
Circulating Tumor DNA (ctDNA) Testing to Predict Response in Solid Tumors
Key Takeaways
- ctDNA offers a non-invasive method for monitoring tumor burden and treatment response, often preceding radiographic progression.
- Adaptive clinical trials using ctDNA-guided therapy show potential in improving progression-free survival and reducing chemotherapy exposure.
ctDNA analysis revolutionizes cancer treatment by predicting responses, detecting resistance, and monitoring minimal residual disease in patients.
Characterizing molecular material from solid tumors using body fluids, such as blood, urine, and cerebrospinal fluid, rather than invasive biopsies, represents a major advancement in oncology.1 One of the most widely studied forms of liquid biopsy utilizes circulating tumor DNA (ctDNA), which consists of DNA fragments released into the bloodstream when tumor cells die.2
Initially developed to identify pharmacologically actionable genes, ctDNA analysis is being explored to monitor treatment response, modify treatment courses, detect the emergence of resistance mutations, and refine prognosis. This article summarizes current data supporting ctDNA as a predictor of response, delving into key studies, remaining challenges, and future research directions.
ctDNA in Advanced Disease: Monitoring Response
CtDNA provides a real-time measure of tumor burden and response to systemic therapy in metastatic and unresectable cancers. The ARTEMIS-PC trial (NCT06043921)3 in unresectable pancreatic cancer demonstrated that ctDNA clearance correlated with higher response rates, prolonged progression-free survival (PFS; 9.0 vs 3.5 months), and earlier relapse detection compared with imaging. Notably, ctDNA changes often preceded radiographic progression by approximately 3 months, suggesting that ctDNA may serve as an early, more sensitive indicator of treatment efficacy.4,5
Similar observations are reported across other solid tumors, including non–small cell lung cancer (NSCLC), where the time between ctDNA positivity and radiologic progression ranges from 2.8 to 12.6 months.6 However, there are currently no prospective studies confirming that switching therapy solely based on a lack of ctDNA response, without evidence of radiologic progression, improves patient outcomes. Because treatment options are often limited in advanced disease, premature therapy changes risk depleting effective lines of treatment without proven survival benefit. Defining optimal thresholds and timing for ctDNA-based intervention remains an unmet need.
Adaptive Study Designs and ctDNA-Guided Therapy
Adaptive clinical trial designs incorporating ctDNA response are 1 approach to evaluating the effects of ctDNA-guided treatment decisions. These study designs allow for real-time modification of treatment arms based on molecular response data. In a recent trial in NSCLC, ctDNA-guided therapy adaptation significantly improved PFS and reduced platinum-based chemotherapy exposure compared with PD-L1 tumor proportion score-informed treatment.7,8 These findings highlight the potential utility of ctDNA as a biomarker to individualize therapy intensity and sequence. Future ctDNA-adaptive trials may help clarify whether early treatment modifications guided by molecular response can enhance survival while sparing additional treatment for some patients with advanced disease.
Detection of Resistance Mutations
Another clinical application of ctDNA lies in the detection of emergent resistance mutations, enabling therapy adjustments before clinical progression. Early studies demonstrated that ctDNA assays can detect resistance mutations before clinical or radiographic progression.6
In estrogen receptor-positive/HER2-negative breast cancer, the PADA-1 and SERENA-6 trials (NCT03079011; NCT04964934)9,10 demonstrated that serial ctDNA testing can identify ESR1 mutations that develop with resistance to aromatase inhibitors. Switching therapy to a selective estrogen receptor degrader upon ctDNA mutation detection significantly improved PFS compared with waiting until radiographic progression.11,12 These results provide compelling evidence that early ctDNA-guided therapeutic intervention may improve outcomes in selected malignancies and support the integration of serial ctDNA monitoring into clinical trial design.
Detecting Molecular Residual Disease (MRD)
In early-stage solid tumors, ctDNA-based tests are being used to detect MRD following curative-intent surgery or definitive radiation. Two main ctDNA assay approaches exist: tumor agnostic and tumor informed.
Tumor-agnostic assays employ predefined gene panels specific to each cancer type. They are advantageous for their rapid turnaround and ability to capture tumor heterogeneity, but they offer lower sensitivity and specificity because they are not tailored to individual mutational profiles.
Tumor-informed assays are patient specific and are designed using mutations identified from a tumor tissue sample. These tests achieve higher sensitivity but require sufficient tumor tissue, longer assay development times, and may miss relapse driven by new clonal genotypes.13
Prognostic Value of MRD Detection
ctDNA-based MRD testing has demonstrated prognostic value across multiple solid tumors. Persistent ctDNA-positivity following surgery or definitive radiation is consistently associated with shorter disease-free survival (DFS) and overall survival (OS).14 A meta-analysis of stage 2 colorectal cancer (CRC) showed that postoperative ctDNA positivity conferred an approximate 3.7-fold higher recurrence risk. In the absence of adjuvant chemotherapy, ctDNA-positive patients had an approximate 5.6-fold higher recurrence risk.15 Dynamic changes in ctDNA levels correlate with tumor volume reduction and disease recurrence, supporting its use as a real-time biomarker of treatment efficacy.16
However, optimal clinical management for patients who are ctDNA-positive following definitive therapy remains undefined. Whether therapy intensification or alteration in response to ctDNA persistence can improve outcomes is an active and critical area for future studies.
MRD and Systemic Therapy Decisions
Several recent studies explored whether ctDNA positivity after surgery can identify patients who benefit from adjuvant systemic therapy. The GALAXY study (UMIN000039205)17 in CRC and IMvigor010 (NCT02450331)18 in urothelial cancer found that patients with ctDNA positivity experienced improved DFS and OS, respectively, with adjuvant therapy, whereas those with ctDNA negativity did not derive benefit from its addition.19,20
In contrast, the DYNAMIC-III trial (ACTRN12617001566325)21 in colon cancer prospectively assigned treatment based on ctDNA status in 1 group, while other participants received standard management. Although this ctDNA-guided, treatment-escalation approach did not improve DFS among ctDNA-positive patients, it safely spared ctDNA-negative patients from additional chemotherapy.22
These studies suggest that ctDNA may serve as a biomarker to tailor the intensity of adjuvant therapy. Still, integration of ctDNA testing into clinical guidelines remains cautious. Current National Comprehensive Cancer Network guidelines for bladder and colon cancer state that while ctDNA is prognostic, it is not yet predictive, and routine use of ctDNA assays for surveillance or therapeutic decision-making outside clinical trials is not recommended.23,24
Challenges and Limitations
Despite its promise, several challenges impede the widespread adoption of ctDNA testing. A significant limitation is the lack of standardized thresholds for defining molecular response. Assay heterogeneity further complicates interpretation, as differences in sensitivity, specificity, and limit of detection can yield different results across different studies.25 Existing data have limited generalizability as they are primarily derived from retrospective and/or small cohort studies. The optimal frequency and timing of ctDNA testing have not been established, and cost-effectiveness remains uncertain, particularly for serial monitoring. Clinical benefit from treatment modification based solely on ctDNA findings has yet to be demonstrated in large, randomized trials. Addressing these gaps requires harmonizing assay methodologies, establishing clinically meaningful ctDNA cutoffs, and rigorously evaluating in prospective, interventional studies whether ctDNA-guided management can translate into improved patient outcomes.
Looking Forward
Future research should focus on refining ctDNA quantification and clinical workflow integration. Defining validated thresholds for ctDNA and optimal sampling intervals is critical to standardizing interpretation. Parallel cost-effectiveness analyses are necessary to support reimbursement and clinical adoption. As sequencing technologies continue to evolve, ctDNA may improve the assessment of therapy response and identification of emerging resistance mutations, facilitating earlier therapeutic interventions. Ultimately, improvements in assay sensitivity (which have improved over the past few years), and workflow integration will enhance ctDNA’s precision, reliability, and clinical utility. This will position ctDNA as a vital component of precision oncology, bridging molecular profiling with dynamic disease monitoring.25
Conclusion
ctDNA analysis is a transformative step in personalized oncology care. Beyond identifying actionable mutations, ctDNA may also predict therapeutic response, detect resistance, and detect MRD. While integration into standard practice depends on validation from ongoing and future studies, accumulating evidence supports ctDNA’s potential to refine prognosis, individualize treatment plans, and reduce chemotherapy exposure in some patient groups. ctDNA-guided adaptive trials will be critical in determining its role in clinical decision-making and solidifying liquid biopsy as a cornerstone of modern oncology.
REFERENCES
Liquid biopsy. NCI Dictionary of Cancer Terms, National Cancer Institute. Accessed October 17, 2025. https://
www.cancer.gov/publications/dictionaries/cancer-terms/def/liquid- biopsyCirculating tumor DNA. NCI Dictionary of Cancer Terms, National Cancer Institute. Accessed October 17, 2025. https://
www.cancer.gov/publications/dictionaries/cancer- terms/def/circulating-tumor-dnaMulticenter study of circulating tumor DNA in patients with pancreatic cancer using a personalized panel (ARTEMIS-PC). Clinicaltrials.gov. Updated September 21, 2023. Accessed October 30, 2025. https://www.clinicaltrials.gov/study/NCT06043921
Shibuki T, Bando H, Ueno M, et al. Utility of circulating tumor DNA in patients with unresectable pancreatic cancer using a patient-specific panel: ARTEMIS-PC study. J Clin Oncol. 2025;43(suppl 4):684. doi:10.1200/JCO.2025.43.4_suppl.684
Norton A. ctDNA analysis shows promise for treatment monitoring in advanced pancreatic cancer. ASCO Daily News. January 24, 2025. Accessed October 17, 2025. https://dailynews.ascopubs.org/do/ctdna-analysis-shows-promise-treatment-monitoring-advanced-pancreatic-cancer
Normanno N, Morabito A, Rachiglio AM, et al. Circulating tumour DNA in early stage and locally advanced NSCLC: ready for clinical implementation?. Nat Rev Clin Oncol. 2025;22(3):215-231. doi:10.1038/s41571-024-00985-w
Rotow JK, Heavey G, Nishino M, et al. Plasma-guided adaptive first-line chemoimmunotherapy for non-small cell lung cancer (NSCLC). J Clin Oncol. 2025;43(suppl 16):8515. doi:10.1200/JCO.2025.43.16_suppl.8515
Rotow JK, Heavey G, Nishino M, et al. Plasma-guided adaptive first-line chemoimmunotherapy for non-small cell lung cancer (NSCLC). Presented at: American Society of Clinical Oncology (ASCO) Annual Meeting; October 10-11, 2025; Chicago, IL. https://
www.asco.org/abstracts- presentations/ABSTRACT489262Palbociclib and circulating tumor DNA for ESR1 mutation detection (PADA-1). Clinicaltrials.gov. Updated November 12, 2024. Accessed October 30, 2025.
https://clinicaltrials.gov/study/NCT03079011 Phase III study to assess AZD9833+ CDK4/6 inhibitor in HR+/HER2-MBC with detectable ESR1m before progression (SERENA-6) (SERENA-6). Clinicaltrials.gov. Updated October 7, 2025. Accessed October 30, 2025. https://clinicaltrials.gov/study/NCT04964934
Bidard FC, Hardy-Bessard AC, Dalenc F, et al. Switch to fulvestrant and palbociclib versus no switch in advanced breast cancer with rising ESR1 mutation during aromatase inhibitor and palbociclib therapy (PADA-1): a randomised,open-label, multicentre, phase 3 trial. Lancet Oncol. 2022;23(11):1367-1377. doi:10.1016/S1470-2045(22)00555-1
Bidard FC, Mayer EL, Park YH, et al. First-line camizestrant for emerging ESR1-mutated advanced breast cancer. N Engl J Med. 2025;393(6):569-580. doi:10.1056/NEJMoa2502929
Cohen SA, Liu MC, Aleshin A. Practical recommendations for using ctDNA in clinical decision making. Nature. 2023;619(7969):259-268. doi:10.1038/s41586-023-06225-y
Kasi PM, Fehringer G, Taniguchi H,et al. Impact of circulating tumor DNA-based detection of molecular residual disease on the conduct and design of clinical trials for solid tumors. JCO Precis Oncol. 2022;6:e2100181. doi:10.1200/PO.21.00181
Negro S, Pulvirenti A, Trento C, et al. Circulating tumor DNA as a real-time biomarker for minimal residual disease and recurrence prediction in stage II colorectal cancer: a systematic review and meta-analysis. Int J Mol Sci. 2025;26(6):2486. doi:10.3390/ijms26062486
Schøler LV, Reinert T, Ørntoft MW, et al. Clinical implications of monitoring circulating tumor DNA in patients with colorectal cancer. Clin Cancer Res. 2017;23(18):5437-5445. doi:10.1158/1078-0432.CCR-17-0510
Genetic alterations and clinical record in radically resected colorectal cancer revealed by liquid biopsy and whole exome analysis. UMIN-CTR. Updated June 3, 2025. Accessed October 30, 2025. https://center6.umin.ac.jp/cgi-open-bin/ctr_e/ctr_view.cgi?recptno=R000044197
A study of atezolizumab versus observation as adjuvant therapy in participants with high-risk muscle-invasive urothelial carcinoma (UC) after surgical resection (IMvigor010). Clinicaltrials.gov. Updated June 18, 2023. Accessed October 30, 2025. https://clinicaltrials.gov/study/NCT02450331
Nakamura Y, Watanabe J, Akazawa N, et al. ctDNA-based molecular residual disease and survival in resectable colorectal cancer. Nat Med. 2024;30(11):3272-3283. doi:10.1038/s41591-024-03254-6
Powles T, Assaf ZJ, Degaonkar V, et al. Updated overall survival by circulating tumor DNA status from the phase 3 IMvigor010 trial: adjuvant atezolizumab versus observation in muscle-invasive urothelial carcinoma. Eur Urol. 2024;85(2):114-122. doi:10.1016/j.eururo.2023.06.007
Circulating tumour DNA analysis informing adjuvant chemotherapy in stage III colon cancer: a multicentre phase II/III randomised controlled study (DYNAMIC-III). ANZCTR. Updated May 11, 2025. Accessed October 30, 2025. https://www.anzctr.org.au/Trial/Registration/TrialReview.aspx?id=373948
Tie J, Wang Y, Lo SN, et al. Circulating tumor DNA analysis guiding adjuvant therapy in stage II colon cancer: 5-year outcomes of the randomized DYNAMIC trial. Nat Med. 2025;31(5):1509-1518. doi:10.1038/s41591-025-03579-w
NCCN. Clinical Practice Guidelines in Oncology. Bladder cancer, version 2.2025. Accessed October 9, 2025. https://www.nccn.org/guidelines/guidelines-detail?category=1&id=1417
NCCN. Clinical Practice Guidelines in Oncology. Colon cancer, version 4.2025. Accessed October 9, 2025. https://www.nccn.org/guidelines/guidelines-detail?id=1428
Stetson D, Labrousse P, Russell H, et al. Next-generation molecular residual disease assays: do we have the tools to evaluate them properly?. J Clin Oncol. 2024;42(23):2736-2740. doi:10.1200/JCO.23.02301
Newsletter
Stay informed on drug updates, treatment guidelines, and pharmacy practice trends—subscribe to Pharmacy Times for weekly clinical insights.
















































































































































































































