Commentary - Journal of Molecular Oncology Research (2025) Volume 9, Issue 2
Advancing precision oncology: Redefining cancer therapy through molecular oncology research
Eva Brazdova Jagelska*
Department of Biophysics, Academy of Sciences, Czech Republic
- *Corresponding Author:
- Eva Brazdova Jagelska
Department of Biophysics, Academy of Sciences, Czech Republic
E-mail: eva.jagelska@biophys.cz
Received: 01-May-2025, Manuscript No. AAMOR -25-166706; Editor assigned: 02-May-2025, PreQC No. AAMOR -25-166706(PQ); Reviewed: 18-May-2025, QC No. AAMOR -25-166706; Revised: 22-May-2025, Manuscript No. AAMOR -25-166706(R); Published: 29-May-2025, DOI: 10.35841/ aamor-9.2.287
Citation: : Jagelska B E. Advancing cancer diagnostics: The promise of liquid biopsy in molecular oncology research. J Mol Oncol Res. 2025;9(2):287.
Introduction
Cancer diagnosis and monitoring have traditionally relied on invasive tissue biopsies, which pose challenges including patient discomfort, sampling bias, and limited feasibility for repeated testing. The emergence of liquid biopsy a minimally invasive method analyzing cancer-derived material circulating in bodily fluids—has transformed the landscape of molecular oncology research. By enabling real-time insights into tumor dynamics, liquid biopsy offers unprecedented opportunities for early detection, treatment personalization, and disease monitoring.
Liquid biopsies primarily detect circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), exosomes, and other tumor-derived biomarkers in blood, urine, or other fluids. These biomarkers provide a comprehensive snapshot of the tumor’s genetic and epigenetic landscape, capturing heterogeneity and evolution that traditional biopsies may miss. This dynamic profiling allows oncologists to adapt therapeutic strategies promptly, improving clinical outcomes [1].
Molecular Insights through Circulating Tumor DNA. One of the most widely studied components in liquid biopsy is ctDNA, fragmented DNA released into the bloodstream by apoptotic or necrotic tumor cells. Sensitive molecular techniques such as digital PCR and next-generation sequencing (NGS) enable the detection of ctDNA mutations, copy number alterations, and methylation patterns. Monitoring ctDNA levels can reveal minimal residual disease (MRD) after surgery or therapy, providing an early warning of relapse before clinical symptoms or imaging changes occur. Furthermore, analysis of ctDNA mutation profiles assists in identifying actionable genetic alterations, guiding targeted therapies and tracking the emergence of resistance mutations [2].
Circulating Tumor Cells and Extracellular Vesicles. Circulating tumor cells, shed from the primary tumor or metastases, serve as another valuable biomarker for liquid biopsy. CTC enumeration and molecular characterization have prognostic significance in multiple cancer types, correlating with disease progression and survival. Extracellular vesicles, including exosomes, carry proteins, RNA, and DNA reflective of the tumor microenvironment. These vesicles facilitate intercellular communication and modulate immune responses, highlighting their potential as both diagnostic markers and therapeutic targets in molecular oncology research [3].
Clinical Applications and Benefits. Liquid biopsy holds transformative potential in several clinical scenarios: Early Detection: Detection of tumor-derived biomarkers in asymptomatic individuals could enable earlier intervention, significantly improving prognosis. Research is ongoing to develop reliable liquid biopsy-based screening tests for cancers such as lung, colorectal, and pancreatic [4].
Treatment Selection and Monitoring: Real-time molecular profiling via liquid biopsy helps tailor therapies to tumor-specific alterations and monitor treatment efficacy. It can detect emerging resistance mutations, allowing timely therapy adjustments without the need for invasive tissue sampling. Disease Surveillance: Post-treatment surveillance using liquid biopsy facilitates early detection of recurrence, enabling prompt therapeutic action and potentially improving long-term survival. Challenges and Future Directions. Despite its promise, liquid biopsy faces several challenges. The low abundance of tumor-derived biomarkers in circulation requires highly sensitive and specific assays to avoid false negatives and positives. Standardization of pre-analytical and analytical procedures is critical for clinical adoption.
The complexity of tumor heterogeneity also demands integrative approaches combining ctDNA, CTCs, and exosomal analyses for comprehensive tumor characterization. Emerging technologies such as single-molecule sequencing, microfluidic enrichment of rare cells, and artificial intelligence-driven data interpretation are addressing these limitations. Ethical and Regulatory Considerations. With expanding clinical use, ethical considerations such as patient consent, data privacy, and implications of incidental findings require careful management. Regulatory frameworks must evolve to ensure the quality, safety, and equitable access to liquid biopsy diagnostics [5].
Conclusion
Liquid biopsy is revolutionizing molecular oncology research by providing a minimally invasive window into tumor biology. Its ability to capture tumor heterogeneity and temporal changes in real time paves the way for precision cancer medicine, improving early detection, treatment personalization, and monitoring. Continued technological innovation, clinical validation, and ethical oversight will be essential to fully realize the potential of liquid biopsy in transforming cancer care.
References
- Agha RA, Franchi T, Sohrabi C, et al. The SCARE 2020 guideline: Updating consensus surgical case report (SCARE) guidelines. Int J Surg. 2020;84:226-30.
- Marone J, Patel S, Page M, et al. Signet cell carcinoma of the colon in a 17 year old child. J Surg Case Rep. 2012; 12(9):3.
- Jasperson KW, Tuohy TM, Neklason DW, et al. Hereditary and familial colon cancer. Gastroenterol. 2010; 138(6):2044-58.
- De Rosa M, Pace U, Rega D, et al. Genetics diagnosis and management of colorectal cancer. Oncol Rep. 2015; 34(3):1087-096.
- Isidori AM, Pozza C, Esposito K, et al. Development and validation of a 6-item version of the female sexual function index (FSFI) as a diagnostic tool for female sexual dysfunction. J Sex Med. 2010; 7:1139-146.
- Pfister DG, Spencer S, Adelstein D, et al. Head and neck cancers version 2. 2020 NCCN clinical practice guidelines in oncology.J Natl Compr Cancer Netw. 2020; 18:873-98.
- Seiwert TY, Foster CC, Blair EA, et al. OPTIMA: A phase II dose and volume de-escalation trial for human papillomavirus-positive oropharyngeal cancer. Ann Oncol. 2019; 30:297-02.
- Chen AM, Felix C, Wang P, et al. Reduced-dose radiotherapy for human papillomavirus-associated squamous-cell carcinoma of the oropharynx: A single-arm phase 2 study. Lancet Oncol. 2017; 18:803-11.
- Lassen P, Lacas B, Pignon J, et al. Prognostic impact of HPV-associated p16-expression and smoking status on outcomes following radiotherapy for oropharyngeal cancer: the MARCH-HPV project. Radiother Oncol. 126 2018; 126:107-15.
- Tofts PS, Brix G, Buckley DL, et al. Estimating kinetic parameters from dynamic contrast-enhanced T1- weighted MRI of a diffusable tracer: Standardized quantities and symbols. J Magn Reson Imaging. 1999; 10: 223-32.
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 Schoalr, Cross Ref
Indexed at, Google Scholar, Cross Ref
Indexed at, Google Scholar, Cross Ref
Indexed at, Google Scholar, Cross Ref