Submitted:
10 June 2025
Posted:
12 June 2025
You are already at the latest version
Abstract
Keywords:
Introduction
Clinical Applications of ctDNA in Genitourinary Malignancies
Prostate Cancer
Urothelial (Bladder) Cancer
Renal Cell Carcinoma (RCC)
Testicular Germ Cell Tumors (TGCT)
Overarching Clinical Applications and Comparison to Standard of Care
- Risk Stratification and Prognostication: Post-treatment ctDNA positivity is a strong independent predictor of recurrence, often with months of lead time over imaging [40,50,51,52]. This allows for better identification of high-risk patients who may benefit from intensified or novel adjuvant strategies [53].
- Real-Time Monitoring of Therapeutic Response: The short half-life of ctDNA allows for dynamic tracking of tumor burden and molecular changes during therapy [54,55]. Declines in ctDNA levels frequently correlate with treatment efficacy and improved outcomes, sometimes preceding radiological response [52,56,57,58,59].
- Identification of Resistance Mechanisms: Serial ctDNA analysis can detect the emergence of resistance mutations (e.g., to targeted therapies or chemotherapy) earlier than clinical progression, allowing for timely treatment adjustments [37,55,61,62]. It also helps characterize tumor heterogeneity [37].
Advanced and Integrative Applications
Unveiling Complex Genomic Landscapes
Capturing Tumor Heterogeneity and Tracking Clonal Evolution
Integrating ctDNA with Other Liquid Biopsy Analytes for a Holistic View
Complementing Imaging with Molecular Insights
Challenges and Ethical Considerations
Scientific and Technical Hurdles
Clinical Validation and Regulatory Oversight
Economic and Health System Implementation Barriers
Ethical, Legal, and Social Implications (ELSI)
Future Perspectives
Anticipated Technological Breakthroughs and Assay Optimization
The Role of Artificial Intelligence and Machine Learning
Towards Optimized and Potentially Malignancy-Specific Assays
Innovative Clinical Trial Designs and Validation Pathways
Transformative Impact Across the GU Cancer Care Continuum
Conclusions
References
- Palve S, Bhardwaj Y, Dusane J, Wankhade P. Blood-borne biomarkers: CT-DNA ushers in a new era of cancer detection. Int J Pharm Sci Drug Res.
- Russano M, Napolitano A, Ribelli G, Iuliani M, Simonetti S, Citarella F, et al. Liquid biopsy and tumor heterogeneity in metastatic solid tumors: the potentiality of blood samples. J Exp Clin Cancer Res. 2020, 39, 95. [Google Scholar] [CrossRef] [PubMed]
- Turabi K, Klute K, Radhakrishnan P. Decoding the dynamics of circulating tumor DNA in liquid biopsies. Cancers (Basel). 2024, 16, 2432. [Google Scholar] [CrossRef] [PubMed]
- Zhao Y, Zhang Z, Qiu JH, Li RY, Sun ZG. Catching cancer signals in the blood: Innovative pathways for early esophageal cancer diagnosis. World J Gastroenterol. 2025, 31, 101838. [Google Scholar]
- Li M, Xie S, Lu C, Zhu L, Zhu L. Application of data science in circulating tumor DNA detection: A promising avenue towards liquid biopsy. Front Oncol. 2021, 11, 692322. [Google Scholar] [CrossRef]
- Moon GY, Dalkiran B, Park HS, Shin D, Son C, Choi JH, et al. Dual biomarker strategies for liquid biopsy: Integrating circulating tumor cells and circulating tumor DNA for enhanced tumor monitoring. Biosensors (Basel). 2025, 15, 74. [Google Scholar]
- Yi X, Ma J, Guan Y, Chen R, Yang L, Xia X. The feasibility of using mutation detection in ctDNA to assess tumor dynamics. Int J Cancer. 2017, 140, 2642–2647. [Google Scholar] [CrossRef]
- Lin C, Liu X, Zheng B, Ke R, Tzeng CM. Liquid biopsy, ctDNA diagnosis through NGS. Life (Basel). 2021, 11, 890. [Google Scholar]
- Ji H, Long X, Gu J, Jin J, Mao X, Wang Z, et al. Longitudinal monitoring of plasma circulating tumour DNA enables the prediction of early relapse in patients with non-Hodgkin lymphoma: A case series. Diagnostics (Basel). 2021, 11, 2055. [Google Scholar] [CrossRef] [PubMed]
- Mair R, Mouliere F, Smith CG, Chandrananda D, Gale D, Marass F, et al. Measurement of plasma cell-free mitochondrial tumor DNA improves detection of glioblastoma in patient-derived orthotopic xenograft models. Cancer Res. 2019, 79, 220–230. [Google Scholar] [CrossRef]
- Underhill, HR. Leveraging the fragment length of circulating tumour DNA to improve molecular profiling of solid tumour malignancies with next-generation sequencing: A pathway to advanced non-invasive diagnostics in precision oncology? Mol Diagn Ther. 2021, 25, 389–408. [Google Scholar] [CrossRef]
- Wang Y, Wang Z, Gang X, Wang G. Liquid biopsy in prostate cancer: current status and future challenges of clinical application. Aging Male. 2021, 24, 58–71. [Google Scholar] [CrossRef] [PubMed]
- Bettegowda C, Sausen M, Leary R, Kinde I, Agrawal N, Bartlett B, et al. Abstract 5606, Detection of circulating tumor DNA in early and late stage human malignancies. Cancer Res. 2014, 74 (19_Supplement), 5606–5606. [CrossRef]
- Diaz LA Jr, Bardelli A. Liquid biopsies: genotyping circulating tumor DNA. J Clin Oncol. 2014, 32, 579–586. [Google Scholar] [CrossRef]
- Gerke MB, Jansen CS, Bilen MA. Circulating tumor DNA in genitourinary cancers: Detection, prognostics, and therapeutic implications. Cancers (Basel). 2024, 16, 2280. [Google Scholar] [CrossRef]
- Wang H, Zhang Y, Zhang H, Cao H, Mao J, Chen X, et al. Liquid biopsy for human cancer: cancer screening, monitoring, and treatment. MedComm. 2024, 5, e564. [Google Scholar] [CrossRef]
- Dawson SJ, Tsui DWY, Murtaza M, Biggs H, Rueda OM, Chin SF, et al. Analysis of circulating tumor DNA to monitor metastatic breast cancer. N Engl J Med. 2013, 368, 1199–1209. [Google Scholar] [CrossRef]
- Stejskal P, Goodarzi H, Srovnal J, Hajdúch M, van ’t Veer LJ, Magbanua MJM. Circulating tumor nucleic acids: biology, release mechanisms, and clinical relevance. Mol Cancer. 2023, 22, 15. [Google Scholar] [CrossRef] [PubMed]
- Warton K, Mahon KL, Samimi G. Methylated circulating tumor DNA in blood: power in cancer prognosis and response. Endocr Relat Cancer. 2016, 23, R157–71. [Google Scholar] [CrossRef]
- Cheng F, Su L, Qian C. Circulating tumor DNA: a promising biomarker in the liquid biopsy of cancer. Oncotarget. 2016, 7, 48832–48841. [Google Scholar] [CrossRef]
- Meriranta L, Alkodsi A, Pasanen A, Lepistö M, Mapar P, Blaker YN, et al. Molecular features encoded in the ctDNA reveal heterogeneity and predict outcome in high-risk aggressive B-cell lymphoma. Blood. 2022, 139, 1863–1877. [Google Scholar] [CrossRef]
- Danesi R, Lo YMD, Oellerich M, Beck J, Galbiati S, Re MD, et al. What do we need to obtain high quality circulating tumor DNA (ctDNA) for routine diagnostic test in oncology? - Considerations on pre-analytical aspects by the IFCC workgroup cfDNA. Clin Chim Acta. 2021, 520, 168–171. [Google Scholar] [CrossRef] [PubMed]
- Sherwood JL, Corcoran C, Brown H, Sharpe AD, Musilova M, Kohlmann A. Optimised pre-analytical methods improve KRAS mutation detection in circulating tumour DNA (ctDNA) from patients with non-small cell lung cancer (NSCLC). PLoS One. 2016, 11, e0150197. [Google Scholar]
- Hollanda CN, Gualberto ACM, Motoyama AB, Pittella-Silva F. Advancing leukemia management through liquid biopsy: Insights into biomarkers and clinical utility. Cancers (Basel) [Internet]. 2025, 17(9). Available from: https://www.mdpi.com/2072-6694/17/9/1438.
- Meddeb R, Dache ZAA, Thezenas S, Otandault A, Tanos R, Pastor B, et al. Quantifying circulating cell-free DNA in humans. Sci Rep. 2019, 9, 5220. [Google Scholar] [CrossRef] [PubMed]
- Asante DB, Tierno D, Grassi G, Scaggiante B. Circulating tumour DNA for ovarian cancer diagnosis and treatment monitoring: What perspectives for clinical use? Int J Mol Sci [Internet]. 2025 Feb 22 [cited 2025 May 22];26(5). Available from: https://www.mdpi.com/1422-0067/26/5/1889.
- van Ginkel JH, Huibers MMH, van Es RJJ, de Bree R, Willems SM. Droplet digital PCR for detection and quantification of circulating tumor DNA in plasma of head and neck cancer patients. BMC Cancer. 2017, 17, 428. [Google Scholar]
- Aiyer S, Kim TH, Collier K, Pollock R, Verschraegen C, Stover DG, et al. Unlocking the potential of ctDNA in sarcomas: A review of recent advances. Cancers (Basel). 2025, 17, 1040. [Google Scholar] [CrossRef]
- Yin H, Zhang M, Zhang Y, Zhang X, Zhang X, Zhang B. Liquid biopsies in cancer. Liquid biopsies in cancer. Mol Biomed. 2025.
- Bohers E, Viailly PJ, Jardin F. CfDNA sequencing: Technological approaches and bioinformatic issues. Pharmaceuticals (Basel). 2021, 14, 596. [Google Scholar] [CrossRef]
- Deveson IW, Gong B, Lai K, LoCoco JS, Richmond TA, Schageman J, et al. Evaluating the analytical validity of circulating tumor DNA sequencing assays for precision oncology. Nat Biotechnol. 2021, 39, 1115–1128. [Google Scholar] [CrossRef]
- Gao Y, Zhao H, An K, Liu Z, Hai L, Li R, et al. Whole-genome bisulfite sequencing analysis of circulating tumour DNA for the detection and molecular classification of cancer. Clin Transl Med. 2022, 12, e1014. [Google Scholar] [CrossRef]
- Merker JD, Oxnard GR, Compton C, Diehn M, Hurley P, Lazar AJ, et al. Circulating tumor DNA analysis in patients with cancer: American society of clinical oncology and College of American Pathologists joint review. J Clin Oncol. 2018, 36, 1631–1641. [Google Scholar] [CrossRef] [PubMed]
- Nakamura Y, Shitara K. Development of circulating tumour DNA analysis for gastrointestinal cancers. ESMO Open. 2020, 5 (Suppl 1), e000600. [CrossRef]
- Eibl RH, Schneemann M. Cell-free DNA as a biomarker in cancer. Extracell Vesicles Circ Nucl Acids. 2022, 3, 195–215. [Google Scholar]
- Lee JS, Cho EH, Kim B, Hong J, Kim YG, Kim Y, et al. Clinical practice guideline for blood-based circulating tumor DNA assays. Ann Lab Med. 2024, 44, 195–209. [Google Scholar] [CrossRef]
- Sorbini M, Carradori T, Togliatto GM, Vaisitti T, Deaglio S. Technical advances in circulating cell-free DNA detection and analysis for personalized medicine in patients’ care. Biomolecules [Internet]. 2024, 14(4). Available from: https://www.mdpi.com/2218-273X/14/4/498.
- Wang Z, Wu Q. Advancements in non-invasive diagnosis of gastric cancer. World J Gastroenterol. 2025.
- Conteduca V, Wetterskog D, Sharabiani M, Grande E, Fernández-Pérez M, Jayaram A, et al. Androgen receptor gene status in plasma DNA associates with worse outcome on enzalutamide or abiraterone for castration-resistant prostate cancer: a multi-institution correlative biomarker study. Ann Oncol. 2017.
- Hasenleithner SO, Speicher M. A clinician’s handbook for using ctDNA throughout the patient journey. Mol Cancer [Internet]. 2022, 21. Available from: https://molecular-cancer.biomedcentral.com/articles/10.1186/s12943-022-01551-7.
- Annala M, Vandekerkhove G, Khalaf D, Taavitsainen S, Beja K, Warner E, et al. Circulating tumor DNA genomics correlate with resistance to abiraterone and enzalutamide in prostate cancer. Cancer Discov. 2018, 8, 444–457. [CrossRef]
- Jänne P, Oxnard G, Thress K, Alden R, Lawrance R. Association between plasma genotyping and outcomes of treatment with osimertinib (AZD9291) in advanced non-small-cell lung cancer. J Clin Oncol. 2016, 34, 3375–3382.
- Belic J, Graf R, Bauernhofer T, Cherkas Y, Ulz P, Waldispuehl-Geigl J, et al. Genomic alterations in plasma DNA from patients with metastasized prostate cancer receiving abiraterone or enzalutamide. Int J Cancer. 2018, 143, 1236–1248. [Google Scholar] [CrossRef]
- Christensen E, Birkenkamp-Demtröder K, Sethi H, Shchegrova S, Salari R, Nordentoft I, et al. Early detection of metastatic relapse and monitoring of therapeutic efficacy by ultra-deep sequencing of plasma cell-free DNA in patients with urothelial bladder carcinoma. J Clin Oncol. 2019, 37, 1547–1557. [Google Scholar] [CrossRef]
- Li S, Xin K, Pan S, Wang Y, Zheng J, Li Z, et al. Blood-based liquid biopsy: insights into early detection, prediction, and treatment monitoring of bladder cancer. Cell Mol Biol Lett. 2023, 28, 28. [Google Scholar]
- Patel KR, Rais-Bahrami S, Basu A. High sensitivity ctDNA assays in genitourinary malignancies: current evidence and future directions. Oncologist. 2024, 29, 731–737. [Google Scholar] [CrossRef] [PubMed]
- Vandekerkhove G, Todenhöfer T, Annala M, Struss W, Wong A, Beja K, et al. Circulating tumor DNA reveals clinically actionable somatic genome of metastatic bladder cancer. Clin Cancer Res. 2017, 23, 6487–6497. [Google Scholar] [CrossRef] [PubMed]
- Sabit H, Attia MG, Mohamed N, Taha PS, Ahmed N, Osama S, et al. Beyond traditional biopsies: the emerging role of ctDNA and MRD on breast cancer diagnosis and treatment. Discov Oncol. 2025, 16, 271. [Google Scholar] [CrossRef]
- kimi AA. Circulating tumor DNA (ctDNA) in kidney cancer: A narrative review. Société Int d’urologie J [Internet]. 2023, Available from: https://www.mdpi.com/2563-6499/4/4/287.
- Duffy MJ, Crown J. Circulating tumor DNA as a biomarker for monitoring patients with solid cancers: Comparison with standard protein biomarkers. Clin Chem. 2022, 68, 1381–1390. [Google Scholar] [CrossRef]
- Moding EJ, Nabet BY, Alizadeh AA, Diehn M. Detecting liquid remnants of solid tumors: Circulating tumor DNA minimal residual disease. Cancer Discov. 2021, 11, 2968–2986. [Google Scholar] [CrossRef]
- Zollinger DR, Rivers E, Fine A, Huang Y, Son J, Kalyan A, et al. Analytical validation of a novel comprehensive genomic profiling informed circulating tumor DNA monitoring assay for solid tumors. PLoS One. 2024, 19, e0302129. [Google Scholar]
- Tie J, Cohen JD, Wang Y, Christie M, Simons K, Lee M, et al. Circulating tumor DNA analyses as markers of recurrence risk and benefit of adjuvant therapy for stage III colon cancer. JAMA Oncol. 2019, 5, 1710–1717. [Google Scholar] [CrossRef]
- Cheng ML, Pectasides E, Hanna GJ, Parsons HA, Choudhury AD, Oxnard GR. Circulating tumor DNA in advanced solid tumors: Clinical relevance and future directions. CA Cancer J Clin. 2021, 71, 176–190. [Google Scholar] [CrossRef]
- Fernandez SV, Tan YF, Rao S, Fittipaldi P, Sheriff F, Borghaei H, et al. Validation of a molecular diagnostic test for circulating tumor DNA by next-gen sequencing. Int J Mol Sci [Internet]. 2023, 24. Available from: https://www.mdpi.com/1422-0067/24/21/15779.
- Magbanua MJM, Brown-Swigart L, Wu HT, Hirst GL, Yau C, Wolf DM, et al. Circulating tumor DNA in neoadjuvant treated breast cancer reflects response and survival [Internet]. bioRxiv. medRxiv; 2020. Available from: http://medrxiv.org/lookup/doi/10.1101/2020.02.03.20019760.
- Kansara M, Bhardwaj N, Thavaneswaran S, Xu C, Lee JK, Chang LB, et al. Early circulating tumor DNA dynamics as a pan-tumor biomarker for long-term clinical outcome in patients treated with durvalumab and tremelimumab. Mol Oncol. 2022, 17, 298–311. [Google Scholar]
- Vega DM, Nishimura KK, Zariffa N, Thompson JC, Hoering A, Cilento V, et al. Changes in circulating tumor DNA reflect clinical benefit across multiple studies of patients with non–small-cell lung cancer treated with immune checkpoint inhibitors. JCO Precis Oncol [Internet]. 2022, 6. Available from: https://ascopubs.org/doi/10.1200/PO.21.00372.
- Zhang Q, Luo J, Wu S, Si H, Gao C, Xu W, et al. Prognostic and predictive impact of circulating tumor DNA in patients with advanced cancers treated with immune checkpoint blockade. Cancer Discov. 2020, 10, 1842–1853. [Google Scholar] [CrossRef]
- Chaudhuri AA, Chabon JJ, Lovejoy AF, Newman AM, Stehr H, Azad TD, et al. Early detection of molecular residual disease in localized lung cancer by circulating tumor DNA profiling. Cancer Discov. 2017, 7, 1394–1403. [Google Scholar] [CrossRef]
- Ma F, Zhu W, Guan Y, Yang L, Xia X, Chen S, et al. ctDNA dynamics: a novel indicator to track resistance in metastatic breast cancer treated with anti-HER2 therapy. Oncotarget. 2016, 7, 66020–66031. [Google Scholar] [CrossRef]
- Tie J, Kinde I, Wang Y, Wong H, Roebert J, Christie M, et al. Circulating tumor DNA as an early marker of therapeutic response in patients with metastatic colorectal cancer. Circulating tumor DNA as an early marker of therapeutic response in patients with metastatic colorectal cancer. Ann Oncol. 2015.
- Siravegna G, Mussolin B, Venesio T, Marsoni S, Seoane J, Dive C, et al. How liquid biopsies can change clinical practice in oncology. Ann Oncol. 2019, 30, 1580–1590. [Google Scholar] [CrossRef]
- Sivapalan L, Murray J, Canzoniero JV, Landon BV, Jackson J, Scott SL, et al. Liquid biopsy approaches to capture tumor evolution and clinical outcomes during cancer immunotherapy. J Immunother Cancer [Internet]. 2023, 11. Available from: https://jitc.bmj.com/lookup/doi/10.1136/jitc-2022-005924.
- Li L, Sun Y. Circulating tumor DNA methylation detection as biomarker and its application in tumor liquid biopsy: advances and challenges. MedComm. 2024, 5, e766. [Google Scholar] [CrossRef] [PubMed]
- Walz AL, Maschietto M, Crompton B, Evageliou N, Dix D, Tytgat G, et al. Tumor biology, biomarkers, and liquid biopsy in pediatric renal tumors. Pediatr Blood Cancer [Internet]. 2023, 70. Available from: https://onlinelibrary.wiley.com/doi/10.1002/pbc.30130.
- Charlton J, Williams RD, Weeks M, Sebire NJ, Popov S, Vujanic G, et al. Methylome analysis identifies a Wilms tumor epigenetic biomarker detectable in blood. Genome Biol. 2014, 15, 434. [Google Scholar]
- van Zogchel LMJ, Lak N, Verhagen O, Tissoudali A, Nuru MG, Gelineau NU, et al. Novel circulating hypermethylated RASSF1A ddPCR for liquid biopsies in patients with pediatric solid tumors. JCO Precis Oncol [Internet]. 2021, 5. Available from: https://ascopubs.org/doi/10.1200/PO.21.00130.
- Papalexis P, Georgakopoulou V, Drossos P, Thymara E, Nonni A, Lazaris AC, et al. Precision medicine in breast cancer (Review). Mol Clin Oncol [Internet]. 2024, 21. Available from: http://www.spandidos-publications.com/10.3892/mco.2024.2776.
- Madanat-Harjuoja L, Renfro L, Klega K, Tornwall B, Thorner A, Nag A, et al. Circulating tumor DNA as a biomarker in patients with stage III and IV Wilms tumor: Analysis from a Children’s Oncology Group trial, AREN0533. J Clin Oncol. 2022, 40, 3047–3056.
- Mattos-Arruda LD, Mattos-Arruda LD, Mattos-Arruda LD, Weigelt B, Cortés J, Won H, et al. Capturing intra-tumor genetic heterogeneity by de novo mutation profiling of circulating cell-free tumor DNA: a proof-of-principle. Ann Oncol. 2014, 25, 1729–1735. [Google Scholar] [CrossRef] [PubMed]
- Yeh P, Hunter T, Sinha D, Ftouni S, Wallach E, Jiang DHS, et al. Circulating tumour DNA reflects treatment response and clonal evolution in chronic lymphocytic leukaemia. Nat Commun [Internet]. 2017, 8. Available from: https://www.nature.com/articles/ncomms14756.
- Chabon J, Simmons A, Lovejoy A, Esfahani MS, Newman AM, Haringsma HJ, et al. Circulating tumour DNA profiling reveals heterogeneity of EGFR inhibitor resistance mechanisms in lung cancer patients. Nat Commun. 2016, 7, 11815. [Google Scholar] [CrossRef]
- Corcoran R, Chabner B. Application of Cell-free DNA Analysis to Cancer Treatment. N Engl J Med. 2018.
- Bamodu OA, Chung CC, Pisanic TR 2nd. Harnessing liquid biopsies: Exosomes and ctDNA as minimally invasive biomarkers for precision cancer medicine. J Liq Biopsy. 2023, 2, 100126. [Google Scholar]
- Hench IB, Hench J, Tolnay M. Liquid biopsy in clinical management of breast, lung, and colorectal cancer. Front Med (Lausanne) [Internet]. 2018, 5. Available from: http://journal.frontiersin.org/article/10.3389/fmed.2018.00009/full.
- Carrasco R, Ingelmo-Torres M, Trullas R, Roldán F, Rodriguez-Carunchio L, Juez L, et al. Tumor-agnostic circulating tumor DNA testing for monitoring muscle-invasive bladder cancer. Int J Mol Sci. 2023, 24, 16578. [Google Scholar] [CrossRef]
- Zhou Y, Wang R, Zeng M, Liu S. Circulating tumor DNA: a revolutionary approach for early detection and personalized treatment of bladder cancer. Front Pharmacol [Internet]. 2025, 16. Available from: https://www.frontiersin.org/articles/10.3389/fphar.2025.1551219/full.
- Onidani K, Shoji H, Kakizaki T, Yoshimoto S, Okaya S, Miura N, et al. Monitoring of cancer patients via next-generation sequencing of patient-derived circulating tumor cells and tumor DNA. Cancer Sci. 2019, 110, 2590–2599. [Google Scholar] [CrossRef] [PubMed]
- Peneder P, Stütz A, Surdez D, Krumbholz M, Semper S, Chicard M, et al. Multimodal analysis of cell-free DNA whole-genome sequencing for pediatric cancers with low mutational burden. Nat Commun [Internet]. 2021, 12. Available from: https://www.nature.com/articles/s41467-021-23445-w.
- Xie J, Hu B, Gong Y, He S, Lin J, Huang Q, et al. A comparative study on ctDNA and tumor DNA mutations in lung cancer and benign cases with a high number of CTCs and CTECs. J Transl Med [Internet]. 2023, 21. Available from: https://translational-medicine.biomedcentral.com/articles/10.1186/s12967-023-04746-8.
- Khanyile R, Chipiti T, Hull R, Dlamini Z. Radiogenomic landscape of metastatic endocrine-positive breast cancer resistant to aromatase inhibitors. Cancers (Basel) [Internet]. 2025, 17. Available from: https://www.mdpi.com/2072-6694/17/5/808.
- Mishra M, Ahmed R, Das DK, Pramanik DD, Dash SK, Pramanik A. Recent advancements in the application of circulating tumor DNA as biomarkers for early detection of cancers. ACS Biomater Sci Eng. 2024, 10, 4740–4756. [Google Scholar] [CrossRef] [PubMed]
- García-Pardo M, Makarem M, Li JJ, Kelly D, Leighl N. Integrating circulating-free DNA (cfDNA) analysis into clinical practice: opportunities and challenges. Br J Cancer. 2022, 127, 592–602. [Google Scholar] [CrossRef]
- Marsavela G, McEvoy A, Pereira M, Reid A, Al-Ogaili Z, Warburton L, et al. Detection of clinical progression through plasma ctDNA in metastatic melanoma patients: a comparison to radiological progression. Br J Cancer. 2021, 126, 401–408. [Google Scholar]
- Godsey JH, Silvestro A, Barrett J, Bramlett K, Chudova DI, Deras I, et al. Generic protocols for the analytical validation of Next-Generation Sequencing-based ctDNA assays: A joint consensus recommendation of the BloodPAC’s Analytical Variables Working Group. Clin Chem. 2020, 66, 1156–1166. [Google Scholar] [CrossRef]
- Fiala C, Diamandis E. Utility of circulating tumor DNA in cancer diagnostics with emphasis on early detection. BMC Med [Internet]. 2018, 16. Available from: https://bmcmedicine.biomedcentral.com/articles/10.1186/s12916-018-1157-9.
- Sotoudeh Anvari M, Gharib A, Abolhasani M, Azari-yam A, Gharalari FH, Safavi M, et al. Pre-analytical practices in the molecular diagnostic tests, A concise review. Iran J Pathol. 2020, 16, 1–19. [Google Scholar]
- Haselmann V, Hedtke M, Neumaier M. Liquid profiling for cancer patient stratification in precision medicine—current status and challenges for successful implementation in standard care. Diagnostics (Basel) [Internet]. 2022, 12. Available from: https://www.mdpi.com/2075-4418/12/3/748.
- Horgan D, Čufer T, Gatto F, Lugowska I, Verbanac D, Carvalho Â, et al. Accelerating the development and validation of liquid biopsy for early cancer screening and treatment tailoring. Healthcare [Internet]. 2022, 10. Available from: https://www.mdpi.com/2227-9032/10/9/1714.
- Hum M, Lee ASG. DNA methylation in breast cancer: early detection and biomarker discovery through current and emerging approaches. J Transl Med. 2025, 23, 465. [Google Scholar] [CrossRef] [PubMed]
- Meddeb R, Pisareva E, Thierry A. Guidelines for the preanalytical conditions for analyzing circulating cell-free DNA. Clin Chem. 2019, 65, 623–633. [Google Scholar] [CrossRef]
- Stetson DB, Ahmed A, Xu X, Nuttall B, Lubinski TJ, Johnson JH, et al. Orthogonal comparison of four plasma NGS tests with tumor suggests technical factors are a major source of assay discordance. JCO Precis Oncol. 2019, 3, 1–9. [Google Scholar]
- Huang Z, Fu Y, Yang H, Zhou Y, Shi M, Li Q, et al. Liquid biopsy in T-cell lymphoma: biomarker detection techniques and clinical application. Mol Cancer [Internet]. 2024, 23. Available from: https://molecular-cancer.biomedcentral.com/articles/10.1186/s12943-024-01947-7.
- Razavi P, Li BT, Brown DN, Jung B, Hubbell E, Shen R, et al. High-intensity sequencing reveals the sources of plasma circulating cell-free DNA variants. News@nat,Com. 2019, 25, 1928–1937.
- Diaz L, Sausen M, Fisher G, Velculescu V. Insights into therapeutic resistance from whole-genome analyses of circulating tumor DNA. Oncotarget. 2013, 4, 1856–1857. [Google Scholar] [CrossRef] [PubMed]
- Leary R, Sausen M, Diaz L, Velculescu V. Cancer detection using whole-genome sequencing of cell free DNA. Oncotarget. 2013, 4, 1119–1120. [Google Scholar] [CrossRef] [PubMed]
- Sausen M, Parpart S, Diaz L. Circulating tumor DNA moves further into the spotlight. Genome Med. 2014, 6, 35–35. [Google Scholar] [CrossRef]
- Ho HY, Chung KSK, Kan CM, Wong SCC. Liquid biopsy in the clinical management of cancers. Int J Mol Sci. 2024, 25, 8594. [Google Scholar] [CrossRef] [PubMed]
- Keppens C, Dequeker E, Patton S, Normanno N, Fenizia F, Butler R, et al. International pilot external quality assessment scheme for analysis and reporting of circulating tumour DNA. BMC Cancer [Internet]. 2018, 18. Available from: https://bmccancer.biomedcentral.com/articles/10.1186/s12885-018-4694-x.
- Dong L, Wang W, Li A, Kansal R, Chen Y, Chen H, et al. Clinical next generation sequencing for precision medicine in cancer. Curr Genomics. 2015, 16, 253–263. [Google Scholar] [CrossRef]
- Liu M, Mu T, Gu J, Xu M, Chen S. The method of minimal residual disease detection with circulating tumor DNA and its clinical applications in colorectal cancer. Cancer Rep. 2025, 8, e70167. [Google Scholar] [CrossRef]
- Zhang H, Wang L, Wu H. Liquid biopsy in ovarian cancer in China and the world: current status and future perspectives. Front Oncol [Internet]. 2023, 13. Available from: https://www.frontiersin.org/articles/10.3389/fonc.2023.1276085/full.
- Desai A, Vázquez TA, Arce KM, Corassa M, Mack PC, Gray JE, et al. CtDNA for the evaluation and management of EGFR-mutant non-small cell lung cancer. Cancers (Basel) [Internet]. 2024, 16. Available from: https://www.mdpi.com/2072-6694/16/5/940.
- Hasenleithner SO, Heitzer E. Liquid profiling for patients with advanced cancer is ready for clinical integration. Liquid profiling for patients with advanced cancer is ready for clinical integration. Memo. 2024.
- Hu Y, Ulrich B, Supplee JG, Kuang Y, Lizotte PH, Feeney N, et al. False-positive plasma genotyping due to clonal hematopoiesis. Clin Cancer Res. 2018, 24, 4437–4443. [Google Scholar] [CrossRef]
- Sheriff S, Saba M, Patel R, Fisher G, Schroeder T, Arnolda G, et al. A scoping review of factors influencing the implementation of liquid biopsy for cancer care. J Exp Clin Cancer Res [Internet]. 2025, 44. Available from: https://jeccr.biomedcentral.com/articles/10.1186/s13046-025-03322-w.
- Adhit KK, Wanjari A, Menon S, K S. Liquid Biopsy: An Evolving Paradigm for Non-invasive Disease Diagnosis and Monitoring in Medicine. Cureus. 2023.
- Alqahtani S, Alqahtani T, Venkatesan K, Sivadasan D, Ahmed R, Elfadil H, et al. Unveiling Pharmacogenomics Insights into Circular RNAs: Toward Precision Medicine in Cancer Therapy. Biomolecules. 2025.
- Ashif M, Akram R. Revolutionizing cancer care: Liquid biopsy redefining surgical biopsy paradigms. Int j multidiscip res [Internet]. 2024, Available from: https://www.ijfmr.com/research-paper.php?id=13089.
- Wang Y, Shao W, Li H, Zhao P, Tian L, Zhang L, et al. Clinical application of minimal residual disease detection by ctDNA testing in non-small cell lung cancer: a narrative review. Transl Lung Cancer Res. 2025, 14, 1007–1020. [Google Scholar] [CrossRef] [PubMed]
- Trosman J, Weldon C, Kurian A, Pasquinelli M, Kircher S, Martin N, et al. Perspectives of private payers on multicancer early-detection tests: informing research, implementation, and policy. Health Affairs Scholar [Internet]. 2023, 1. Available from: https://academic.oup.com/healthaffairsscholar/article/doi/10.1093/haschl/qxad005/7203674.
- Wu S, Thawani R. Tumor-agnostic therapies in practice: Challenges, innovations, and future perspectives. Cancers (Basel) [Internet]. 2025, 17. Available from: https://www.mdpi.com/2072-6694/17/5/801.
- Conran CA, Brendler C, Xu J. Personalized prostate cancer care: from screening to treatment. Asian J Androl. 2016, 18, 505–508. [Google Scholar] [CrossRef]
- Houwink E, Sollie A, Numans M, Cornel M. Proposed roadmap to stepwise integration of genetics in family medicine and clinical research. Clin Transl Med. 2013, 2, 5–5. [Google Scholar] [CrossRef]
- Tommel J, Kenis D, Lambrechts N, Brohet R, Swysen J, Mollen L, et al. Personal genomes in practice: Exploring citizen and healthcare professionals’ perspectives on personalized genomic medicine and personal health data spaces using a mixed-methods design. Genes (Basel) [Internet]. 2023, 14. Available from: https://www.mdpi.com/2073-4425/14/4/786.
- Houwink E, van Luijk SV, Henneman L, van der Vleuten CVD, Dinant GJ, Cornel M. Genetic educational needs and the role of genetics in primary care: a focus group study with multiple perspectives. BMC Fam Pract. 2011, 12, 5–5. [Google Scholar] [CrossRef]
- Pasic M, Samaan S, Yousef G. Genomic medicine: new frontiers and new challenges. Clin Chem. 2013, 59, 158–167. [CrossRef] [PubMed]
- An L, Liu Y, Liu Y. Advancements in Circulating Tumor Cell Detection for Early Cancer Diagnosis: An Integration of Machine Learning Algorithms with Microfluidic Technologies. Biosensors. 2025.
- Wang Y, Feng W. Cancer-related psychosocial challenges. Gen Psychiatr [Internet]. 2022, 35. Available from: https://gpsych.bmj.com/lookup/doi/10.1136/gpsych-2022-100871.
- Callier SL, Abudu R, Mehlman M, Singer M, Neuhauser D, Anan CC, et al. Ethical, legal, and social implications of personalized genomic medicine research: Current literature and suggestions for the future. Z-Medicine eJournal [Internet]. 2016, Available from: https://onlinelibrary.wiley.com/doi/10.1111/bioe.12285.
- Lemke A, Wolf WA, Hebert-Beirne J, Smith ME. Public and biobank participant attitudes toward genetic research participation and data sharing. Public Health Genomics. 2010, 13, 368–377. [Google Scholar] [CrossRef]
- Chua IS, Gaziel-Yablowitz M, Korach Z, Kehl K, Levitan N, Arriaga Y, et al. Artificial intelligence in oncology: Path to implementation. Cancer Med. 2021, 10, 4138–4149. [Google Scholar] [CrossRef] [PubMed]
- Pando A, Trivedi R, Pauwels J, Nowakowska J, Cavina B, Falkman L, et al. Unlocking the promise of liquid biopsies in precision oncology. The Journal of Liquid Biopsy [Internet]. 2024, 3. Available from: https://linkinghub.elsevier.com/retrieve/pii/S295019542400016X.
- Anderson MW, Schrijver I. Next generation DNA sequencing and the future of genomic medicine. Genes (Basel). 2010, 1, 38–69. [Google Scholar] [CrossRef] [PubMed]
- Manolio T, Chisholm R, Ozenberger B, Roden D, Williams MS, Wilson R, et al. Implementing genomic medicine in the clinic: the future is here. Genet Med. 2013, 15, 258–267. [Google Scholar] [CrossRef]
- Milko L, Khoury M. Editorial: DNA-based population screening for precision public health. Front Genet [Internet]. 2022, 13. Available from: https://www.frontiersin.org/articles/10.3389/fgene.2022.1061329/full.
- Wijetunga N, Yahalom J, Imber B, FitzGerald T, Hilal L, Hong JH, et al. The art of war: using genetic insights to understand and harness radiation sensitivity in hematologic malignancies. Front Oncol [Internet]. 2025, 14. Available from: https://www.frontiersin.org/articles/10.3389/fonc.2024.1478078/full.
- Adalsteinsson VA, Ha G, Freeman SS, Choudhury AD, Stover DG, Parsons HA, et al. Scalable whole-exome sequencing of cell-free DNA reveals high concordance with metastatic tumors. Nat Commun. 2017, 8, 1324. [Google Scholar] [CrossRef]
- Mathios D, Johansen JS, Cristiano S, Medina J, Phallen J, Larsen K, et al. Detection and characterization of lung cancer using cell-free DNA fragmentomes. Nat Commun [Internet]. 2021, 12. Available from: https://www.nature.com/articles/s41467-021-24994-w.
- Ueno-Yokohata H, Okita H, Nakasato K, Hishiki T, Shirai R, Tsujimoto S, et al. Preoperative diagnosis of clear cell sarcoma of the kidney by detection of BCOR internal tandem duplication in circulating tumor DNA. Genes Chromosomes Cancer. 2018, 57, 525–529. [Google Scholar] [CrossRef] [PubMed]
- Spreafico A, Hansen A, Abdul Razak AA, Bedard P, Siu L. The future of clinical trial design in oncology. Cancer Discov. 2021, 11, 822–837. [Google Scholar] [CrossRef] [PubMed]
- Betz M, Massard V, Gilson P, Witz A, Dardare J, Harlé A, et al. ESR1 gene mutations and liquid biopsy in ER-positive breast cancers: A small step forward, a giant leap for personalization of endocrine therapy? Cancers (Basel). 2023, 15, 5169. [Google Scholar] [CrossRef]
- Chen X, Gole J, Gore A, He Q, Lu M, Min J, et al. Non-invasive early detection of cancer four years before conventional diagnosis using a blood test. Nat Commun [Internet]. 2020, 11. Available from: https://www.nature.com/articles/s41467-020-17316-z.
- Aggarwal C, Thompson JC, Black TA, Katz SI, Fan R, Yee SS, et al. Clinical implications of plasma-based genotyping with the delivery of personalized therapy in metastatic non-small cell lung cancer. JAMA Oncol. 2019, 5, 173–180. [Google Scholar] [CrossRef] [PubMed]
- Brannon AR, Jayakumaran G, Diosdado M, Patel J, Razumova A, Hu Y, et al. Enhanced specificity of clinical high-sensitivity tumor mutation profiling in cell-free DNA via paired normal sequencing using MSK-ACCESS. Nat Commun. 2021.
- Mack P, Banks K, Espenschied C, Burich RA, Zill OA, Lee CE, et al. Spectrum of driver mutations and clinical impact of circulating tumor DNA analysis in non–small cell lung cancer: Analysis of over 8000 cases. Cancer. 2020, 126, 3219–3228. [Google Scholar] [CrossRef]
- Moser T, Waldispuehl-Geigl J, Belic J, Weber S, Zhou Q, Hasenleithner SO, et al. On-treatment measurements of circulating tumor DNA during FOLFOX therapy in patients with colorectal cancer. NPJ Precis Oncol [Internet]. 2020, 4. Available from: https://www.nature.com/articles/s41698-020-00134-3.
- Weber S, van der Leest P, Donker HC, Schlange T, Timens W, Tamminga M, et al. Dynamic changes of circulating tumor DNA predict clinical outcome in patients with advanced non–small-cell lung cancer treated with Immune checkpoint inhibitors. JCO Precis Oncol. 2021, 5, 1540–1553. [Google Scholar]
- Azad TD, Chaudhuri AA, Fang P, Qiao Y, Esfahani MS, Chabon JJ, et al. Circulating tumor DNA analysis for detection of minimal residual disease after chemoradiotherapy for localized esophageal cancer. Gastroenterology. 2020, 158, 494–505.e6. [Google Scholar] [CrossRef]
- Dagogo-Jack I, Brannon A, Ferris LA, Campbell CD, Lin JJ, Schultz KR, et al. Tracking the Evolution of Resistance to ALK Tyrosine Kinase Inhibitors through Longitudinal Analysis of Circulating Tumor DNA. Tracking the Evolution of Resistance to ALK Tyrosine Kinase Inhibitors through Longitudinal Analysis of Circulating Tumor DNA. JCO Precis Oncol. 2018.
- Recondo G, Bahcall M, Spurr LF, Che J, Ricciuti B, Leonardi G, et al. Molecular mechanisms of acquired resistance to MET tyrosine kinase inhibitors in patients with MET Exon 14–mutant NSCLC. Clin Cancer Res. 2020, 26, 2615–2625. [Google Scholar] [CrossRef] [PubMed]
- Zhou Q, Perakis SO, Ulz P, Mohan S, Riedl J, Talakic E, et al. Cell-free DNA analysis reveals POLR1D-mediated resistance to bevacizumab in colorectal cancer. Genome Med [Internet]. 2020, 12. Available from: https://genomemedicine.biomedcentral.com/articles/10.1186/s13073-020-0719-6.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
