AccScience Publishing / EJMO / Online First / DOI: 10.36922/EJMO025360379
ORIGINAL RESEARCH ARTICLE

Sulforaphane selectively inhibits glucose metabolism in PIK3CA-mutated ovarian cancer cells

Zijiao Li1† Yinli Su2† Liying Qin3 Han Wu1 Wan Fu1 Xinyu Wang1 Longyang Li1 Xuerou Wang1 Di Chen4* Ya Xie1* Linlin Li3*
Show Less
1 Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
2 Department of Obstetrics and Gynecology, Fuyang Municipal People’s Hospital, Fuyang, Anhui, China
3 Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
4 Department of Pharmacology, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
†These authors contributed equally to this work.
Received: 4 September 2025 | Revised: 25 September 2025 | Accepted: 9 October 2025 | Published online: 12 December 2025
(This article belongs to the Special Issue Tumor Immune Microenvironment and Intervention Strategies)
© 2025 by the Author(s). This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution -Noncommercial 4.0 International License (CC-by the license) ( https://creativecommons.org/licenses/by-nc/4.0/ )
Abstract

Introduction: PIK3CA mutations are prevalent in ovarian cancer (OC) and drive cancer progression by activating the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) pathway. Sulforaphane (SFN), a natural compound derived from cruciferous vegetables, exhibits antitumor effects; however, its specific mechanisms, especially those related to metabolic reprogramming in PIK3CA-mutated cancers, remain unclear.

Objective: This study investigates the alterations in glucose metabolism and the selective inhibitory effects of SFN in PIK3CA-mutated OC cells.

Methods: Clinical samples from OC patients were analyzed to determine PIK3CA mutation status and its prognostic relevance to patient outcome. PIK3CA-mutated and wild-type OC cell lines were used for in vitro analysis. Glycolysis and mitochondrial respiration were assessed using the Seahorse XF Analyzer. Gene and protein expression were analyzed through RNA sequencing, real-time quantitative polymerase chain reaction, and Western blotting. Metabolite levels were m-easured through untargeted mass spectrometry. The effects of SFN were validated in vitro and in a mouse xenograft model.

Results: PIK3CA mutations were associated with poorer progression-free survival in OC patients. PIK3CA-mutated OC cells exhibited enhanced glycolysis and tricarboxylic acid (TCA) cycle activity compared to wild-type cells. SFN selectively inhibited the proliferation of PIK3CA-mutated cells by suppressing glycolysis and the TCA cycle, downregulating key glycolytic enzymes (e.g., hexokinase 1 and 2) and reducing TCA cycle intermediates (e.g., α-ketoglutaric acid and citrate). Mechanistically, SFN inhibited the PI3K/Akt pathway, resulting in reduced Akt phosphorylation. In vivo, SFN more effectively inhibited tumor growth in mice bearing PIK3CA-mutated xenografts.

Conclusion: PIK3CA mutations enhance both glycolysis and the TCA cycle in OC. SFN selectively inhibits the growth of PIK3CA-mutated OC cells by targeting the PI3K/Akt pathway, leading to the suppression of glucose metabolism. These findings highlight the therapeutic potential of SFN for PIK3CA-mutated OC.

Keywords
PIK3CA mutation
Ovarian cancer
Glucose metabolism
Funding
This study was funded by the Natural Science Foundation of Henan (242300421273), the Beijing Science and Technology Innovation Medical Development Foundation (KC2021-JX-0186-130), the CSCO-Zai Ding Cancer Treatment Research Fund project (Y-zai2022/ms-0246), the National Outstanding Youth Science Fund Project of the National Natural Science Foundation of China, (81802770), and the Henan Provincial Health and Health Commission, Joint Co-construction Project of Medical Science and Technology Tackling Program in Henan Province (2018020056).
Conflict of interest
The authors declare no competing interests.
References
  1. Bray F, Laversanne M, Sung H, et al. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2024;74:229-263. doi: 10.3322/caac.21834

 

  1. Siegel RL, Giaquinto AN, Jemal A. Cancer statistics, 2024. CA Cancer J Clin. 2024;74:12-49. doi: 10.3322/caac.21820

 

  1. Stine ZE, Schug ZT, Salvino JM, Dang CV. Targeting cancer metabolism in the era of precision oncology. Nat Rev Drug Discov. 2022;21:141-162. doi: 10.1038/s41573-021-00339-6

 

  1. Warburg O. The metabolism of carcinoma cells. J Cancer Res. 1925;9:148-163. doi: 10.1158/jcr.1925.148

 

  1. Elia I, Haigis MC. Metabolites and the tumour microenvironment: From cellular mechanisms to systemic metabolism. Nat Metabol. 2021;3:21-32. doi: 10.1038/s42255-020-00317-z

 

  1. Hegde S, Akbar H, Wellendorf AM, et al. Inhibition of RHOA activity preserves the survival and hemostasis function of long-term cold-stored platelets. Blood. 2024;144:1732-1746. doi: 10.1182/blood.2023021453

 

  1. Xiao J, Wang S, Chen L, et al. 25-Hydroxycholesterol regulates lysosome AMP kinase activation and metabolic reprogramming to educate immunosuppressive macrophages. Immunity. 2024;57:1087-1104.e1087. doi: 10.1016/j.immuni.2024.03.021

 

  1. Ladraa S, Zerbib L, Bayard C, et al. PIK3CA gain-of-function mutation in adipose tissue induces metabolic reprogramming with Warburg-like effect and severe endocrine disruption. Sci Adv. 2022;8:eade7823.doi: 10.1126/sciadv.ade7823

 

  1. Ikenoue T, et al. Functional analysis of PIK3CA gene mutations in human colorectal cancer. Cancer Res. 2005;65:4562-4567. doi: 10.1158/0008-5472.can-04-4114

 

  1. Samuels Y, Wang Z, Bardelli A, et al. High frequency of mutations of the PIK3CA gene in human cancers. Science. 2004;304:554. doi: 10.1126/science.1096502

 

  1. Turner NC, Im SA, Saura C, et al. Inavolisib-based therapy in PIK3CA-mutated advanced breast cancer. New Engl J Med. 2024;391:1584-1596. doi: 10.1056/nejmoa2404625

 

  1. Wu Q, Ma J, Wei J, et al. FOXD1-AS1 regulates FOXD1 translation and promotes gastric cancer progression and chemoresistance by activating the PI3K/AKT/mTOR pathway. Mol Oncol. 2021;15:299-316. doi: 10.1002/1878-0261.12728

 

  1. Xu JM, Wang Y, Wang YL, et al. PIK3CA mutations contribute to acquired cetuximab resistance in patients with metastatic colorectal cancer. Clin Cancer Res. 2017;23: 4602-4616. doi: 10.1158/1078-0432.ccr-16-2738

 

  1. Su WY, Tian LY, Guo LP, Huang LQ, Gao WY. PI3K signaling-regulated metabolic reprogramming: From mechanism to application. Biochim Biophys Acta Rev Cancer. 2023;1878:188952. doi: 10.1016/j.bbcan.2023.188952

 

  1. Herms A, Colom B, Piedrafita G, et al. Organismal metabolism regulates the expansion of oncogenic PIK3CA mutant clones in normal esophagus. Nat Genet. 2024;56:2144-2157. doi: 10.1038/s41588-024-01891-8

 

  1. Fusco N, Malapelle U, Fassan M, et al. PIK3CA mutations as a molecular target for hormone receptor-positive, HER2-negative metastatic breast cancer. Front Oncol. 2021;11:644737. doi: 10.3389/fonc.2021.644737

 

  1. Vatte C, Al Amri AM, Cyrus C, et al. Helical and kinase domain mutations of PIK3CA, and their association with hormone receptor expression in breast cancer. Oncol Lett. 2019;18:2427-2433. doi: 10.3892/ol.2019.10565

 

  1. Tan N, Wong M, Nannini MA, et al. Bcl-2/Bcl-xL inhibition increases the efficacy of MEK inhibition alone and in combination with PI3 kinase inhibition in lung and pancreatic tumor models. Mol Cancer Ther. 2013;12:853-864. doi: 10.1158/1535-7163.mct-12-0949

 

  1. Mishra R, Patel H, Alanazi S, Kilroy MK, Garrett JT. PI3K inhibitors in cancer: Clinical implications and adverse effects. Int J Mol Sci. 2021;22:3464. doi: 10.3390/ijms22073464

 

  1. Shi Z, Zeng H, Zhao B, et al. Sulforaphane reverses the enhanced NSCLC metastasis by regulating the miR-7-5p/c- Myc/LDHA axis in the acidic tumor microenvironment. Phytomedicine. 2024;133:155874. doi: 10.1016/j.phymed.2024.155874

 

  1. Huang L, Wang J, Wang X, et al. Sulforaphane suppresses bladder cancer metastasis via blocking actin nucleation-mediated pseudopodia formation. Cancer Lett. 2024;601:217145. doi: 10.1016/j.canlet.2024.217145

 

  1. Li L, Ma P, Nirasawa S, Liu H. Formation, immunomodulatory activities, and enhancement of glucosinolates and sulforaphane in broccoli sprouts: A review for maximizing the health benefits to human. Crit Rev Food Sci Nutr. 2024;64:7118-7148. doi: 10.1080/10408398.2023.2181311

 

  1. Gong TT, Liu XD, Zhan ZP, Wu QJ. Sulforaphane enhances the cisplatin sensitivity through regulating DNA repair and accumulation of intracellular cisplatin in ovarian cancer cells. Exp Cell Res. 2020;393:112061. doi: 10.1016/j.yexcr.2020.112061

 

  1. Justin S, Rutz J, Maxeiner S, et al. Chronic sulforaphane administration inhibits resistance to the mTOR-inhibitor everolimus in bladder cancer cells. Int J Mol Sci. 2020;21:4026.

 

  1. Calcabrini C, Maffei F, Turrini E, Fimognari C. Sulforaphane potentiates anticancer effects of doxorubicin and cisplatin and mitigates their toxic effects. Front Pharmacol. 2020;11:567. doi: 10.3389/fphar.2020.00567

 

  1. Kallifatidis G, Labsch S, Rausch V, et al. Sulforaphane increases drug-mediated cytotoxicity toward cancer stem-like cells of pancreas and prostate. Mol Ther. 2011;19:188-195. doi: 10.1038/mt.2010.216

 

  1. Bose C, Awasthi S, Sharma R, et al. Sulforaphane potentiates anticancer effects of doxorubicin and attenuates its cardiotoxicity in a breast cancer model. PLoS One. 2018;13:e0193918. doi: 10.1371/journal.pone.0193918

 

  1. Liu P, Zhang B, Li Y, Yuan Q. Potential mechanisms of cancer prevention and treatment by sulforaphane, a natural small molecule compound of plant-derived. Mol Med. 2024;30:94. doi: 10.1186/s10020-024-00842-7

 

  1. Du Bois A, Reuss A, Pujade-Lauraine E, et al. Role of surgical outcome as prognostic factor in advanced epithelial ovarian cancer: A combined exploratory analysis of 3 prospectively randomized phase 3 multicenter trials: By the arbeitsgemeinschaft gynaekologische onkologie studiengruppe ovarialkarzinom (AGO-OVAR) and the Groupe d’Investigateurs nationaux pour les etudes des cancers de l’Ovaire (GINECO). Cancer. 2009;115:1234-1244. doi: 10.1002/cncr.24149

 

  1. Shayesteh L, Lu Y, Kuo WL, et al. PIK3CA is implicated as an oncogene in ovarian cancer. Nat Genet. 1999;21:99-102. doi: 10.1038/5042

 

  1. Tan Y, Li J, Zhao G, et al. Metabolic reprogramming from glycolysis to fatty acid uptake and beta-oxidation in platinum-resistant cancer cells. Nat Commun. 2022;13:4554. doi: 10.1038/s41467-022-32101-w

 

  1. Li X, Mak VC, Zhou Y, et al. Deregulated Gab2 phosphorylation mediates aberrant AKT and STAT3 signaling upon PIK3R1 loss in ovarian cancer. Nat Commun. 2019;10:716. doi: 10.1038/s41467-019-08574-7

 

  1. Hortobagyi GN, Stemmer SM, Burris HA, et al. Updated results from MONALEESA-2, a phase III trial of first-line ribociclib plus letrozole versus placebo plus letrozole in hormone receptor-positive, HER2-negative advanced breast cancer. Ann Oncol. 2018;29:1541-1547. doi: 10.1093/annonc/mdy155

 

  1. Yaeger R, Chatila WK, Lipsyc MD, et al. Clinical sequencing defines the genomic landscape of metastatic colorectal cancer. Cancer Cell. 2018;33:125-136.e3. doi: 10.1016/j.ccell.2017.12.004

 

  1. McNicholas M, De Cola A, Bashardanesh Z, et al. A compendium of syngeneic, transplantable pediatric high-grade glioma models reveals subtype-specific therapeutic vulnerabilities. Cancer Discov. 2023;13:1592-1615. doi: 10.1158/2159-8290.Cd-23-0004

 

  1. Vasan N, Toska E, Scaltriti M. Overview of the relevance of PI3K pathway in HR-positive breast cancer. Ann Oncol. 2019;30:x3-x11. doi: 10.1093/annonc/mdz281

 

  1. Song KW, Edgar KA, Hanan EJ, et al. RTK-dependent inducible degradation of mutant PI3Kα drives GDC-0077 (Inavolisib) efficacy. Cancer Discov. 2022;12:204-219. doi: 10.1158/2159-8290.Cd-21-0072

 

  1. Jiang W, He T, Liu S, et al. The PIK3CA E542K and E545K mutations promote glycolysis and proliferation via induction of the β-catenin/SIRT3 signaling pathway in cervical cancer. J Hematol Oncol. 2018;11:139. doi: 10.1186/s13045-018-0674-5

 

  1. Pradeep Prabhu P, Mohanty B, Lobo CL, et al. Harnessing the nutriceutics in early-stage breast cancer: Mechanisms, combinational therapy, and drug delivery. J Nanobiotechnol. 2024;22:574. doi: 10.1186/s12951-024-02815-8

 

  1. Sundaram MK, Preetha R, Haque S, et al. Dietary isothiocyanates inhibit cancer progression by modulation of epigenome. Semin Cancer Biol. 2022;83:353-376. doi: 10.1016/j.semcancer.2020.12.021

 

  1. Joković N, Pešić S, Vitorović J, et al. Glucosinolates and their hydrolytic derivatives: Promising phytochemicals with anticancer potential. Phytother Res PTR. 2024;39:10. doi: 10.1002/ptr.8419

 

  1. Tomooka F, Kaji K, Nishimura N, et al. Sulforaphane potentiates gemcitabine-mediated anti-cancer effects against intrahepatic cholangiocarcinoma by inhibiting HDAC activity. Cells. 2023;12:687. doi: 10.3390/cells12050687

 

  1. Dandawate PR, Subramaniam D, Jensen RA, Anant S. Targeting cancer stem cells and signaling pathways by phytochemicals: Novel approach for breast cancer therapy. Semin Cancer Biol. 2016;40-41:192-208. doi: 10.1016/j.semcancer.2016.09.001

 

  1. Chen X, Jiang Z, Zhou C, et al. Activation of Nrf2 by sulforaphane inhibits high glucose-induced progression of pancreatic cancer via AMPK dependent signaling. Cell Physiol Biochem. 2018;50:1201-1215. doi: 10.1159/000494547

 

  1. Jo S, Seo M, Nguyen TH, et al. Biosynthesis-encoded lipogenic Acetyl-CoA measurement using NMR reveals glucose-driven lipogenesis and glutamine’s alternative roles in kidney cancer. J Am Chem Soc. 2024;146:33753-33762. doi: 10.1021/jacs.4c11809
Share
Back to top
Eurasian Journal of Medicine and Oncology, Electronic ISSN: 2587-196X Print ISSN: 2587-2400, Published by AccScience Publishing