation with Trilaciclib in recurrent advanced ovarian cancer: a case report. Front Oncol 2024; 14:1343239. [PMID: 38764584 PMCID: PMC11099831 DOI: 10.3389/fonc.2024.1343239] [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: 11/23/2023] [Accepted: 04/04/2024] [Indexed: 05/21/2024] Open
Abstract
Ovarian cancer is a prevalent malignant tumor of the female reproductive system, often remaining concealed until it reaches an advanced stage. The standard treatment protocol includes cytoreductive surgery for ovarian cancer plus postoperative consolidation chemotherapy and maintenance therapy, although it carries a high recurrence rate. During the treatment period, chemotherapy can lead to bone marrow suppression, a condition known as Chemotherapy-Induced Myelosuppression (CIM). This suppression may necessitate dose reduction or chemotherapy treatment cycle delay. In severe cases, CIM can result in infection, fever, and potential harm to the patient's life. Here, we report a case of a female patient with ovarian malignant tumor of biochemical recurrence who treated with chemotherapy combined with Trilaciclib, following previous perioperative chemotherapy with occurrence of severe CIM. It involves an intravenous injection of Trilaciclib before chemotherapy, which significantly abates the side effects of chemotherapy, reduces the occurrence of severe CIM, improves the patients' quality of life, and decreases the economic burden of hospitalization. We hope that this retrospective analysis of the case may serve as a reference in preventing and treating severe CIM during chemotherapy in some patients with malignant tumors, ultimately benefiting more patients with tumors.
Collapse
Affiliation(s)
- Huaming Tan
- Medical College, Shantou University Medical College, Shantou, China
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
- Cancer Center, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| | - Xiuchen Han
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Chao Li
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Wenli Liu
- Medical College, Shenzhen University, Shenzhen, China
| | - Kanghong Li
- Gynecology Department, Longgang District Maternity and Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, China
| | - Xiugui Sheng
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
- Cancer Center, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| | - Shuying Qi
- Medical College, Shantou University Medical College, Shantou, China
- Department of Reproductive Medicine, Longgang Central Hospital of Shenzhen, Shenzhen, China
| |
Collapse
|
15
|
Wang J, Zheng Q, Zhao Y, Chen S, Chen L. HMGB1 enhances the migratory and invasive abilities of A2780/DDP cells by facilitating epithelial to mesenchymal transition via GSK‑3β. Exp Ther Med 2024; 27:102. [PMID: 38356665 PMCID: PMC10865443 DOI: 10.3892/etm.2024.12390] [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: 02/21/2023] [Accepted: 10/25/2023] [Indexed: 02/16/2024] Open
Abstract
The aim of the present study was to investigate the impact and mechanism of high mobility group box 1 (HMGB1) on the regulation of cell migration and invasion in A2780/DDP cisplatin-resistant ovarian cancer cells. After transfecting small interfering (si)RNA-HMGB1 into A2780/DDP cells, Transwell migration and invasion assays were conducted to assess alterations in the cell migratory and invasive abilities. Additionally, western blotting analyses were performed to examine changes in HMGB1, phosphorylated (p)-GSK-3β, GSK-3β, E-cadherin and vimentin expression levels. The results of the present study demonstrated that the migratory and invasive abilities of A2780/DDP cells were significantly higher compared with those of A2780 cells. Additionally, the expression levels of HMGB1, p-GSK-3β and the mesenchymal phenotype marker, vimentin, in A2780/DDP cells were significantly elevated relative to the levels in A2780 cells. Conversely, the expression level of the epithelial phenotype marker, E-cadherin, was markedly decreased compared with that in A2780 cells. Following transfection of A2780/DDP cells with siRNA-HMGB1, there was a significant reduction in the rate of cell migration and invasion. Simultaneously, the expression levels of HMGB1, p-GSK-3β and vimentin were downregulated while the level of E-cadherin was upregulated. It was therefore concluded that the high expression of HMGB1 in A2780/DDP cells enhanced the cell migration and invasion abilities by facilitating epithelial to mesenchymal transition via GSK-3β.
Collapse
Affiliation(s)
- Jinhua Wang
- Department of Obstetrics and Gynecology, Fujian Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Qiaomei Zheng
- Department of Obstetrics and Gynecology, Fujian Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Yanjing Zhao
- Department of Surgery, 92403 Military Hospital, Fuzhou, Fujian 350015, P.R. China
| | - Shaozhan Chen
- Department of Obstetrics and Gynecology, Fujian Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Lihong Chen
- Department of Obstetrics and Gynecology, Fujian Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| |
Collapse
|
16
|
Zhan J, Li Z, Lin C, Wang D, Yu L, Xiao X. The role of circRNAs in regulation of drug resistance in ovarian cancer. Front Genet 2023; 14:1320185. [PMID: 38152652 PMCID: PMC10751324 DOI: 10.3389/fgene.2023.1320185] [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: 10/11/2023] [Accepted: 12/04/2023] [Indexed: 12/29/2023] Open
Abstract
Ovarian cancer is one of the female reproductive system tumors. Chemotherapy is used for advanced ovarian cancer patients; however, drug resistance is a pivotal cause of chemotherapeutic failure. Hence, it is critical to explore the molecular mechanisms of drug resistance of ovarian cancer cells and to ameliorate chemoresistance. Noncoding RNAs (ncRNAs) have been identified to critically participate in drug sensitivity in a variety of human cancers, including ovarian cancer. Among ncRNAs, circRNAs sponge miRNAs and prevent miRNAs from regulation of their target mRNAs. CircRNAs can interact with DNA or proteins to modulate gene expression. In this review, we briefly describe the biological functions of circRNAs in the development and progression of ovarian cancer. Moreover, we discuss the underneath regulatory molecular mechanisms of circRNAs on governing drug resistance in ovarian cancer. Furthermore, we mention the novel strategies to overcome drug resistance via targeting circRNAs in ovarian cancer. Due to that circRNAs play a key role in modulation of drug resistance in ovarian cancer, targeting circRNAs could be a novel approach for attenuation of chemoresistance in ovarian cancer.
Collapse
Affiliation(s)
- Jun Zhan
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, Sichuan, China
| | - Zhiyi Li
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, Sichuan, China
| | - Changsheng Lin
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, Sichuan, China
| | - Dingding Wang
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, Sichuan, China
| | - Lei Yu
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, Sichuan, China
| | - Xue Xiao
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|