Abstract
Background
Dysregulation of alternative splicing (AS) has been identified as a promising target for cancer therapy. Nevertheless, the precise molecular mechanisms by which AS influences ovarian cancer (OC) progression have not yet been fully elucidated.
Methods
A comprehensive bioinformatics analysis was conducted to identify and screen core splicing factors in OC. The splicing factor EFTUD2 was found to be significantly overexpressed in clinical OC samples. Subsequent in vitro and in vivo assays elucidated the oncogenic role of EFTUD2 in OC. RNA-seq and AS events analysis were employed to determine the key downstream target regulated by EFTUD2. ASOs targeting EFTUD2 were developed for efficacy validation.
Results
EFTUD2 was identified as a critical splicing factor in the pathogenesis of OC, and EFTUD2 knockdown impeded OC tumorigenesis and progression. The EFTUD2-ASO significantly inhibited tumor growth in vivo. Mechanistically, EFTUD2 was shown to promote the malignant biological behavior of OC by facilitating the efficient splicing of DDX41 and maintaining the oncogenic expression of its functional proteins. Knockdown of DDX41 partially mitigated the EFTUD2-induced malignant progression of OC cells.
Conclusions
Our findings suggest that the EFTUD2/DDX41 axis is a viable target for OC. ASO-mediated silencing of EFTUD2 presents promising new therapeutic options for OC patients.
This is a preview of subscription content, access via your institution
Access options
Subscribe to this journal
Receive 24 print issues and online access
269,00 € per year
only 11,21 € per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout







Similar content being viewed by others
Data availability
The data that support the findings of this study are available from the corresponding author upon reasonable request.
References
Nasiri F, Farrokhi K, Safarzadeh Kozani P, Mahboubi Kancha M, Dashti Shokoohi S, Safarzadeh Kozani P. CAR-T cell immunotherapy for ovarian cancer: hushing the silent killer. Front Immunol. 2023;14:1302307.
Wei L, Li Y, Chen J, Wang Y, Wu J, Yang H, et al. Alternative splicing in ovarian cancer. Cell Commun Signal. 2024;22:507.
Armstrong DK, Alvarez RD, Backes FJ, Bakkum-Gamez JN, Barroilhet L, Behbakht K, et al. NCCN Guidelines® Insights: Ovarian Cancer, Version 3.2022. J Natl Compr Canc Netw. 2022;20:972–80.
Armstrong DK, Alvarez RD, Bakkum-Gamez JN, Barroilhet L, Behbakht K, Berchuck A, et al. Ovarian Cancer, Version 2.2020, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2021;19:191–226.
Menon U, Gentry-Maharaj A, Burnell M, Ryan A, Singh N, Manchanda R, et al. Tumour stage, treatment, and survival of women with high-grade serous tubo-ovarian cancer in UKCTOCS: an exploratory analysis of a randomised controlled trial. Lancet Oncol. 2023;24:1018–28.
Zhang M, Cheng S, Jin Y, Zhao Y, Wang Y. Roles of CA125 in diagnosis, prediction, and oncogenesis of ovarian cancer. Biochim Biophys Acta Rev Cancer. 2021;1875:188503.
Kuroki L, Guntupalli SR. Treatment of epithelial ovarian cancer. Bmj. 2020;371:m3773.
Webb PM, Jordan SJ. Global epidemiology of epithelial ovarian cancer. Nat Rev Clin Oncol. 2024;21:389–400.
Konstantinopoulos PA, Matulonis UA. Clinical and translational advances in ovarian cancer therapy. Nat Cancer. 2023;4:1239–57.
Bradley RK, Anczuków O. RNA splicing dysregulation and the hallmarks of cancer. Nat Rev Cancer. 2023;23:135–55.
Kahles A, Lehmann KV, Toussaint NC, Hüser M, Stark SG, Sachsenberg T, et al. Comprehensive Analysis of Alternative Splicing Across Tumors from 8,705 Patients. Cancer Cell. 2018;34:211–24.e6.
Xu N, Ren Y, Bao Y, Shen X, Kang J, Wang N, et al. PUF60 promotes cell cycle and lung cancer progression by regulating alternative splicing of CDC25C. Cell Rep. 2023;42:113041.
Wan L, Lin KT, Rahman MA, Ishigami Y, Wang Z, Jensen MA, et al. Splicing Factor SRSF1 Promotes Pancreatitis and KRASG12D-Mediated Pancreatic Cancer. Cancer Discov. 2023;13:1678–95.
Deng L, Liao L, Zhang YL, Yang SY, Hu SY, Andriani L, et al. SF3A2 promotes progression and cisplatin resistance in triple-negative breast cancer via alternative splicing of MKRN1. Sci Adv. 2024;10:eadj4009.
Wang Z, Wang S, Qin J, Zhang X, Lu G, Liu H, et al. Splicing factor BUD31 promotes ovarian cancer progression through sustaining the expression of anti-apoptotic BCL2L12. Nat Commun. 2022;13:6246.
Li Y, Chen Z, Peng J, Yuan C, Yan S, Yang N, et al. The splicing factor SNRPB promotes ovarian cancer progression through regulating aberrant exon skipping of POLA1 and BRCA2. Oncogene. 2023;42:2386–401.
Zhu X, Li C, Gao Y, Zhang Q, Wang T, Zhou H, et al. The feedback loop of EFTUD2/c-MYC impedes chemotherapeutic efficacy by enhancing EFTUD2 transcription and stabilizing c-MYC protein in colorectal cancer. J Exp Clin Cancer Res. 2024;43:7.
Tu M, He L, You Y, Li J, Yao N, Qu C, et al. EFTUD2 maintains the survival of tumor cells and promotes hepatocellular carcinoma progression via the activation of STAT3. Cell Death Dis. 2020;11:830.
Lv C, Li XJ, Hao LX, Zhang S, Song Z, Ji XD, et al. Over-activation of EFTUD2 correlates with tumor propagation and poor survival outcomes in hepatocellular carcinoma. Clin Transl Oncol. 2022;24:93–103.
Sato N, Maeda M, Sugiyama M, Ito S, Hyodo T, Masuda A, et al. Inhibition of SNW1 association with spliceosomal proteins promotes apoptosis in breast cancer cells. Cancer Med. 2015;4:268–77.
Hong D, Kurzrock R, Kim Y, Woessner R, Younes A, Nemunaitis J, et al. AZD9150, a next-generation antisense oligonucleotide inhibitor of STAT3 with early evidence of clinical activity in lymphoma and lung cancer. Sci Transl Med. 2015;7:314ra185.
Wei Y, Chen Z, Li Y, Song K. The splicing factor WBP11 mediates MCM7 intron retention to promote the malignant progression of ovarian cancer. Oncogene. 2024;43:1565–78.
Yang G, Yang Y, Song Z, Chen L, Liu F, Li Y, et al. Spliceosomal GTPase Eftud2 deficiency-triggered ferroptosis leads to Purkinje cell degeneration. Neuron. 2024;112:3452–69.e9.
Dvinge H, Bradley RK. Widespread intron retention diversifies most cancer transcriptomes. Genome Med. 2015;7:45.
Arna AB, Patel H, Singh RS, Vizeacoumar FS, Kusalik A, Freywald A, et al. Synthetic lethal interactions of DEAD/H-box helicases as targets for cancer therapy. Front Oncol. 2022;12:1087989.
Beyer S, Müller L, Mitter S, Keilmann L, Meister S, Buschmann C, et al. High RIG-I and EFTUD2 expression predicts poor survival in endometrial cancer. J Cancer Res Clin Oncol. 2023;149:4293–303.
Ramasamy T, Ruttala HB, Munusamy S, Chakraborty N, Kim JO. Nano drug delivery systems for antisense oligonucleotides (ASO) therapeutics. J Control Release. 2022;352:861–78.
Kim Y. Drug discovery perspectives of antisense oligonucleotides. Biomol Ther (Seoul). 2023;31:241–52.
Kim Y, Jo M, Schmidt J, Luo X, Prakash TP, Zhou T, et al. Enhanced potency of GalNAc-conjugated antisense oligonucleotides in hepatocellular cancer models. Mol Ther. 2019;27:1547–57.
Wan L, Dreyfuss G. Splicing-Correcting Therapy for SMA. Cell. 2017;170:5.
Ma WK, Voss DM, Scharner J, Costa ASH, Lin KT, Jeon HY, et al. ASO-based PKM splice-switching therapy inhibits hepatocellular carcinoma growth. Cancer Res. 2022;82:900–15.
Reilley MJ, McCoon P, Cook C, Lyne P, Kurzrock R, Kim Y, et al. STAT3 antisense oligonucleotide AZD9150 in a subset of patients with heavily pretreated lymphoma: results of a phase 1b trial. J Immunother Cancer. 2018;6:119.
Bohnsack KE, Yi S, Venus S, Jankowsky E, Bohnsack MT. Cellular functions of eukaryotic RNA helicases and their links to human diseases. Nat Rev Mol Cell Biol. 2023;24:749–69.
Moriyama M, Koshiba T, Ichinohe T. Influenza A virus M2 protein triggers mitochondrial DNA-mediated antiviral immune responses. Nat Commun. 2019;10:4624.
Cheloor Kovilakam S, Gu M, Dunn WG, Marando L, Barcena C, Nik-Zainal S, et al. Prevalence and significance of DDX41 gene variants in the general population. Blood. 2023;142:1185–92.
Makishima H, Bowman TV, Godley LA. DDX41-associated susceptibility to myeloid neoplasms. Blood. 2023;141:1544–52.
Yang F, Long N, Anekpuritanang T, Bottomly D, Savage JC, Lee T, et al. Identification and prioritization of myeloid malignancy germline variants in a large cohort of adult patients with AML. Blood. 2022;139:1208–21.
Qi Z, Yan F, Chen D, Xing W, Li Q, Zeng W, et al. Identification of prognostic biomarkers and correlations with immune infiltrates among cGAS-STING in hepatocellular carcinoma. Biosci Rep. 2020;40:BSR20202603.
Kobatake K, Ikeda K, Nakata Y, Yamasaki N, Kanai A, Sekino Y, et al. DDX41 expression is associated with tumor necrosis in clear cell renal cell carcinoma and in cooperation with VHL loss leads to worse prognosis. Urol Oncol. 2022;40:456.e9–456.e18.
An J, Luo Z, An W, Cao D, Ma J, Liu Z. Identification of spliceosome components pivotal to breast cancer survival. RNA Biol. 2021;18:833–42.
Polprasert C, Schulze I, Sekeres MA, Makishima H, Przychodzen B, Hosono N, et al. Inherited and Somatic Defects in DDX41 in Myeloid Neoplasms. Cancer Cell. 2015;27:658–70.
Middleton R, Gao D, Thomas A, Singh B, Au A, Wong JJ, et al. IRFinder: assessing the impact of intron retention on mammalian gene expression. Genome Biol. 2017;18:51.
Monteuuis G, Wong JJL, Bailey CG, Schmitz U, Rasko JEJ. The changing paradigm of intron retention: regulation, ramifications and recipes. Nucleic Acids Res. 2019;47:11497–513.
Monteuuis G, Schmitz U, Petrova V, Kearney PS, Rasko JEJ. Holding on to Junk Bonds: intron retention in cancer and therapy. Cancer Res. 2021;81:779–89.
Ge Y, Porse BT. The functional consequences of intron retention: alternative splicing coupled to NMD as a regulator of gene expression. Bioessays. 2014;36:236–43.
Zhen N, Zhu J, Mao S, Zhang Q, Gu S, Ma J, et al. Alternative Splicing of lncRNAs From SNHG Family Alters snoRNA Expression and Induces Chemoresistance in Hepatoblastoma. Cell Mol Gastroenterol Hepatol. 2023;16:735–55.
Chen Y, Xu X, Ding K, Tang T, Cai F, Zhang H, et al. TRIM25 promotes glioblastoma cell growth and invasion via regulation of the PRMT1/c-MYC pathway by targeting the splicing factor NONO. J Exp Clin Cancer Res. 2024;43:39.
Li T, Fu J, Zeng Z, Cohen D, Li J, Chen Q, et al. TIMER2.0 for analysis of tumor-infiltrating immune cells. Nucleic Acids Res. 2020;48:W509–14.
Liu J, Liu C, Ma Y, Pan X, Chu R, Yao S, et al. STING inhibitors sensitize platinum chemotherapy in ovarian cancer by inhibiting the CGAS-STING pathway in cancer-associated fibroblasts (CAFs). Cancer Lett. 2024;588:216700.
Acknowledgements
This work was supported by the Tai-Shan Scholar Program of Shandong Province, China (tstp20231248); and the Natural Science Foundation of Shandong Province (ZR2023MH183). We thank American Journal Experts (AJE) for English language editing. The authors also thank the Laboratory Animal Center of Shandong University for mouse housing and care.
Author information
Authors and Affiliations
Contributions
KS and YWL conceived and designed the study. YLL, ZSC, ZXS, YLC, and NY collected and assembled the data. YLL, ZSC, and HMX analyzed and interpreted the data. KS, YWL, YD, and NY provided administrative, technical, or material support. KS and YWL supervised the study. YLL and YWL wrote, reviewed, and revised the manuscript. All authors reviewed the results and approved the final version of the manuscript.
Corresponding authors
Ethics declarations
Competing interests
The authors declare no competing interests.
Ethics approval and consent to participate
Ethical approval was obtained from the Ethics Committee of Qilu Hospital, Shandong University (KYLL-202410-045). The animal experiment received approval from the Animal Care and Use Committee of Cheeloo College of Medicine, Shandong University (24044). All experiments were performed in compliance with relevant guidelines and regulations.
Consent for publication
The authors consent to publish the paper.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Supplementary information
Rights and permissions
Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.
About this article
Cite this article
Liu, Y., Chen, Z., Duan, Y. et al. Spliceosomal GTPase EFTUD2 mediates DDX41 intron retention to promote the malignant progression of ovarian cancer. Br J Cancer (2025). https://doi.org/10.1038/s41416-025-03079-1
Received:
Revised:
Accepted:
Published:
DOI: https://doi.org/10.1038/s41416-025-03079-1