AccScience Publishing / EJMO / Online First / DOI: 10.36922/EJMO025230251
ORIGINAL RESEARCH ARTICLE

Dual roles of FGF2 and PGR in lactate-driven metabolic reprogramming and immune evasion: Implications for prostate cancer outcomes

Bo Peng1† Rongbo Xue2† Xuhua Qiao2 Chundong Ji2*
Show Less
1 Department of Urology, Jianyang People’s Hospital, Chengdu, Sichuan, China
2 Department of Urology, Affiliated Hospital of Panzhihua University, Panzhihua Hospital of Integrated Traditional Chinese and Western Medicine, Clinical and Basic Research Team of Urinary System Diseases, Affiliated Hospital of Panzhihua University, Research and Innovation Platform of Panzhihua Urology, Panzhihua, Sichuan, China
†These authors contributed equally to this work.
Received: 8 June 2025 | Revised: 15 October 2025 | Accepted: 10 November 2025 | Published online: 16 December 2025
© 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: Prostate cancer (PCa) continues to present significant therapeutic challenges, particularly in advanced stages. Lactate, a pivotal metabolite within the tumor microenvironment, promotes tumor progression through mechanisms involving metabolic reprogramming and immune suppression. Fibroblast growth factor 2 (FGF2) and progesterone receptor (PGR) represent lactate metabolism-associated genes implicated in PCa pathogenesis; however, their immunomodulatory functions and prognostic relevance remain incompletely characterized.

Methods: Multi-omics data derived from public repositories (e.g., TCGA-PRAD, GEO, ImmPort) were systematically integrated to screen out genes related to lactate metabolism and immune cell infiltration. Analyses encompassed functional enrichment, immune infiltration profiling (QuanTIseq), and single-cell transcriptomics. Cox regression, coupled with least absolute shrinkage and selection operator regularization, identified FGF2 and PGR as hub genes. A clinical–molecular prognostic signature was subsequently developed and rigorously validated employing receiver operating characteristic curves, decision curve analysis, and calibration plots.

Results: FGF2 and PGR demonstrated enrichment in biological processes, including angiogenesis and phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin pathway signaling, alongside associations with immunosuppressive cell infiltration (e.g., tumor-associated macrophages [TAM] and regulatory T cells). Single-cell resolution analysis revealed predominant expression within fibroblast and macrophage populations. Although elevated co-expression levels correlated with a prolonged progression-free interval, they concurrently corresponded to increased drug resistance, as indicated by higher half-maximal inhibitory concentration values. A combined prognostic model incorporating these genes and established clinical factors demonstrated robust predictive accuracy (area under the curve = 0.85).

Conclusion: FGF2 and PGR modulate lactate-mediated immune–metabolic crosstalk within the PCa tumor microenvironment, influencing TAM polarization and regulatory T-cell activity, thereby exerting context-dependent pro- and anti-tumorigenic effects. These findings highlight that FGF2 and PGR are promising therapeutic targets for combination strategies, although further mechanistic elucidation and clinical validation are warranted.

Keywords
Lactate metabolism
Immune microenvironment
Prostate cancer prognosis
Fibroblast growth factor 2
Progesterone receptor
Funding
This study was supported by the Science and Technology Bureau of Panzhihua City (2021zx-5-2) and the Panzhihua Medical Research Center (PYYZ-2022-02) and the Health Commission of Sichuan Province Medical Science and Technology Program(24WSXT005).
Conflict of interest
The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
References
  1. Wang L, Lu B, He M, Wang Y, Wang Z, Du L. Prostate cancer incidence and mortality: Global status and temporal trends in 89 countries from 2000 to 2019. Front Public Health. 2022;10:811044. doi: 10.3389/fpubh.2022.811044

 

  1. Andersen LB, Nørgaard M, Rasmussen M, et al. Immune cell analyses of the tumor microenvironment in prostate cancer highlight infiltrating regulatory T cells and macrophages as adverse prognostic factors. J Pathol. 2021;255(2):155-165. doi: 10.1002/path.5757

 

  1. Watanabe M, Kanao K, Suzuki S, et al. Increased infiltration of CCR4mor microenvironment in prostate cancer highlight infiltrating regulatory T poor prognosis. Prostate. 2019;79(14):1658-1665. doi: 10.1002/pros.23890

 

  1. Petya A, Erika LP. Lactic acid and lactate: Revisiting the physiological roles in the tumor microenvironment. Trends Immunol. 2022;43(12):969-977. doi: 10.1016/j.it.2022.10.005

 

  1. Harmon C, Robinson MW, Hand F, et al. Lactate-mediated acidification of tumor microenvironment induces apoptosis of liver-resident NK cells in colorectal liver metastasis. Cancer Immunol Res. 2019;7(2):335-346. doi: 10.1158/2326-6066.CIR-18-0481

 

  1. Xie D, Zhu S, Bai L. Lactic acid in tumor microenvironments causes dysfunction of NKT cells by interfering with mTOR signaling. Sci China Life Sci. 2016;59(12):1290-1296. doi: 10.1007/s11427-016-0348-7

 

  1. Tu VY, Ayari A, O’Connor RS. Beyond the lactate paradox: How lactate and acidity impact T cell therapies against cancer. Antibodies (Basel). 2021;10(3):25. doi: 10.3390/antib10030025

 

  1. Barbieri L, Velica P, Gameiro PA, et al. Lactate exposure shapes the metabolic and transcriptomic profile of CD8+ T cells. Front Immunol. 2023;14:1101433. doi: 10.3389/fimmu.2023.1101433

 

  1. Zhang Y, Peng Q, Zheng J, et al. The function and mechanism of lactate and lactylation in tumor metabolism and microenvironment. Genes Dis. 2023;10(5):2029-2037. doi: 10.1016/j.gendis.2022.10.006

 

  1. Goswami KK, Banerjee S, Bose A, Baral R. Lactic acid in alternative polarization and function of macrophages in tumor microenvironment. Hum Immunol. 2022;83(5):409-417. doi: 10.1016/j.humimm.2022.02.007

 

  1. Liu H, Liang Z, Zhou C, et al. Mutant KRAS triggers functional reprogramming of tumor-associated macrophages in colorectal cancer. Signal Transduct Target Ther. 2021;6(1):144. doi: 10.1038/s41392-021-00534-2

 

  1. Zhang Y, Zhang X, Meng Y, Xu X, Zuo D. The role of glycolysis and lactate in the induction of tumor-associated macrophages immunosuppressive phenotype. Int Immunopharmacol. 2022;110:108994. doi: 10.1016/j.intimp.2022.108994

 

  1. Tuomela K, Levings MK. Acidity promotes the differentiation of immunosuppressive regulatory T cells. Eur J Immunol. 2023;53(6):2350511. doi: 10.1002/eji.202350511

 

  1. Gu J, Zhou J, Chen Q, et al. Tumor metabolite lactate promotes tumorigenesis by modulating MOESIN lactylation and enhancing TGF-β signaling in regulatory T cells. Cell Rep. 2022;39(12):110986. doi: 10.1016/j.celrep.2022.110986

 

  1. Kumagai S, Koyama S, Itahashi K, et al. Lactic acid promotes PD-1 expression in regulatory T cells in highly glycolytic tumor microenvironments. Cancer Cell. 2022;40(2):201-218. doi: 10.1016/j.ccell.2022.01.001

 

  1. Yang Y, Wu Y, Chen H, et al. Research progress on the interaction between glucose metabolic reprogramming and lactylation in tumors. Front Immunol. 2025;16:1595162. doi: 10.3389/fimmu.2025.1595162

 

  1. Boufaied N, Chetta P, Hallal T, et al. Obesogenic high-fat diet and MYC cooperate to promote lactate accumulation and tumor microenvironment remodeling in prostate cancer. Cancer Res. 2024;84(11):1834-1855. doi: 10.1158/0008-5472.CAN-23-0519

 

  1. Zhang Y, Huang Y, Hong Y, et al. Lactate acid promotes PD-1+ Tregs accumulation in the bone marrow with high tumor burden of Acute myeloid leukemia. Int Immunopharmacol. 2024;130:111765. doi: 10.1016/j.intimp.2024.111765

 

  1. Zhou J, Shao Q, Lu Y, et al. Monocarboxylate transporter upregulation in induced regulatory T cells promotes resistance to anti-PD-1 therapy in hepatocellular carcinoma patients. Front Oncol. 2022;12:960066. doi: 10.3389/fonc.2022.960066

 

  1. Wang J, Guo R, Zhang L, et al. Reversing the “cold” tumor microenvironment: The role of neoantigen vaccines in prostate cancer. J Transl Med. 2025;23(1):835. doi: 10.1186/s12967-025-06867-8

 

  1. Chen J, Ma N, Chen B, et al. Synergistic effects of immunotherapy and adjunctive therapies in prostate cancer management. Crit Rev Oncol/Hematol. 2025;207:104604. doi: 10.1016/j.critrevonc.2024.104604

 

  1. Michael P, Roversi G, Brown K, Sharifi N. Adrenal steroids and resistance to hormonal blockade of prostate and breast cancer. Endocrinology. 2023;164(3):bqac218. doi: 10.1210/endocr/bqac218

 

  1. Lai Y, Liu C, Wang S, et al. Identification of a steroid hormone-associated gene signature predicting the prognosis of prostate cancer through an integrative bioinformatics analysis. Cancers (Basel). 2022;14(6):1565. doi: 10.3390/cancers14061565

 

  1. Hu ZI, Hellmann MD, Wolchok JD, et al. Acquired resistance to immunotherapy in MMR-D pancreatic cancer. J Immunother Cancer. 2018;6(1):127. doi: 10.1186/s40425-018-0448-1

 

  1. Pacholczak-Madej R, Bartoletti M, Musacchio L, Püsküllüoglu M, Blecharz P, Lorusso D. Immunotherapy in MMR-d/MSI-H recurrent/metastatic endometrial cancer. Expert Rev Anticancer Ther. 2024;24(8):717-729. doi: 10.1080/14737140.2024.2367472

 

  1. Zhang M, Dai X, Chen G, et al. Analysis of the distribution characteristics of prostate cancer and its environmental factors in China. Environ Sci Pollut Res Int. 2023;30(11):29349-29368. doi: 10.1007/s11356-022-24266-0

 

  1. Le TK, Duong QH, Baylot V, et al. Castration-resistant prostate cancer: From uncovered resistance mechanisms to current treatments. Cancers (Basel). 2023;15(20):5047. doi: 10.3390/cancers15205047

 

  1. Rajput M, Singh R, Singh N, Singh RP. EGFR-mediated Rad51 expression potentiates intrinsic resistance in prostate cancer via EMT and DNA repair pathways. Life Sci. 2021;286:120031. doi: 10.1016/j.lfs.2021.120031

 

  1. Crowley F, Sterpi M, Buckley C, Margetich L, Handa S, Dovey Z. A review of the pathophysiological mechanisms underlying castration-resistant prostate cancer. Res Rep Urol. 2021;13:457-472. doi: 10.2147/RRU.S264722

 

  1. Guan W, Li F, Zhao Z, Zhang Z, Hu J, Zhang Y. Tumor-associated macrophage promotes the survival of cancer cells upon docetaxel chemotherapy via the CSF1/CSF1R– CXCL12/CXCR4 Axis in castration-Resistant prostate cancer. Genes (Basel). 2021;12(5):773. doi: 10.3390/genes12050773

 

  1. Li X, Mu P. The critical interplay of CAF plasticity and resistance in prostate cancer. Cancer Res. 2023;83(18): 2990-2992. doi: 10.1158/0008-5472.CAN-23-2260

 

  1. Lin J, Elkon J, Ricart B, et al. Phase I study of entinostat in combination with enzalutamide for treatment of patients with castration-resistant prostate cancer. J Clin Oncol. 2021;96. doi: 10.1200/jco.2021.39.6_suppl.96

 

  1. Sluzalska KD, Slawski J, Sochacka M, Lampart A, Otlewski J, Zakrzewska M. Intracellular partners of fibroblast growth factors 1 and 2 - implications for functions. Cytokine Growth Factor Rev. 2021;57:93-111. doi: 10.1016/j.cytogfr.2020.05.004

 

  1. Akl MR, Nagpal P, Ayoub NM, et al. Molecular and clinical significance of fibroblast growth factor 2 (FGF2/bFGF) in malignancies of solid and hematological cancers for personalized therapies. Oncotarget. 2016;7(28):44735-44762. doi: 10.18632/oncotarget.8203

 

  1. Li D, Huang P, Xia L, Leng W, Qin S. Cancer-associated fibroblasts promote gastric cancer cell proliferation by paracrine FGF2-driven ribosome biogenesis. Int Immunopharmacol. 2024;131:111836. doi: 10.1016/j.intimp.2024.111836

 

  1. Narisawa T, Naito S, Mitsuda Y, et al. Exploratory analysis on crosstalk between intra-tumor immunity and FGF/FGFR pathway in clear cell renal cell carcinoma. J Clin Oncol. 2022;40:e6525. doi: 10.1200/jco.2022.40.16_suppl.e16525

 

  1. Singh A, Mishra R, Mazumder A. Breast cancer and its therapeutic targets: A comprehensive review. Chem Biol Drug Des. 2024;103(1):e14384. doi: 10.1111/cbdd.14384

 

  1. Boonyaratanakornkit V, McGowan EM, Marquez-Garban DC, et al. Progesterone receptor signaling in the breast tumor microenvironment. Adv Exp Med Biol. 2021;1329:443-474. doi: 10.1007/978-3-030-73119-9_21

 

  1. Zhou W, Su Y, Zhang Y, Han B, Liu H, Wang X. Endothelial cells promote docetaxel resistance of prostate cancer cells by inducing ERG expression and activating Akt/mTOR signaling pathway. Front Oncol. 2020;10:584505. doi: 10.3389/fonc.2020.584505

 

  1. Huang Y, Chen W, Yu C, et al. FGF2 drives osteosarcoma metastasis through activating FGFR1-4 receptor pathway-mediated ICAM-1 expression. Biochem Pharmacol. 2023;218:115853. doi: 10.1016/j.bcp.2023.115853

 

  1. Eiro N, Fernández-Gómez JM, Gonzalez-Ruiz De León C, et al. Gene expression profile of stromal factors in cancer-associated fibroblasts from prostate cancer. Diagnostics (Basel). 2022;12(7):1605. doi: 10.3390/diagnostics12071605

 

  1. Sun Y, Malaer JD, Mathew PA. Lectin-like transcript 1 as an natural killer cell-mediated immunotherapeutic target for triple negative breast cancer and prostate cancer. J Cancer Metastasis Treat. 2019;2019(5):80. doi: 10.20517/2394-4722.2019.29

 

  1. Katzenellenbogen JA. PET imaging agents (FES, FFNP, and FDHT) for estrogen, androgen, and progesterone receptors to improve management of breast and prostate cancers by functional imaging. Cancers (Basel). 2020;12(8):2020. doi: 10.3390/cancers12082020

 

  1. Spirina LV, Kovaleva IV, Usynin EA, Goorbunov AK, Kondakova IV. Progesterone receptor expression in the benign prostatic hyperplasia and prostate cancer tissues, relation with transcription, growth factors, hormone reception and components of the AKT/mTOR signaling pathway. Asian Pac J Cancer Prev. 2020;21(2):423-429. doi: 10.31557/APJCP.2020.21.2.423

 

  1. Liao W, Sui X, Hou G, et al. Trends in estrogen and progesterone receptors in prostate cancer: A bibliometric analysis. Front Oncol. 2023;13:1111296. doi: 10.3389/fonc.2023.1111296

 

  1. Yang M, Li JC, Tao C, et al. PAQR6 upregulation is associated with AR signaling and unfavorite prognosis in prostate cancers. Biomolecules. 2021;11(9):1383. doi: 10.3390/biom11091383
Share
Back to top
Eurasian Journal of Medicine and Oncology, Electronic ISSN: 2587-196X Print ISSN: 2587-2400, Published by AccScience Publishing