AccScience Publishing / TD / Online First / DOI: 10.36922/TD025190036
Cite this article
10
Download
110
Views
Journal Browser
Volume | Year
Issue
Search
News and Announcements
View All
ORIGINAL RESEARCH ARTICLE

RIBC2 expression as a potential diagnostic and prognostic biomarker associated with tumor progression and immune modulation in breast cancer

Zhuoyu Li1 Luobin Lin1 Zhiwei Liao1 Shimin Lin1 Wenmei Wu1*
Show Less
1 Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
Tumor Discovery, 025190036 https://doi.org/10.36922/TD025190036
Received: 8 May 2025 | Revised: 6 September 2025 | Accepted: 8 September 2025 | Published online: 10 October 2025
© 2025 by the Author(s). This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution 4.0 International License ( https://creativecommons.org/licenses/by/4.0/ )
Abstract

Breast cancer remains one of the deadliest diseases around the world. This study systematically investigates the expression profile and clinical significance of RIBC2 in breast cancer. By integrating multi-omics data from public databases, such as The Cancer Genome Atlas and The Genotype-Tissue Expression project, combined with bioinformatic analyses and molecular docking simulations, we demonstrated that RIBC2 is significantly overexpressed in breast cancer tissues. Elevated RIBC2 expression was positively correlated with higher T stage, advanced pathological stage, and poorer overall survival. Functional enrichment analyses indicated that RIBC2 may facilitate tumor invasion by regulating extracellular matrix remodeling and activating the mitogen-activated protein kinase signaling pathway. Multivariate Cox regression analysis confirmed high RIBC2 expression as an independent poor prognostic factor, with its inclusion slightly increasing the area under the curve of the prognostic model from 0.762 to 0.764. Moreover, immune infiltration analysis revealed that high RIBC2 expression was associated with increased infiltration of regulatory T cells and M2-polarized macrophages, indicating a potential role in shaping an immune suppressive tumor microenvironment. Spearman correlation analysis between RIBC2 and immune checkpoints showed significant positive associations only for lymphocyte activation gene 3 protein (p<0.001, absolute correlation ≈ 0.1) and human leukocyte antigen-C (p=0.045, absolute correlation ≈ 0.08), indicating RIBC2 may selectively modulate specific immunosuppressive pathways. Drug sensitivity analysis further suggested that RIBC2 overexpression may contribute to chemotherapy resistance. The methylation level of RIBC2 was found to be significantly negatively correlated with its expression, suggesting that hypermethylation in the promoter or regulatory regions may suppress gene expression through epigenetic silencing mechanisms. Furthermore, quantitative polymerase chain reaction revealed a 4.2-fold RIBC2 upregulation in triple-negative breast cancer cells (MDA-MB-231) compared to normal mammary cells (Hs578bst), exceeding levels in luminal subtypes (MCF-7). Collectively, these findings highlight RIBC2 as a promising prognostic biomarker and provide a theoretical foundation for developing combination therapeutic strategies targeting RIBC2.

Keywords
Breast cancer
RIBC2
Prognostic marker
Metastasis
Immunity
Funding
This work was supported by the Guangdong Provincial Traditional Chinese Medicine Research Project (No. 20251216) and the College Students’ Innovation and Entrepreneurship Training Program (No. S202410573014).
Conflict of interest
The authors declare they have no competing interests.
References
  1. Liu M, Li Z, Yang J, et al. Cell-specific biomarkers and targeted biopharmaceuticals for breast cancer treatment. Cell Prolif. 2016;49(4):409-20. doi: 10.1111/cpr.12266

 

  1. Barzaman K, Karami J, Zarei Z, et al. Breast cancer: Biology, biomarkers, and treatments. Int Immunopharmacol. 2020;84:106535. doi: 10.1016/j.intimp.2020.106535

 

  1. Singh DD, Yadav DK. TNBC: Potential targeting of multiple receptors for a therapeutic breakthrough, nanomedicine, and immunotherapy. Biomedicines. 2021;9(8):876. doi: 10.3390/biomedicines9080876

 

  1. Bhave MA, Quintanilha JCF, Tukachinsky H, et al. Comprehensive genomic profiling of ESR1, PIK3CA, AKT1, and PTEN in HR(+)HER2(-) metastatic breast cancer: Prevalence along treatment course and predictive value for endocrine therapy resistance in real-world practice. Breast Cancer Res Treat. 2024;207(3):599-609. doi: 10.1007/s10549-024-07376-w

 

  1. Bardia A, Cortes J, Bidard F, et al. Elacestrant in ER+, HER2- metastatic breast cancer with ESR1-mutated tumors: Subgroup analyses from the phase III EMERALD trial by prior duration of endocrine therapy plus CDK4/6 inhibitor and in clinical subgroups. Clin Cancer Res. 2024;30(19):4299- 4309. doi: 10.1158/1078-0432.CCR-24-1073

 

  1. Cheng X. A comprehensive review of HER2 in cancer biology and therapeutics. Genes (Basel). 2024;15(7):903. doi: 10.3390/genes15070903

 

  1. Hu Y, Wang C, Liang H, Li J, Yang Q. The treatment landscape of triple-negative breast cancer. Med Oncol. 2024;41(10):236. doi: 10.1007/s12032-024-02456-9

 

  1. Swain SM, Shastry M, Hamilton E. Targeting HER2-positive breast cancer: Advances and future directions. Nat Rev Drug Discov. 2023;22(2):101-126. doi: 10.1038/s41573-022-00579-0

 

  1. Tang Y, Olufemi L, Wang M, Nie D. Role of Rho GTPases in breast cancer. Front Biosci. 2008;13:759-76. doi: 10.2741/2718

 

  1. Humphries B, Wang Z, Yang C. Rho GTPases: Big players in breast cancer initiation, metastasis and therapeutic responses. Cells. 2020;9(10):2167. doi: 10.3390/cells9102167

 

  1. Ghoroghi S, Mary B, Larnicol A, et al. Ral GTPases promote breast cancer metastasis by controlling biogenesis and organ targeting of exosomes. Elife. 2021;10:e61539. doi: 10.7554/eLife.61539

 

  1. Xie J, Yang A, Liu Q, et al. Single-cell RNA sequencing elucidated the landscape of breast cancer brain metastases and identified ILF2 as a potential therapeutic target. Cell Prolif. 2024;57(11):e13697. doi: 10.1111/cpr.13697

 

  1. Xie J, Deng X, Xie Y, et al. Multi-omics analysis of disulfidptosis regulators and therapeutic potential reveals glycogen synthase 1 as a disulfidptosis triggering target for triple-negative breast cancer. MedComm (2020). 2024;5(3):e502. doi: 10.1002/mco2.502

 

  1. Sun C, Wang A, Zhou Y, et al. Spatially resolved multi-omics highlights cell-specific metabolic remodeling and interactions in gastric cancer. Nat Commun. 2023;14(1):2692. doi: 10.1038/s41467-023-38360-5

 

  1. Yousuf S, Qiu M, Voith Von Voithenberg L, et al. Spatially resolved multi-omics single-cell analyses inform mechanisms of immune dysfunction in pancreatic cancer. Gastroenterology. 2023;165(4):891-908.e14. doi: 10.1053/j.gastro.2023.05.036

 

  1. Zhou Y, Lin Z, Xie S, et al. Interplay of chronic obstructive pulmonary disease and colorectal cancer development: Unravelling the mediating role of fatty acids through a comprehensive multi-omics analysis. J Transl Med. 2023;21(1):587. doi: 10.1186/s12967-023-04278-1

 

  1. Liu Z, Chen N, Li R, Ma Y, Qiayimaerdan A, Ma CL. WGCNA reveals a biomarker for cancer-associated fibroblasts to predict prognosis in cervical cancer. J Chin Med Assoc. 2024;87(9):885-897. doi: 10.1097/JCMA.0000000000001129

 

  1. He Z, Chen Z, Tan M, et al. A review on methods for diagnosis of breast cancer cells and tissues. Cell Prolif. 2020;53(7):e12822. doi: 10.1111/cpr.12822

 

  1. Wang T, Dai L, Shen S, et al. Comprehensive molecular analyses of a macrophage-related gene signature with regard to prognosis, immune features, and biomarkers for immunotherapy in hepatocellular carcinoma based on WGCNA and the LASSO algorithm. Front Immunol. 2022;13:843408. doi: 10.3389/fimmu.2022.843408

 

  1. Xiong M, Wang Y, Guo W, et al. A common variant rs2272804 in the 5’UTR of RIBC2 inhibits downstream gene expression by creating an upstream open reading frame. Eur Rev Med Pharmacol Sci. 2020;24(7):3839-3848. doi: 10.26355/eurrev_202004_20851

 

  1. Katsuma K, Shimada K, Tonai S, et al. The absence of both RIBC1 and RIBC2 induces decreased sperm motility and litter size in male mice. Andrology. 2025.doi: 10.1111/andr.70045

 

  1. Kwon KY, Jeong H, Jang DG, Kwon T, Park TJ. Ckb and Ybx2 interact with Ribc2 and are necessary for the ciliary beating of multi-cilia. Genes Genomics. 2023;45(2):157-167. doi: 10.1007/s13258-022-01350-w

 

  1. Li S, Han F, Qi N, et al. Determination of a six-gene prognostic model for cervical cancer based on WGCNA combined with LASSO and Cox-PH analysis. World J Surg Oncol. 2021;19(1):277. doi: 10.1186/s12957-021-02384-2

 

  1. Xu M, Jiang B, Man Z, Zhu H. TRIM37 promotes gallbladder cancer proliferation by activating the Wnt/beta-catenin pathway via ubiquitination of Axin1. Transl Oncol. 2023;35:101732. doi: 10.1016/j.tranon.2023.101732

 

  1. Roy M, Fowler AM, Ulaner GA, Mahajan A. Molecular classification of breast cancer. PET Clin. 2023;18(4):441-458. doi: 10.1016/j.cpet.2023.04.002

 

  1. Ibadurrahman W, Hanif N, Hermawan A. Functional network analysis of p85 and PI3K as potential gene targets and mechanism of oleanolic acid in overcoming breast cancer resistance to tamoxifen. J Genet Eng Biotechnol. 2022;20(1):66. doi: 10.1186/s43141-022-00341-4

 

  1. Turner NC, Im S, Saura C, et al. Inavolisib-based therapy in PIK3CA-mutated advanced breast cancer. N Engl J Med. 2024;391(17):1584-1596. doi: 10.1056/NEJMoa2404625

 

  1. Xu X, Zhang M, Xu F, Jiang S. Wnt signaling in breast cancer: Biological mechanisms, challenges and opportunities. Mol Cancer. 2020;19(1):165. doi: 10.1186/s12943-020-01276-5

 

  1. Luond F, Sugiyama N, Bill R, et al. Distinct contributions of partial and full EMT to breast cancer malignancy. Dev Cell. 2021;56(23):3203-3221.e11. doi: 10.1016/j.devcel.2021.11.006

 

  1. Krishnamurthy N, Kurzrock R. Targeting the Wnt/ beta-catenin pathway in cancer: Update on effectors and inhibitors. Cancer Treat Rev. 2018;62:50-60. doi: 10.1016/j.ctrv.2017.11.002

 

  1. Pan Y, Yu Y, Wang X, Zhang T. Tumor-associated macrophages in tumor immunity. Front Immunol. 2020;11:583084. doi: 10.3389/fimmu.2020.583084

 

  1. Ren Y, Chen Z, Sun J, et al. The correlation between infiltration of FoxP3(+) Tregs, CD66b(+) TANs and CD163(+) TAMs in colorectal cancer. Cent Eur J Immunol. 2022;47(1):1-7. doi: 10.5114/ceji.2022.114004

 

  1. Hartkopf AD, Taran F, Wallwiener M, et al. PD-1 and PD-L1 immune checkpoint blockade to treat breast cancer. Breast Care (Basel). 2016;11(6):385-390. doi: 10.1159/000453569

 

  1. Dvir K, Giordano S, Leone JP. Immunotherapy in breast cancer. Int J Mol Sci. 2024;25(14):7517. doi: 10.3390/ijms25147517

 

  1. Luobin L, Wanxin H, Yingxin G, et al. Nanomedicine-induced programmed cell death in cancer therapy: Mechanisms and perspectives. Cell Death Discov. 2024; 10(1):386. doi: 10.1038/s41420-024-02121-0
Share
Back to top
Tumor Discovery, Electronic ISSN: 2810-9775 Print ISSN: 3060-8597, Published by AccScience Publishing