EDITORIAL article
Front. Genet.
Sec. Cancer Genetics and Oncogenomics
Volume 16 - 2025 | doi: 10.3389/fgene.2025.1611885
This article is part of the Research TopicScreening and discovering novel biological biomarkers by omic-data to revolutionize tumor managementView all 9 articles
Editorial: Screening and discovering novel biological biomarkers by omic-data to revolutionize tumor management
Provisionally accepted- 1Henan University, Kaifeng, China
- 2Children's Hospital of Pittsburgh, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
- 3School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
- 4State University of Northern Rio de Janeiro, Campos dos Goytacazes, Rio de Janeiro, Brazil
- 5School of Basic Medical Sciences, Henan University, Zhengzhou, China, Zhengzhou, China
Select one of your emails
You have multiple emails registered with Frontiers:
Notify me on publication
Please enter your email address:
If you already have an account, please login
You don't have a Frontiers account ? You can register here
revolutionized cancer research, enabling systematic biomarker discovery through multi-omics analyses (Goodwin et al., 2016). For example, single-cell RNA sequencing (scRNA-Seq) and spatial transcriptomics have enabled the resolution of tumor heterogeneity at subcellular levels (Longo et al., 2021;Gulati et al., 2025).These innovations underscore the shift from bulk tissue analysis to spatially and temporally resolved molecular profiling, which is critical for deciphering dynamic tumor-immune interactions (Hsieh et al., 2022). The integration of multi-omics data has accelerated the identification of biomarkers with clinical utility. Below, we contextualize the studies in this research topic within recent breakthroughs in oncology.Multi-omics integration allows a comprehensive view of the mechanisms underlying any disease. Ahmed N et al. characterized genetic and epigenetic changes in AML progression using Omni-C, ATAC-seq, and RNA-seq data. Differential interaction analysis showed significant 3D chromatin landscape reorganization between relapse and diagnosis samples (Ahmed et al., 2024). Uveal melanoma (UVM) is the most common primary intraocular malignancy. However, treatment outcomes are unsatisfactory, and the long-term prognosis remains dismal. To address this challenge, Zhang M et al. employed bioinformatics and identified that two TRP channel-related long noncoding RNAs (TCRLs), AC092535.4 and LINC01637, could serve as novel prognostic biomarkers for UVM and may present potential therapeutic targets (Zhang et al., 2024). Using multi-omics data, Zhao Y et al. found that FANCI is significantly upregulated in multiple tumor types. High FANCI expression correlates with poor prognosis in specific cancers and is associated with higher immune cell infiltration and tumor mutation burden. FANCI emerges as a promising biomarker for cancer prognosis and diagnosis, with potential as a novel therapeutic target (Zhao et al., 2025).Kinesin family proteins and metabolic reprogramming are emerging as central players in tumor progression (Chen et al., 2024b). The expression of KIF18B was correlated with immune infiltration in the tumor microenvironment. KIF18B is a key factor affecting the prognosis of glioblastoma (GBM) patients, and its targeting may provide a new therapeutic method for GBM patients (Su et al., 2025). Yang B et al. demonstrated that KIF18B correlated with hepatocellular carcinoma progression potentially via activation of the Wnt/β-catenin-signaling pathway, suggesting its broader role in epigenetic regulation (Yang et al., 2020). Targeting metabolic enzymes like GFPT1 has gained traction. In vivo, silencing of GFPT1 attenuated the immune escape of breast cancer cells by reducing PD-L1 levels (Tang et al., 2024). These findings align with Liang et al.'s observation that high GFPT1 level was associated with increased cytoplasmic translation, activation of oncogenic pathways, and infiltration of M2 macrophages, indicating that GFPT1 may be a novel prognostic biomarker and an indicator of chemotherapy response in invasive breast carcinoma (Liang et al., 2024).Immune-related signatures are reshaping therapeutic strategies. Ni D, et al. constructed an 8-gene prognostic model (AK2, CXCL11, TYK2, ANGPT4, IL20RA, MET, ENPP6, and CA12) in pancreatic ductal adenocarcinoma (PDAC), enhancing prognostic accuracy and potentially therapeutic decision-making in PDAC, offering valuable insights for evaluation in clinical practice (Ni et al., 2024). Wan C, et al. found that high MARCKS, MACC1, and GRB10 expression in their tumors correlated with poorer survival rates in endometrial cancer patients. High expression of NINJ2 correlated with higher survival rates and higher sensitivity to radiation therapy (Wan et al., 2025).Immunotherapy based on immune checkpoint inhibitors (ICIs) has become a prominent focus in the development of novel anti-tumor drugs and has been tested in several human clinical trials. Thus, an immune-related gene prognostic index (IRGPI) was developed by Chen D et al., providing a systematic analysis of distinct and molecular characteristics in papillary renal cell carcinoma (PRCC) (Chen et al., 2024a). This research topic exemplifies the power of multi-omics approaches in discovering novel biological biomarkers and unraveling the molecular complexity of cancer. By identifying biomarkers that reflect tumor biology, immune interactions, and therapeutic vulnerabilities, these studies pave the way for precision oncology. As we advance, integrating multi-omics data with AI and innovative therapeutics will be pivotal in transforming cancer care, ultimately improving survival and quality of life for patients worldwide.Keywords: biomarkers, multi-omics data, therapeutic targets, immunotherapy, cancer
Keywords: biomarkers, Multi-omics data, therapeutic targets, Immunotherapy, Cancer
Received: 15 Apr 2025; Accepted: 13 Jun 2025.
Copyright: © 2025 Wang, Jiang, Wu, Medina-Acosta, M.Sc., Ph.D. and Yan. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) or licensor are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
* Correspondence: Yinan Jiang, Children's Hospital of Pittsburgh, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15224, Pennsylvania, United States
Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.