Commentary - Journal of Molecular Oncology Research (2025) Volume 9, Issue 3
Unraveling the molecular pathways: Signal transduction in tumorigenesis and its implications in molecular oncology research
Sunghee Baek *
Department of Biochemistry and Molecular Biology, Seoul National University, South Korea
- *Corresponding Author:
- Sunghee Baek
Department of Biochemistry and Molecular Biology, Seoul National University, South Korea
E-mail: sungheebaek@snu.ac.kr
Received: 01-Aug-2025, Manuscript No. AAMOR -25-166750; Editor assigned: 02-Aug-2025, PreQC No. AAMOR -25-166750(PQ); Reviewed: 18-Aug-2025, QC No. AAMOR -25-166750; Revised: 22-Aug-2025, Manuscript No. AAMOR -25-166750(R); Published: 29-Aug-2025, DOI: 10.35841/ aamor-9.3.297
Citation: : Baek S. Unraveling the molecular pathways: Signal transduction in tumorigenesis and its implications in molecular oncology research. J Mol Oncol Res. 2025;9(3):297.
Introduction
Cancer arises not merely from genetic mutations, but from a cascade of disrupted cellular communication known as signal transduction. These intricate biochemical pathways govern essential functions such as cell proliferation, differentiation, apoptosis, and response to external stimuli. When these pathways are hijacked, either through mutations or external environmental stressors, they contribute to tumorigenesis—the transformation of normal cells into malignant ones. Within the field of molecular oncology research, understanding signal transduction mechanisms has emerged as a cornerstone for developing targeted therapies. Modern oncological strategies now focus not only on identifying mutations but also on mapping the signaling networks that sustain cancer progression. This article delves into the complexity of signal transduction in tumorigenesis, the major pathways involved, and their potential as therapeutic targets [1, 2].
The next frontier in signal transduction research lies in systems biology and computational modeling. By integrating genomics, transcriptomics, and proteomics data, researchers are now constructing dynamic signaling maps to predict how tumors respond to treatment in real-time. Machine learning algorithms are also being used to identify novel druggable targets and predict therapy resistance. Furthermore, signal transduction-based diagnostics are on the rise. Liquid biopsies measuring circulating tumor DNA (ctDNA) or phosphorylated proteins offer non-invasive ways to monitor pathway activity and treatment efficacy. Some of the most commonly implicated pathways in tumorigenesis include: MAPK/ERK Pathway: Often activated by receptor tyrosine kinases (RTKs), this pathway promotes cell division. Mutations in RAS or RAF genes can lead to persistent activation, contributing to melanoma, colon, and lung cancers. PI3K/AKT/mTOR Pathway: Regulates metabolism, growth, and survival. Its dysregulation is associated with resistance to apoptosis and is frequently seen in breast, ovarian, and prostate cancers [3, 4].
Additionally, feedback loops and adaptive signaling often render single-target therapies ineffective in the long run. Hence, a deeper molecular understanding is needed to design multi-targeted therapies or identify predictive biomarkers that can guide precision treatment. The tumor microenvironment (TME), including immune cells, fibroblasts, and extracellular matrix components, plays a crucial role in modulating signal transduction. Paracrine signals from the TME can activate pro-survival and metastatic pathways in cancer cells. For example, cancer-associated fibroblasts may secrete growth factors that activate the WNT or Notch pathways in nearby tumor cells. JAK/STAT Pathway: Involved in cytokine signaling and immune response. Persistent STAT3 activation supports tumor immune evasion and chronic inflammation. WNT/β-Catenin Pathway: Critical in cell fate determination and self-renewal. Its abnormal activation is linked to colorectal and liver cancers. Understanding how these signaling circuits interact allows researchers to identify nodal points for intervention—where one disruption could suppress an entire cancer-driving network [5, 6].
Targeting TME-driven signaling is gaining attention as a complementary strategy. Therapies that disrupt stromal support or immune checkpoint inhibitors that modulate JAK/STAT signaling have shown remarkable success, especially in immunogenic tumors. Recent advances in molecular oncology research have led to the development of inhibitors targeting specific components of these pathways. Small molecule inhibitors, monoclonal antibodies, and siRNA-based approaches have shown significant potential in clinical applications. EGFR inhibitors (like gefitinib) have been approved for non-small cell lung cancer. mTOR inhibitors (such as everolimus) are used in renal and breast cancers [7, 8].
BRAF inhibitors (like vemurafenib) are effective in treating BRAF-mutant melanoma. Moreover, combination therapies are becoming the norm. By targeting multiple pathways simultaneously or sequentially, oncologists can prevent resistance and achieve more durable responses. Signal transduction refers to the transmission of molecular signals from a cell's exterior to its interior, ultimately influencing gene expression and cellular behavior. In cancer, these signals are often aberrant, leading to unchecked growth and survival. Cross-Talk Between Pathways and Resistance Mechanisms. A significant challenge in targeting signal transduction is pathway cross-talk—the ability of one pathway to compensate for another when inhibited. For example, inhibition of the MAPK pathway may lead to upregulation of the PI3K pathway, allowing cancer cells to escape therapy-induced apoptosis [9, 10].
Conclusion
Signal transduction serves as the command center of cellular behavior, and its disruption is central to cancer development. With the rise of molecular oncology research, the therapeutic landscape is rapidly evolving toward more precise and dynamic interventions. By targeting the very signals that drive tumorigenesis, we move closer to transforming cancer into a manageable condition—one where therapies are guided by molecular logic rather than trial and error. Continued research, cross-disciplinary collaboration, and ethical deployment of these technologies will ensure that signal transduction remains not only a subject of academic inquiry but a clinical reality that benefits patients worldwide.
References
- Yahya EB, Alqadhi AM. Recent trends in cancer therapy: A review on the current state of gene delivery. Life Sci. 2021;269:119087.
- Sheikh-Hosseini M, Larijani B, Gholipoor Kakroodi Z, et al. Gene therapy as an emerging therapeutic approach to breast cancer: new developments and challenges. Human Gene Ther. 2021;32(21-22):1330-45.
- Gustafson MP, Ligon JA, Bersenev A, et al. Emerging frontiers in immuno-and gene therapy for cancer. Cytotherapy. 2023;25(1):20-32.
- Kobelt D, Pahle J, Walther W. A brief introduction to current cancer gene therapy.Gene Ther Canc Method Protoc. 2022.
- Patel MP, Shah MS, Athalye MN, et al. Gene therapy for cancer treatment: recent trends and clinical evidence. Gene Deliv Sys. 2022:163-80.
- Bezeljak U. Cancer gene therapy goes viral: Viral vector platforms come of age. Radiol Oncolo. 2022;56(1):1-3.
- Younis MA, Khalil IA, Harashima H. Gene therapy for hepatocellular carcinoma: Highlighting the journey from theory to clinical applications. Advan Therap. 2020;3(11):2000087.
- Ou X, Ma Q, Yin W, et al. CRISPR/Cas9 gene-editing in cancer immunotherapy: Promoting the present revolution in cancer therapy and exploring more. Fronti Cell Developm Biolo. 2021;9:674467.
- Shchaslyvyi AY, Antonenko SV, Tesliuk MG, et al. Current state of human gene therapy: Approved products and vectors. Pharmace. 2023;16(10):1416.
- Zittersteijn HA, Gonçalves MA, Hoeben RC. A primer to gene therapy: Progress, prospects, and problems. J Inher Metabo Dise. 2021;44(1):54-71.
Indexed at, Google Scholar, Cross Ref
Indexed at, Google Scholar, Cross Ref
Indexed at, Google Scholar, Cross Ref
Indexed at, Google Scholar, Cross Ref
Indexed at, Google Scholar, Cross Ref
Indexed at, Google Scholar, Cross Ref
Indexed at, Google Scholar, Cross Ref
Indexed at, Google Scholar, Cross Ref
Indexed at, Google Scholar, Cross Ref