Next Article in Journal
Noninvasive Cell Population Profiling of Normal and Dysplastic Cervical Biofluids by Multicolor Flow Cytometry as a Promising Tool for Companion Diagnostics
Previous Article in Journal
Unveiling Enhancer RNAs in Gliomas: A Systematic Review and Qualitative Synthesis
Previous Article in Special Issue
Methylation Analyses in Liquid Biopsy of Lung Cancer Patients: A Novel and Intriguing Approach Against Resistance to Target Therapies and Immunotherapies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Circulating Tumor DNA as a Prognostic and Predictive Biomarker in Lung Cancer

1
Department of Oncology, Albert Einstein College of Medicine/Montefiore Medical Center/Montefiore Einstein Comprehensive Cancer Center, Bronx, New York, NY 10461, USA
2
Department of Internal Medicine, NYC Health + Hospitals/Jacobi Medical Center, Bronx, New York, NY 10461, USA
*
Author to whom correspondence should be addressed.
Cancers 2025, 17(20), 3327; https://doi.org/10.3390/cancers17203327
Submission received: 1 September 2025 / Revised: 6 October 2025 / Accepted: 8 October 2025 / Published: 15 October 2025

Abstract

Simple Summary

Liquid biopsy is a type of blood test which detects circulating tumor DNA (ctDNA). This is a key advance in precision medicine which can help in diagnosing, treating, and monitoring lung cancer. This article reviews the current applications of ctDNA in the context of lung cancer, particularly non-small cell lung cancer (NSCLC). Research to date reveal ctDNA is useful in detecting actionable genomic alterations, helps guide treatment, and may predict how the cancer may respond to treatment and evolve. Minimal residual disease detection is another key area of interest. ctDNA detection and tracking have powerful potential uses, and many studies are currently underway.

Abstract

Background/Objectives: Lung cancer remains a leading cause of cancer-related mortality worldwide. In recent years, the development of liquid biopsy, or ctDNA detection in body fluids, particularly blood, has been shown to be effective in detection, genotyping, prognostication, and evaluating therapy response, particularly in non-small cell lung cancer (NSCLC). Methods: In this review, we present a summary of the current landscape of ctDNA, applications, and limitations, as well as future areas of research. Results/Conclusions: Though not yet in its prime, ctDNA detection and tracking have powerful current and potential uses, including treatment selection, prognostication, and risk stratification.

1. Introduction

Lung cancer is one of the most diagnosed cancers worldwide at 12.4% of total cancer diagnoses [1] and the second most diagnosed cancer in the United States in both men and women. It remains one of the leading causes of cancer-related deaths, causing approximately 131,584 deaths in the United States alone in 2023 [2].
Around 85% of lung cancer is non-small cell lung cancer (NSCLC) [3]. In recent years, the discovery of driver mutations in NSCLC and the utilization of mutation-targeted therapies have significantly improved therapeutic options for patients. Due to this, tumor genotyping is of the utmost importance and is traditionally performed on specimens that are obtained through invasive means, like tissue biopsies. Liquid biopsy is a non-invasive method which analyzes circulating tumor DNA (ctDNA) and can assist in genotyping the tumor [4,5,6].
Cell-free DNA (cfDNA) comprises partially degraded DNA fragments that are not encased in cells. Circulating tumor DNA (ctDNA) in particular comprises short, double-stranded DNA fragments that are released by tumor cells into blood and, depending on the tumor, other biologic fluids like cerebrospinal fluid or urine. ctDNA shed from cancer cells holds unique genetic alterations from where the tumor originated [7,8].
Various international oncology societies currently recommend the use of liquid biopsy for tumor molecular genotyping in lung cancer.
Liquid biopsy is a term used to describe sampling of body fluid, typically blood, to detect tumor cells and cancer cell DNA [6,8]. There are multiple advantages, including being minimally invasive. ctDNA is also thought to better reflect intra-tumoral heterogeneity, given the ability to capture information from different tumor sub-clones. The turnaround time of ctDNA detection is relatively quick, and it can be performed in the office without needing to plan for a biopsy [9,10].
There are various methods to detect ctDNA. Currently, ctDNA testing is typically performed using polymerase chain reaction (PCR)-based or next-generation sequencing (NGS), though NGS is more commercially utilized [11,12,13]. Digital PCR-based methods detect specific DNA sequences and have been preferred in this context over prior PCR methods, but they interrogate only specific numbers of mutations at a time and cannot easily detect copy number alterations and rearrangements [14,15,16]. TAM-seq and CAPP-seq are NGS-based assays. They retain high sensitivity and specificity and can detect multiple mutations of interest, as well as copy number alterations [17,18]. However, for NGS assays, turnaround times can be longer, the cost is higher than for PCR, and they require bioinformatics result interpretation [16,17,18,19].
Fragmentomics analyzes the size patterns of cfDNA and is another area of ongoing research [20,21,22]. Methylation patterns of ctDNA can mirror those in cancerous tissue, and studies have been investigating applications in detection and monitoring [23].
Most commonly, ctDNA testing is performed on blood samples, but it can also be performed in cerebrospinal fluid (CSF) or body fluid; for example, pleural fluid ctDNA analysis of patients with brain metastasis has revealed either the absence or very low levels of ctDNA, possibly because of the blood–brain barrier [24]. However, one study found that ctDNA in CSF had a higher sensitivity than plasma for CNS genomic alterations [25], and another study found that the EGFR mutation detected was the same in the CSF and blood [26,27]. Other body fluids have also been studied, such as urine, saliva, and bronchoalveolar lavage fluid, but they do not have clinically relevant applications yet [28,29,30,31].
In NSCLC, ctDNA has been evidenced to be effective in detection, genotyping, and evaluating therapy response, though further work is required in establishing standardized practices.

2. Applications

2.1. Detection/Screening

In patients who qualify for lung cancer screening, low-dose CT is recommended for early detection, but there are suboptimal compliance rates due to cost and difficulty [20]. Detection of cancers with ctDNA is being studied as a tool to aid in screening to augment low-dose CT chest screening [32,33]. Limitations include early-stage cancers being harder to detect as ctDNA level has been shown to be proportional to tumor burden [31,32,34,35]. There has been work on testing machine learning models to diagnose lung cancer, combining ctDNA, imaging, patient risk factors, and fragmentation features [20,36].
ctDNA levels can fluctuate in response to treatment and thus can aid in monitoring disease progression and/or recurrence. Overall, there is a positive correlation with tumor burden, and higher ctDNA levels in the blood have been linked to poorer prognosis in NSCLC [37,38,39]. There are various clinical trials that aim to study the role of ctDNA in lung cancer screening and stratification such as the Lung Cancer Mutation Consortium (NCT04712877) and DNA Evaluation of Fragments for Early Interception—Lung Cancer Training Study (DELFI-L101 Study) (NCT04825834) [24].

2.2. Genotyping and Detecting Actionable Genomic Alterations (AGAs)

ctDNA can be used to identify specific mutations in lung cancer and guide selection of targeted therapy, which has significantly improved outcomes, especially in recent years [32]. It is current practice to test for EGFR, KRAS, ALK, ROS1, BRAF, NTRK 1/2/3, MET, RET, ERBB2 (HER2), and NRG1 via liquid or tissue biopsy at diagnosis, recurrence, and/or progression. Figure 1 shows the prevalence of different mutations in lung adenocarcinoma, which makes up about 45% of NSCLC. Around 65% of these patients may harbor a clinically actionable genomic alteration [1,2,3,4,5,6,40].
Molecular genotyping from liquid biopsy can be as effective as from a tissue specimen, depending on the specimen characteristics. Specific genomic alterations can also be monitored longitudinally, like in copy number mutations over time, like EGFR after surgery. Liquid biopsy can be performed at any stage of disease including at the time of progression to evaluate for any new potential actional genomic alterations to guide treatment decisions [15,41,42].
There has been some evidence of increased adherence to standard of care and overall more prompt care, though this relationship is likely not causal. For instance, Thompson et al. demonstrated that the inclusion of plasma-based NGS testing led to higher rates of guideline-recommended treatment (74% vs. 46%, p = 0.0005), but it was noted that physicians had test results available before the first patient visit much more frequently when a plasma-based strategy was used, which is likely a contributing factor [43].

2.3. Utilizing ctDNA in Resectable Disease

Approximately 25% to 30% of patients with NSCLC present at stages I to IIIA [11,12,13]. Surgery remains the definitive treatment for early-stage NSCLC. Unfortunately, 30–50% of NSCLC patients will relapse after surgery. Adjuvant treatment has improved overall survival (OS) by around 5%, but a significant proportion of patients develop recurrence, resulting in a median OS rate ranging from 84% at stage IA to 36% at stage IIIA. Minimal residual disease (MRD) refers to the small number of remaining cancer cells, or micrometastasis, during the course or after the completion of treatment. An effective strategy to eradicate MRD and therefore minimize the chance of post-operative recurrence consists of adjuvant therapy. Currently disease is followed using imaging and monitored using the Response Evaluation Criteria in Solid Tumors (RECIST) criteria. CT imaging is most commonly used, and small lesions may be missed given the limit of detection is 2–3 mm. Using ctDNA to track MRD and perform appropriate risk stratification and treatment decisions is an area of research which is becoming more studied.
ctDNA MRD analyses can be tumor-informed or tumor-agnostic [44] and depend on prior knowledge of tissue sequencing. Tumor-informed analysis requires sequencing of tumor tissue to identify mutations to track and is more sensitive. One of the main approaches used has been to employ NGS to track mutations since this facilitates the tracking of multiple mutations in parallel, minimizes the number of tests needed, and increases sensitivity [34], but it requires longer assay development times and may not capture newly evolved mutations [33,45]. Tumor-agnostic methods utilize epigenetic features such as DNA methylation and DNA fragmentation patterns to detect ctDNA. Currently, the sensitivity of these methods is worse compared to tumor-informed approaches and thus, at this time, they are less applicable in MRD tracking [45].
Studies using a variety of tumor-informed NGS-based ctDNA MRD assays have demonstrated that the detection of residual ctDNA after completion of treatment means a higher risk of recurrence [11,12,13,39,45]. There has been variation among them in terms of when ctDNA is drawn after definitive treatment. Lower ctDNA shedding has also been associated with the adenocarcinoma histological subtype as opposed to squamous cell carcinoma [13].
There are two main methodologies for the MRD testing timeline—longitudinal or serial measurements over time and landmark or singular designated time points [19]. Though there is no clear recommendation for one methodology, studies will typically utilize one of the other. In a systematic review of 13 studies that performed ctDNA analysis for post-operative MRD detection in stage I-III NSCLC patients, higher overall sensitivity was achieved in studies that performed longitudinal MRD analysis compared to landmark analysis [18,19].
Chauduri et al. showed that longitudinal tracking of multiple mutations can significantly increase the sensitivity of post-treatment MRD detection (94% vs. 58% for a single mutation p = 0.001) [11,12]. In a prospective study by Zhang et al., landmark analysis was utilized at one month post-operatively and at one month post-adjuvant therapy. For those who received adjuvant chemotherapy, 86.6% of patients with negative MRD status remained disease-free [39]. Different studies utilize different time points in both methodologies, but obtaining measurements at baseline, post-operatively, and before and after systemic therapy seems pertinent, as these landmark timepoints are clinically relevant, though more trials are needed [39].
One of the key applications of interest in MRD testing is guiding treatment decisions on when to de-escalate treatment, such as in decisions around adjuvant therapy.

2.3.1. Chemoimmunotherapy in Resectable Disease

Neoadjuvant chemotherapy and/or immunotherapy is an important treatment strategy in resectable NSCLC [46]. The phase II NADIM study that included patients with resectable stage IIIA disease showed that clearance of ctDNA after three cycles of neoadjuvant nivolumab plus chemotherapy was significantly correlated with longer progression-free survival (PFS) (HR: 0.16, 95% CI: 0.03–0.73) and overall survival (OS) (HR: 0.05, 95% CI: 0–0.62). It is of note that analyses excluded patients with undetectable ctDNA at baseline [47].
The phase III AEGEAN trial used peri-operative durvalumab plus neoadjuvant chemotherapy in patients with resectable stage II-IIIB NSCLC and showed that patients achieving ctDNA clearance after three cycles of neoadjuvant therapy had significantly better event-free survival (EFS) outcomes compared with those with residual ctDNA, especially in the durvalumab-plus-chemotherapy arm (HR: 0.26; 95% CI: 0.13–0.54) [48]. Patients with no detectable ctDNA before surgery had better survival outcomes even in the absence of pathologic complete response [48,49].
It is important to note that the negative prognostic value of ctDNA clearance is suboptimal at this time. Analyses from CheckMate 816 (neoadjuvant nivolumab plus chemotherapy) and CheckMate 77T (peri-operative nivolumab plus chemotherapy trials in resectable NSCLC) both showed that although clearance of ctDNA was correlated with a higher pCR rate, as many as 50% of patients with ctDNA clearance did not achieve pCR, implying that ctDNA negativity may not necessarily translate into eradication of residual tumor cells [50].

2.3.2. Targeted Therapies in Resectable Disease

Targeted therapies have significantly changed the landscape of treatment for EGFR-mutated NSCLC in general, though it is not yet clear how MRD can guide therapy duration and personalize treatment to patients. Herbst et al. in post hoc ADAURA trial analysis looked at patients that would benefit beyond three years using MRD to guide therapy. In this study, MRD preceded DFS events in most patients [51]. DFS and MRD event-free status was maintained for most patients during adjuvant osimertinib treatment and posttreatment follow-up, with most MRD or DFS events occurring after osimertinib treatment discontinuation or completion [52].
The TRACERx study used ctDNA to track a median of 200 mutations in resected NSCLC tissue. It utilized a tumor-informed approach (anchored multiplex PCR) to detect MRD in early-stage NSCLC after surgery. It found that 25% of patients were positive for MRD within 120 days of surgery, and 3- to 6-monthly ctDNA surveillance identified relapse in a further ~20% of landmark-ctDNA-negative patients. A bioinformatic tool, ECLIPSE, was used to track sub-clonal architecture at low ctDNA levels, which identified patients with polyclonal metastatic dissemination and was associated with poor clinical outcomes [51,53].
Jung et al. studied stage I–IIIA EGFR-mutated NSCLC patients who underwent curative resection. ctDNA was measured prior to surgery, after curative surgery, and then at different time points with increasing interval lengths up to 5 years or until clinically definite recurrence [54]. Patients were then classified into three groups: baseline ctDNA-negative, baseline ctDNA-positive but post-operative MRD-negative, and baseline ctDNA-positive and post-operative MRD-positive. The study found that the 3-year DFS rate was significantly different among the three groups (84% versus 78% versus 50%, p = 0.02), suggesting the possible future development of ctDNA MRD status-based risk stratification [54]. Table 1 shows a concise summary of key studies highlighting resectable NSCLC and ctDNA applications and outcomes.

2.4. Locally Advanced, Metastatic, and Recurrent NSCLC

Like in resectable disease, tracking changes using RECIST criteria represents the primary quantification of response and progression [55,59,62,63,64]. In advanced disease, the utility of ctDNA lies in the ability to track it over time to measure progression prior to radiographic progression, and, in theory, change treatment if needed, to improve patient outcomes. Additionally, ctDNA tracking after chemoradiation or consolidation therapy to guide therapy is another area of interest [56,65,66,67,68]. This is not yet a guideline-directed application.
Studies have shown that detecting ctDNA after chemoradiation is associated with worse prognosis, prompting further investigation [69]. Moding et al. studied samples from patients treated with chemoradiation for locally advanced NSCLC, including patients receiving consolidation immunotherapy. Patients ctDNA-negative after chemoradiation had better outcomes, and outcomes of those with MRD after chemoradiation were significantly improved by consolidation immunotherapy [69]. In a prospective observational study, Gale et al. found that patients with ctDNA negativity in all plasma samples and those with ctDNA clearance (previously positive) had longer PFS and OS than patients with persistent ctDNA detection [57]. In certain therapeutic scenarios, the relationship of ctDNA may help guide one towards a specific therapeutic [70,71,72]. For patients being treated for advanced disease with amivantamab plus lazertinib, there have been improved outcomes in high-risk patients with baseline positive ctDNA, as well as without ctDNA clearance, compared to osimertinib [73].
Some studies demonstrate the importance of a molecular response in the relevant biomarker or molecular target irrespective of the target. In metastatic HER2 NSCLC, plasma ctDNA was tested before first-line therapy, and it was found that patients with detectable baseline ctDNA had significantly shorter OS (hazard ratio (HR), 5.25; 95% CI, 1.2–23.9; p = 0.019) [74]. The phase II VISION study evaluated tepotinib in patients with advanced or metastatic NSCLC with a confirmed MET exon 14 skipping mutation and found a high concordance between the molecular cfDNA response and clinical response based on RECIST tumor measurement. Four patients who had decreased levels of cfDNA during treatment did have clinical progression [75]. In patients with KRASG12C-mutated NSCLC treated with sotorasib, those with detectable KRASG12C had inferior progression-free survival (hazard ratio [HR] 2.13 [95% confidence interval [CI]: 1.06–4.30], p ¼ 0.031) and overall survival (HR 2.61 [95% CI: 1.16–5.91], p ¼ 0.017). The disease control rate was significantly higher in those with a molecular response. KRAS amplifications were identified as recurrent treatment-emergent alterations which represent a proposed mechanism of resistance [76].
Table 2 shows a concise summary of key studies highlighting advanced NSCLC and ctDNA and a summary of relevant outcomes. In short, ctDNA detection after an intervention may be a prognostic indicator.

2.5. Resistance

Detecting resistance early to guide treatments and evaluate if there is an impact on outcomes is also a purpose for which ctDNA is being studied [84,87,88,89]. Liquid biopsy is particularly effective in detecting genotypic variations in the background of clonal drift and molecular heterogeneity.
Most patients treated with first- and second-generation TKIs develop resistance early [16,17]; osimertinib is a preferred TKI for this reason [79,81,90,91,92]. However, resistance to osimertinib does occur, and knowing resistance patterns may help predict the course of treatment. The most common mechanism of resistance to osimertinib (which inhibits the T790M isoform) is the development of the C797S isoform, as well as L792X, G796S, L718Q, S768I, G796R, G796D, and G724S [40,93,94]. EGFR gene amplifications and copy number alterations are also mechanisms of resistance [95]. Other mechanisms of resistance include novel fusion events, ERBB2 amplification, and activation of the RAS-MAPK or RAS–PI3K pathway [96,97]. Kato et al. in 2021 performed deep sequencing (CAPP-seq) to analyze ctDNA and were able to identify potentially targetable genetic alterations in patients with osimertinib resistance [43].
Yamaguchi et al. evaluated ctDNA during osimertinib administration as a second-line or further treatment to identify the relationship between EGFR mutation levels and outcomes in patients with advanced NSCLC. In patients with EGFR T790M-positive NSCLC who were receiving osimertinib after prior EGFR-TKI treatment, ctDNA was collected at pretreatment, after 1 month, and at the time of POD. The detection rate of copy numbers of exon 19 deletion, L858R, and T790M in plasma samples was significantly lower 1 month after osimertinib than at pretreatment and significantly higher at PD than at 1 month, whereas that of C797S was significantly higher at PD than at 1 month. No statistically significant difference was observed in the copy numbers of exon 19 deletion, L858R, T790M, and C797S between complete response or partial response and stable disease or PD. Testing based on ctDNA may be helpful in predicting outcomes of osimertinib treatment in T790M-positive NSCLC after previous EGFR-TKI treatment [84].

3. Future Directions

Though there are promising data, further studies are paramount in evaluating the role of using ctDNA to guide care and therapeutic decisions in NSCLC in a way that effects patient outcomes. Both resectable and advanced NSCLC are areas that have seen several promising studies showing prognostic and predictive value, and there are myriad clinical trials that will hopefully help guide clinicians on when and how to change therapeutic course and impacts on survival. Particularly, the ability to risk-stratify patients and escalate or de-escalate based on ctDNA detection would be paramount in limiting toxicities and improving clinical outcomes. Using ctDNA in screening is still immature but could augment low-dose CT and, in the future, could aid in risk stratification. We hope that further studies in these directions lead to consistent incorporation and recommendations for clinicians.

4. Conclusions

The development of liquid biopsy has been a significant development, especially to detect actionable genomic alterations and to choose the appropriate therapy. Though ctDNA has clear indications in guidelines for detecting key biomarkers and has promising applications, it is not uniformly integrated into care and has not reached its full potential yet. The sensitivity and negative predictive value of current ctDNA testing are still low, especially for early-stage cancers. Further research is needed to standardize ctDNA analysis and validate its clinical utility for lung cancer screening, diagnosis, and prognostication.
Additionally, variants can be detected that are not related to the tumor [clonal hematopoiesis of indeterminate potential (CHIP)]. It is also important to note that there are no standard approved practices for MRD testing at this time. There are numerous ongoing clinical trials to evaluate the value of ctDNA in various stages and clinical applications, and ctDNA is effective in the clinical setting so far. Table 3 shows a list, including primary outcomes.

Author Contributions

H.C. was responsible for the idea, drafting, and revisions; P.D. was responsible for drafting, the literature search, writing, and editing; and S.D. performed the literature search and revised. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No datasets were generated or analyzed during the current study.

Acknowledgments

The authors would like to thank the reviewers for their thoughtful questions and comments.

Conflicts of Interest

The authors declare no competing conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
AGAActionable genomic alterations.
BLBaseline.
CFDNACell-free DNA.
CICIConsolidation immune checkpoint inhibition.
CNSCentral nervous system.
CPICheckpoint inhibitor.
CRTChemoradiation therapy.
CTDNACirculating tumor DNA.
DFSDisease-free survival.
DNADeoxyribonucleic acid.
EGFREpidermal growth factor receptor.
ICIImmune checkpoint inhibition.
MPRMajor pathologic response.
MRMolecular response.
MRDMinimal residual disease.
NSCLCNon-small cell lung cancer.
OSOverall survival.
ORROverall response rate.
pCRPathologic complete response.
PDProgression of disease.
PFSProgression-free survival.
PT/PTsPatient/Patients.
RECISTResponse Evaluation Criteria in Solid Tumors.
RFSRecurrence-free survival.
RTRadiotherapy.
TKITyrosine kinase inhibitors.
USPSTFUnited States Preventive Services Taskforce.

References

  1. Bray, F.; Laversanne, M.; Sung, H.; Ferlay, J.; Siegel, R.L.; Soerjomataram, I.; Jemal, A. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2024, 74, 229–263. [Google Scholar] [CrossRef]
  2. Centers for Disease Control and Prevention. U.S. Cancer Statistics Lung Cancer Stat Bite; U.S. Department of Health and Human Services: Washington, DC, USA, 2025. Available online: https://www.cdc.gov/lung-cancer/statistics/index.html (accessed on 25 July 2025).
  3. Zhang, Y.-L.; Yuan, J.-Q.; Wang, K.-F.; Fu, X.-H.; Han, X.-R.; Threapleton, D.; Yang, Z.-Y.; Mao, C.; Tang, J.-L. The prevalence of EGFR mutation in patients with non-small cell lung cancer: A systematic review and meta-analysis. Oncotarget 2016, 7, 78985–78993. [Google Scholar] [CrossRef]
  4. Merker, J.D.; Oxnard, G.R.; Compton, C.; Diehn, M.; Hurley, P.; Lazar, A.J.; Lindeman, N.; Lockwood, C.M.; Rai, A.J.; Schilsky, R.L.; 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]
  5. Hiatt, J.B.; Doebley, A.-L.; Arnold, H.U.; Adil, M.; Sandborg, H.; Persse, T.W.; Ko, M.; Wu, F.; Villalonga, A.Q.; Santana-Davila, R.; et al. Molecular phenotyping of small cell lung cancer using targeted cfDNA profiling of transcriptional regulatory regions. Sci. Adv. 2024, 10, eadk2082. [Google Scholar] [CrossRef] [PubMed]
  6. Duffy, M.J. Circulating tumor DNA (ctDNA) as a biomarker for lung cancer: Early detection, monitoring and therapy prediction. Tumor Biol. 2023, 46, S283–S295. [Google Scholar] [CrossRef]
  7. Stroun, M.; Anker, P.; Maurice, P.; Lyautey, J.; Lederrey, C.; Beljanski, M. Neoplastic Characteristics of the DNA Found in the Plasma of Cancer Patients. Oncology 1989, 46, 318–322. [Google Scholar] [CrossRef]
  8. Wan, J.C.M.; Massie, C.; Garcia-Corbacho, J.; Mouliere, F.; Brenton, J.D.; Caldas, C.; Pacey, S.; Baird, R.; Rosenfeld, N. Liquid biopsies come of age: Towards implementation of circulating tumour DNA. Nat. Rev. Cancer 2017, 17, 223–238. [Google Scholar] [CrossRef]
  9. Gray, J.E.; Markovets, A.; Reungwetwattana, T.; Majem, M.; Nogami, N.; Peled, N.; Lee, J.-S.; Cho, B.C.; Chewaskulyong, B.; John, T.; et al. Longitudinal Analyses of Circulating Tumor DNA for the Detection of EGFR Mutation-Positive Advanced NSCLC Progression During Treatment: Data from FLAURA and AURA3. J. Thorac. Oncol. 2024, 19, 1525–1538. [Google Scholar] [CrossRef]
  10. Rolfo, C.; Mack, P.; Scagliotti, G.V.; Aggarwal, C.; Arcila, M.E.; Barlesi, F.; Bivona, T.; Diehn, M.; Dive, C.; Dziadziuszko, R.; et al. Liquid Biopsy for Advanced NSCLC: A Consensus Statement From the International Association for the Study of Lung Cancer. J. Thorac. Oncol. 2021, 16, 1647–1662. [Google Scholar] [CrossRef]
  11. Chaudhuri, A.A.; Chabon, J.J.; Lovejoy, A.F.; Newman, A.M.; Stehr, H.; Azad, T.D.; Khodadoust, M.S.; Esfahani, M.S.; Liu, C.L.; Zhou, L.; 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] [PubMed]
  12. Pellini, B.; Chaudhuri, A.A. Circulating Tumor DNA Minimal Residual Disease Detection of Non–Small-Cell Lung Cancer Treated with Curative Intent. J. Clin. Oncol. 2022, 40, 567–575. [Google Scholar] [CrossRef]
  13. Abbosh, C.; Birkbak, N.J.; Wilson, G.A.; Jamal-Hanjani, M.; Constantin, T.; Salari, R.; Le Quesne, J.; Moore, D.A.; Veeriah, S.; Rosenthal, R.; et al. Phylogenetic ctDNA analysis depicts early-stage lung cancer evolution. Nature 2017, 545, 446–451. [Google Scholar] [CrossRef]
  14. Bestvina, C.M.; Garassino, M.C.; Neal, J.W.; Wakelee, H.A.; Diehn, M.; Vokes, E.E. Early-Stage Lung Cancer: Using Circulating Tumor DNA to Get Personal. J. Clin. Oncol. 2023, 41, 4093–4096. [Google Scholar] [CrossRef]
  15. Diaz, L.A., Jr.; Bardelli, A. Liquid Biopsies: Genotyping Circulating Tumor DNA. J. Clin. Oncol. 2014, 32, 579–586. [Google Scholar] [CrossRef] [PubMed]
  16. Singh, A.P.; Li, S.; Cheng, H. Circulating DNA in EGFR-mutated lung cancer. Ann. Transl. Med. 2017, 5, 379. [Google Scholar] [CrossRef]
  17. Singh, A.P.; Cheng, H.; Guo, X.; Levy, B.; Halmos, B. Circulating Tumor DNA in Non–Small-Cell Lung Cancer: A Primer for the Clinician. JCO Precis. Oncol. 2017, 1, 1–13. [Google Scholar] [CrossRef] [PubMed]
  18. Verzè, M.; Pluchino, M.; Leonetti, A.; Corianò, M.; Bonatti, F.; Armillotta, M.P.; Perrone, F.; Casali, M.; Minari, R.; Tiseo, M. Role of ctDNA for the detection of minimal residual disease in resected non-small cell lung cancer: A systematic review. Transl. Lung Cancer Res. 2022, 11, 2588–2600. [Google Scholar] [CrossRef] [PubMed]
  19. Marinello, A.; Tagliamento, M.; Pagliaro, A.; Conci, N.; Cella, E.; Vasseur, D.; Remon, J.; Levy, A.; Dall’oLio, F.G.; Besse, B. Circulating tumor DNA to guide diagnosis and treatment of localized and locally advanced non-small cell lung cancer. Cancer Treat. Rev. 2024, 129, 102791. [Google Scholar] [CrossRef]
  20. Mathios, D.; Johansen, J.S.; Cristiano, S.; Medina, J.E.; Phallen, J.; Larsen, K.R.; Bruhm, D.C.; Niknafs, N.; Ferreira, L.; Adleff, V.; et al. Detection and characterization of lung cancer using cell-free DNA fragmentomes. Nat. Commun. 2021, 12, 5060. [Google Scholar] [CrossRef]
  21. Mazzone, P.J.; Bach, P.B.; Carey, J.; Schonewolf, C.A.; Bognar, K.; Ahluwalia, M.S.; Cruz-Correa, M.; Gierada, D.; Kotagiri, S.; Lloyd, K.; et al. Clinical Validation of a Cell-Free DNA Fragmentome Assay for Augmentation of Lung Cancer Early Detection. Cancer Discov. 2024, 14, 2224–2242. [Google Scholar] [CrossRef]
  22. Cristiano, S.; Leal, A.; Phallen, J.; Fiksel, J.; Adleff, V.; Bruhm, D.C.; Jensen, S.Ø.; Medina, J.E.; Hruban, C.; White, J.R.; et al. Genome-wide cell-free DNA fragmentation in patients with cancer. Nature 2019, 570, 385–389. [Google Scholar] [CrossRef]
  23. Li, Y.; Fan, Z.; Meng, Y.; Liu, S.; Zhan, H. Blood-based DNA methylation signatures in cancer: A systematic review. Biochim. Biophys. Acta (BBA)-Mol. Basis Dis. 2022, 1869, 166583. [Google Scholar] [CrossRef] [PubMed]
  24. Barlesi, F.; Gervais, R.; Lena, H.; Hureaux, J.; Berard, H.; Paillotin, D.; Bota, S.; Monnet, I.; Chajara, A.; Robinet, G. Pemetrexed and cisplatin as first-line chemotherapy for advanced non-small-cell lung cancer (NSCLC) with asymptomatic inoperable brain metastases: A multicenter phase II trial (GFPC 07-01). Ann. Oncol. 2011, 22, 2466–2470. [Google Scholar] [CrossRef] [PubMed]
  25. De Mattos-Arruda, L.; Mayor, R.; Ng, C.K.Y.; Weigelt, B.; Martínez-Ricarte, F.; Torrejon, D.; Oliveira, M.; Arias, A.; Raventos, C.; Tang, J.; et al. Cerebrospinal fluid-derived circulating tumour DNA better represents the genomic alterations of brain tumours than plasma. Nat. Commun. 2015, 6, 8839. [Google Scholar] [CrossRef]
  26. Sasaki, S.; Yoshioka, Y.; Ko, R.; Katsura, Y.; Namba, Y.; Shukuya, T.; Kido, K.; Iwakami, S.; Tominaga, S.; Takahashi, K. Diagnostic significance of cerebrospinal fluid EGFR mutation analysis for leptomeningeal metastasis in non-small-cell lung cancer patients harboring an active EGFR mutation following gefitinib therapy failure. Respir. Investig. 2015, 54, 14–19. [Google Scholar] [CrossRef] [PubMed]
  27. Hata, A.; Nanjo, S.; Okuda, C.; Kaji, R.; Masago, K.; Fujita, S.; Irie, K.; Okada, H.; Fukushima, S.; Katakami, N. 451PD Osimertinib at 80 mg for refractory leptomeningeal metastases in T790M-positive EGFR-mutant non-small cell lung cancer. Ann. Oncol. 2016, 27, mdw594.015. [Google Scholar] [CrossRef]
  28. Reckamp, K.L.; Melnikova, V.O.; Karlovich, C.; Sequist, L.V.; Camidge, D.R.; Wakelee, H.; Perol, M.; Oxnard, G.R.; Kosco, K.; Croucher, P.; et al. A Highly Sensitive and Quantitative Test Platform for Detection of NSCLC EGFR Mutations in Urine and Plasma. J. Thorac. Oncol. 2016, 11, 1690–1700. [Google Scholar] [CrossRef]
  29. Zhao, J.; Ye, X.; Xu, Y.; Chen, M.; Zhong, W.; Sun, Y.; Yang, Z.; Zhu, G.; Gu, Y.; Wang, M. EGFR mutation status of paired cerebrospinal fluid and plasma samples in EGFR mutant non-small cell lung cancer with leptomeningeal metastases. Cancer Chemother. Pharmacol. 2016, 78, 1305–1310. [Google Scholar] [CrossRef]
  30. Wei, F.; Lin, C.-C.; Joon, A.; Feng, Z.; Troche, G.; Lira, M.E.; Chia, D.; Mao, M.; Ho, C.-L.; Su, W.-C.; et al. Noninvasive Saliva-based EGFR Gene Mutation Detection in Patients with Lung Cancer. Am. J. Respir. Crit. Care Med. 2014, 190, 1117–1126. [Google Scholar] [CrossRef]
  31. Park, S.; Hur, J.Y.; Lee, K.Y.; Lee, J.C.; Rho, J.K.; Shin, S.H.; Choi, C.-M. Assessment of EGFR mutation status using cell-free DNA from bronchoalveolar lavage fluid. CCLM 2017, 55, 1489–1495. [Google Scholar] [CrossRef]
  32. García-Pardo, M.; Czarnecka-Kujawa, K.; Law, J.H.; Salvarrey, A.M.; Fernandes, R.; Fan, Z.J.; Waddell, T.K.; Yasufuku, K.; Liu, G.; Donahoe, L.L.; et al. Association of Circulating Tumor DNA Testing Before Tissue Diagnosis with Time to Treatment Among Patients with Suspected Advanced Lung Cancer. JAMA Netw. Open 2023, 6, e2325332. [Google Scholar] [CrossRef] [PubMed]
  33. Moding, E.J.; Nabet, B.Y.; Alizadeh, A.A.; Diehn, M. Detecting Liquid Remnants of Solid Tumors: Circulating Tumor DNA Minimal Residual Disease. Cancer Discov. 2021, 11, 2968–2986. [Google Scholar] [CrossRef]
  34. Bartolomucci, A.; Nobrega, M.; Ferrier, T.; Dickinson, K.; Kaorey, N.; Nadeau, A.; Castillo, A.; Burnier, J.V. Circulating tumor DNA to monitor treatment response in solid tumors and advance precision oncology. npj Precis. Oncol. 2025, 9, 84. [Google Scholar] [CrossRef]
  35. Newman, A.M.; Bratman, S.V.; To, J.; Wynne, J.F.; Eclov, N.C.W.; Modlin, L.A.; Liu, C.L.; Neal, J.W.; Wakelee, H.A.; Merritt, R.E.; et al. An ultrasensitive method for quantitating circulating tumor DNA with broad patient coverage. Nat. Med. 2014, 20, 548–554. [Google Scholar] [CrossRef]
  36. Xue, R.; Li, X.; Yang, L.; Yang, M.; Zhang, B.; Zhang, X.; Li, L.; Duan, X.; Yan, R.; He, X.; et al. Evaluation and integration of cell-free DNA signatures for detection of lung cancer. Cancer Lett. 2024, 604, 217216. [Google Scholar] [CrossRef] [PubMed]
  37. Thompson, J.C.; Aggarwal, C.; Wong, J.; Nimgaonkar, V.; Hwang, W.-T.; Andronov, M.; Dibardino, D.M.; Hutchinson, C.T.; Ma, K.C.; Lanfranco, A.; et al. Plasma Genotyping at the Time of Diagnostic Tissue Biopsy Decreases Time-to-Treatment in Patients with Advanced NSCLC—Results From a Prospective Pilot Study. JTO Clin. Res. Rep. 2022, 3, 100301. [Google Scholar] [CrossRef]
  38. Raez, L.E.; Brice, K.; Dumais, K.; Lopez-Cohen, A.; Wietecha, D.; Izquierdo, P.A.; Santos, E.S.; Powery, H.W. Liquid Biopsy Versus Tissue Biopsy to Determine Front Line Therapy in Metastatic Non-Small Cell Lung Cancer (NSCLC). Clin. Lung Cancer 2022, 24, 120–129. [Google Scholar] [CrossRef]
  39. Chen, K.; Zhao, H.; Shi, Y.; Yang, F.; Wang, L.T.; Kang, G.; Nie, Y.; Wang, J. Perioperative Dynamic Changes in Circulating Tumor DNA in Patients with Lung Cancer (DYNAMIC). Clin. Cancer Res. 2019, 25, 7058–7067. [Google Scholar] [CrossRef] [PubMed]
  40. Bouchard, N.; Daaboul, N. Lung Cancer: Targeted Therapy in 2025. Curr. Oncol. 2025, 32, 146. [Google Scholar] [CrossRef]
  41. Li, R.-Y.; Liang, Z.-Y. Circulating tumor DNA in lung cancer: Real-time monitoring of disease evolution and treatment response. Chin. Med. J. 2020, 133, 2476–2485. [Google Scholar] [CrossRef]
  42. Zviran, A.; Schulman, R.C.; Shah, M.; Hill, S.T.K.; Deochand, S.; Khamnei, C.C.; Maloney, D.; Patel, K.; Liao, W.; Widman, A.J.; et al. Genome-wide cell-free DNA mutational integration enables ultra-sensitive cancer monitoring. Nat. Med. 2020, 26, 1114–1124. [Google Scholar] [CrossRef]
  43. Kato, R.; Hayashi, H.; Sakai, K.; Suzuki, S.; Haratani, K.; Takahama, T.; Tanizaki, J.; Nonagase, Y.; Tanaka, K.; Yoshida, T.; et al. CAPP-seq analysis of circulating tumor DNA from patients with EGFR T790M–positive lung cancer after osimertinib. Int. J. Clin. Oncol. 2021, 26, 1628–1639. [Google Scholar] [CrossRef]
  44. Peng, M.; Huang, Q.; Yin, W.; Tan, S.; Chen, C.; Liu, W.; Tang, J.; Wang, X.; Zhang, B.; Zou, M.; et al. Circulating Tumor DNA as a Prognostic Biomarker in Localized Non-small Cell Lung Cancer. Front. Oncol. 2020, 10, 561598. [Google Scholar] [CrossRef]
  45. Kan, C.F.K.; Unis, G.D.; Li, L.Z.; Gunn, S.; Li, L.; Soyer, H.P.; Stark, M.S. Circulating Biomarkers for Early Stage Non-Small Cell Lung Carcinoma Detection: Supplementation to Low-Dose Computed Tomography. Front. Oncol. 2021, 11, 555331. [Google Scholar] [CrossRef]
  46. Kuang, P.P.; Li, N.; Liu, Z.; Sun, T.Y.; Wang, S.Q.; Hu, J.; Ou, W.; Wang, S.Y. Circulating Tumor DNA Analyses as a Potential Marker of Recurrence and Effectiveness of Adjuvant Chemotherapy for Resected Non-Small-Cell Lung Cancer. Front. Oncol. 2021, 10, 595650. [Google Scholar] [CrossRef]
  47. Provencio, M.; Serna-Blasco, R.; Nadal, E.; Insa, A.; García-Campelo, M.R.; Casal Rubio, J.; Dómine, M.; Majem, M.; Rodríguez-Abreu, D.; Martínez-Martí, A.; et al. Overall survival and biomarker analysis of neoadjuvant nivolumab plus chemotherapy in operable stage IIIA non-small-cell lung cancer (NADIM phase II trial). J. Clin. Oncol. 2022, 40, 2924–2933. [Google Scholar] [CrossRef]
  48. Reck, M.; Gale, D.; Harpole, D.; Taube, J.; Mitsudomi, T.; Hochmair, M.; Winder, T.; Zhu, Z.; Lai, Z.; Stewart, R.; et al. LBA49 Associations of ctDNA clearance (CL) during neoadjuvant Tx with pathological response and event-free survival (EFS) in pts with resectable NSCLC (R-NSCLC): Expanded analyses from AEGEAN. Ann. Oncol. 2024, 35, S1239. [Google Scholar] [CrossRef]
  49. Deutsch, J.S.; Cimino-Mathews, A.; Thompson, E.; Provencio, M.; Forde, P.M.; Spicer, J.; Girard, N.; Wang, D.; Anders, R.A.; Gabrielson, E.; et al. Association between pathologic response and survival after neoadjuvant therapy in lung cancer. Nat. Med. 2024, 30, 218–228. [Google Scholar] [CrossRef]
  50. Forde, P.M.; Spicer, J.; Lu, S.; Provencio, M.; Mitsudomi, T.; Awad, M.M.; Felip, E.; Broderick, S.R.; Brahmer, J.R.; Swanson, S.J.; et al. CheckMate 816 Investigators. Neoadjuvant nivolumab plus chemotherapy in resectable lung cancer. N. Engl. J. Med. 2022, 386, 1973–1985. [Google Scholar] [CrossRef]
  51. Herbst, R.S.; Wang, M.; Chen, L. When immunotherapy meets surgery in non-small cell lung cancer. Cancer Cell 2022, 40, 603–605. [Google Scholar] [CrossRef]
  52. Zhou, C.; Das Thakur, M.; Srivastava, M.; Zou, W.; Xu, H.; Ballinger, M.; Felip, E.; Wakelee, H.; Altorki, N.; Reck, M.; et al. 2O IMpower010: Biomarkers of disease-free survival (DFS) in a phase III study of atezolizumab (atezo) vs. best supportive care (BSC) after adjuvant chemotherapy in stage IB-IIIA NSCLC. Ann. Oncol. 2021, 32, S134. [Google Scholar] [CrossRef]
  53. Abbosh, C.; Frankell, A.M.; Harrison, T.; Kisistok, J.; Garnett, A.; Johnson, L.; Veeriah, S.; Moreau, M.; Chesh, A.; Chaunzwa, T.L.; et al. Tracking early lung cancer metastatic dissemination in TRACERx using ctDNA. Nature 2023, 616, 553–562. [Google Scholar] [CrossRef]
  54. Jung, H.-A.; Ku, B.M.; Kim, Y.J.; Park, S.; Sun, J.-M.; Lee, S.-H.; Ahn, J.S.; Cho, J.H.; Kim, H.K.; Choi, Y.S.; et al. Longitudinal Monitoring of Circulating Tumor DNA From Plasma in Patients with Curative Resected Stages I to IIIA EGFR-Mutant Non–Small Cell Lung Cancer. J. Thorac. Oncol. 2023, 18, 1199–1208. [Google Scholar] [CrossRef]
  55. Guo, N.; Lou, F.; Ma, Y.; Li, J.; Yang, B.; Chen, W.; Ye, H.; Zhang, J.-B.; Zhao, M.-Y.; Wu, W.-J.; et al. Circulating tumor DNA detection in lung cancer patients before and after surgery. Sci. Rep. 2016, 6, 33519. [Google Scholar] [CrossRef]
  56. Waldeck, S.; Mitschke, J.; Wiesemann, S.; Rassner, M.; Andrieux, G.; Deuter, M.; Mutter, J.; Lüchtenborg, A.M.; Kottmann, D.; Titze, L.; et al. Early assessment of circulating tumor DNA after curative-intent resection predicts tumor recurrence in early-stage and locally advanced non-small-cell lung cancer. Mol. Oncol. 2022, 16, 527–537. [Google Scholar] [CrossRef]
  57. Gale, D.; Heider, K.; Ruiz-Valdepenas, A.; Hackinger, S.; Perry, M.; Marsico, G.; Rundell, V.; Wulff, J.; Sharma, G.; Knock, H.; et al. Residual ctDNA after treatment predicts early relapse in patients with early-stage non-small cell lung cancer. Ann. Oncol. 2022, 33, 500–510. [Google Scholar] [CrossRef]
  58. Oh, Y.; Yoon, S.M.; Lee, J.; Park, J.H.; Lee, S.; Hong, T.; Chung, L.I.; Sudhaman, S.; Riddell, T.; Palsuledesai, C.C.; et al. Personalized, tumor-informed, circulating tumor DNA assay for detecting minimal residual disease in non-small cell lung cancer patients receiving curative treatments. Thorac. Cancer 2024, 15, 1095–1102. [Google Scholar] [CrossRef]
  59. Ohara, S.; Suda, K.; Sakai, K.; Nishino, M.; Chiba, M.; Shimoji, M.; Takemoto, T.; Fujino, T.; Koga, T.; Hamada, A.; et al. Prognostic implications of preoperative versus postoperative circulating tumor DNA in surgically resected lung cancer patients: A pilot study. Transl. Lung Cancer Res. 2020, 9, 1915–1923. [Google Scholar] [CrossRef]
  60. Tan, A.C.; Lai, G.G.Y.; Saw, S.P.L.; Chua, K.L.M.; Takano, A.; Ong, B.H.; Koh, T.P.T.; Jain, A.; Tan, W.L.; Ng, Q.S.; et al. Detection of circulating tumor DNA with ultradeep sequencing of plasma cell-free DNA for monitoring minimal residual disease and early detection of recurrence in early-stage lung cancer. Cancer 2024, 130, 1758–1765. [Google Scholar] [CrossRef]
  61. Rosenlund, L.; Guldbrandsen, K.; Ahlborn, L.B.; Bloch, M.; Skougaard, K.; Albrecht-Beste, E.; Nellemann, H.M.; Krakauer, M.; Gørtz, P.M.; Fledelius, J.; et al. ctDNA can detect minimal residual disease in curative treated non-small cell lung cancer patients using a tumor agnostic approach. Lung Cancer 2025, 203, 108528. [Google Scholar] [CrossRef]
  62. Schuurbiers, M.M.F.; Smith, C.G.; Hartemink, K.J.; Rintoul, R.C.; Gale, D.; Monkhorst, K.; Mandos, B.L.R.; Paterson, A.L.; Broek, D.v.D.; Rosenfeld, N.; et al. Recurrence prediction using circulating tumor DNA in patients with early-stage non-small cell lung cancer after treatment with curative intent: A retrospective validation study. PLoS Med. 2025, 22, e1004574. [Google Scholar] [CrossRef]
  63. Tan, A.; Lai, G.; Saw, S.; Chua, K.; Takano, A.; Ong, B.; Koh, T.; Jain, A.; Tan, W.; Ng, Q.; et al. MA07.06 Circulating Tumor DNA for Monitoring Minimal Residual Disease and Early Detection of Recurrence in Early Stage Lung Cancer. J. Thorac. Oncol. 2021, 16, S907. [Google Scholar] [CrossRef]
  64. Zhang, J.-T.; Dong, S.; Gu, W.Q.; Zhao, N.; Liang, Y.; Tang, W.F.; Liu, S.Y.; Wang, F.; Wang, G.S.; Peng, B.; et al. Adjuvant Therapy-Free Strategy for Stage IB to IIIA Non–Small-Cell Lung Cancer Patients After Radical Resection Based on Longitudinal Undetectable Molecular Residual Disease: Prospective, Multicenter, Single-Arm Study (CTONG 2201). Clin. Lung Cancer. 2024, 25, e1–e4. [Google Scholar] [CrossRef]
  65. Zhao, X.; Dai, F.; Mei, L.; Huang, D.; Shen, X.; Zhang, H.; She, X.; Ma, Z. The Potential Use of Dynamics Changes of ctDNA and cfDNA in the Perioperative Period to Predict the Recurrence Risk in Early NSCLC. Front. Oncol. 2021, 11, 671963. [Google Scholar] [CrossRef]
  66. Li, N.; Wang, B.X.; Li, J.; Shao, Y.; Li, M.T.; Li, J.J.; Kuang, P.P.; Liu, Z.; Sun, T.Y.; Wu, H.Q.; et al. Perioperative circulating tumor DNA as a potential prognostic marker for operable stage I to IIIA non-small cell lung cancer. Cancer 2022, 128, 708–718. [Google Scholar] [CrossRef]
  67. Xia, L.; Mei, J.; Kang, R.; Deng, S.; Chen, Y.; Yang, Y.; Feng, G.; Deng, Y.; Gan, F.; Lin, Y.; et al. Perioperative ctDNA-Based Molecular Residual Disease Detection for Non-Small Cell Lung Cancer: A Prospective Multicenter Cohort Study (LUNGCA-1). Clin. Cancer Res. 2022, 28, 3308–3317. [Google Scholar] [CrossRef]
  68. Qiu, B.; Guo, W.; Zhang, F.; Lv, F.; Ji, Y.; Peng, Y.; Chen, X.; Bao, H.; Xu, Y.; Shao, Y.; et al. Dynamic recurrence risk and adjuvant chemotherapy benefit prediction by ctDNA in resected NSCLC. Nat. Commun. 2021, 12, 6770. [Google Scholar] [CrossRef]
  69. Moding, E.J.; Liu, Y.; Nabet, B.Y.; Chabon, J.J.; Chaudhuri, A.A.; Hui, A.B.; Bonilla, R.F.; Ko, R.B.; Yoo, C.H.; Gojenola, L.; et al. Circulating tumor DNA dynamics predict benefit from consolidation immunotherapy in locally advanced non-small-cell lung cancer. Nat. Cancer 2020, 1, 176–183. [Google Scholar] [CrossRef]
  70. Desai, A.; Vázquez, T.A.; Arce, K.M.; Corassa, M.; Mack, P.C.; Gray, J.E.; Pellini, B. ctDNA for the Evaluation and Management of EGFR-Mutant Non-Small Cell Lung Cancer. Cancers 2024, 16, 940. [Google Scholar] [CrossRef]
  71. O’Kane, G.M.; Bradbury, P.A.; Feld, R.; Leighl, N.B.; Liu, G.; Pisters, K.-M.; Kamel-Reid, S.; Tsao, M.S.; Shepherd, F.A. Uncommon EGFR mutations in advanced non-small cell lung cancer. Lung Cancer 2017, 109, 137–144. [Google Scholar] [CrossRef]
  72. Arcila, M.E.; Nafa, K.; Chaft, J.E.; Rekhtman, N.; Lau, C.; Reva, B.A.; Zakowski, M.F.; Kris, M.G.; Ladanyi, M. EGFR Exon 20 Insertion Mutations in Lung Adenocarcinomas: Prevalence, Molecular Heterogeneity, and Clinicopathologic Characteristics. Mol. Cancer Ther. 2013, 12, 220–229. [Google Scholar] [CrossRef]
  73. Felip, E.; Cho, B.C.; Gutiérrez, V.; Alip, A.; Besse, B.; Lu, S.; Spira, A.I.; Girard, N.; Califano, R.; Gadgeel, A.M.; et al. Amivantamab plus lazertinib versus osimertinib in first-line EGFR-mutant advanced non-small-cell lung cancer with biomarkers of high-risk disease: A secondary analysis from MARIPOSA. Ann. Oncol. 2024, 35, 805–816. [Google Scholar] [CrossRef]
  74. Liu, S.-Y.; Erazo, T.; Jee, J.; Arfe, A.; Gupta, A.; Pike, L.R.; Santini, F.C.; Daly, B.; Schoenfeld, A.; Eichholz, J.; et al. Optimal systemic treatment and real-world clinical application of ctDNA in patients with metastatic HER2-mutant lung cancer. Eur. J. Cancer 2024, 210, 114257. [Google Scholar] [CrossRef]
  75. Anagnostou, V.; Ho, C.; Nicholas, G.; Juergens, R.A.; Sacher, A.; Fung, A.S.; Wheatley-Price, P.; Laurie, S.A.; Levy, B.; Brahmer, J.R.; et al. ctDNA response after pembrolizumab in non-small cell lung cancer: Phase 2 adaptive trial results. Nat. Med. 2023, 29, 2559–2569. [Google Scholar] [CrossRef]
  76. Lovly, C.M.; Salama, A.K.; Salgia, R. Tumor heterogeneity and therapeutic resistance. Am. Soc. Clin. Oncol. Educ. Book 2016, 36, e585–e593. [Google Scholar] [CrossRef]
  77. Song, Y.; Hu, C.; Xie, Z.; Wu, L.; Zhu, Z.; Rao, C.; Liu, L.; Chen, Y.; Liang, N.; Chen, J.; et al. Written on behalf of AME Lung Cancer Collaborative Group. Circulating tumor DNA clearance predicts prognosis across treatment regimen in a large real-world longitudinally monitored advanced non-small cell lung cancer cohort. Transl. Lung Cancer Res. 2020, 9, 269–279. [Google Scholar] [CrossRef] [PubMed]
  78. Paik, P.K.; Felip, E.; Veillon, R.; Sakai, H.; Cortot, A.B.; Garassino, M.C.; Mazieres, J.; Viteri, S.; Senellart, H.; Van Meerbeeck, J.; et al. Tepotinib in Non–Small-Cell Lung Cancer with MET Exon 14 Skipping Mutations. N. Engl. J. Med. 2020, 383, 931–943. [Google Scholar] [CrossRef]
  79. Jun, S.; Shukla, N.A.; Durm, G.; Hui, A.B.; Cao, S.; Ganti, A.K.; Jabbour, S.K.; Kunder, C.; Alizadeh, A.A.; Hanna, N.H.; et al. Analysis of Circulating Tumor DNA Predicts Outcomes of Short-Course Consolidation Immunotherapy in Unresectable Stage III NSCLC. J. Thorac. Oncol. 2024, 19, 1427–1437. [Google Scholar] [CrossRef] [PubMed]
  80. Pan, Y.; Zhang, J.-T.; Gao, X.; Chen, Z.-Y.; Yan, B.; Tan, P.-X.; Yang, X.-R.; Gao, W.; Gong, Y.; Tian, Z.; et al. Dynamic circulating tumor DNA during chemoradiotherapy predicts clinical outcomes for locally advanced non-small cell lung cancer patients. Cancer Cell 2023, 41, 1763–1773.e4. [Google Scholar] [CrossRef]
  81. Yang, Y.; Zhang, T.; Wang, J.; Wang, J.; Xu, Y.; Zhao, X.; Ou, Q.; Shao, Y.; Wang, X.; Wu, Y.; et al. The clinical utility of dynamic ctDNA monitoring in inoperable localized NSCLC patients. Mol. Cancer 2022, 21, 117. [Google Scholar] [CrossRef] [PubMed]
  82. Horndalsveen, H.; Haakensen, V.D.; Madebo, T.; Gronberg, B.H.; Halvorsen, T.O.; Koivunen, J.; Oselin, K.; Cicenas, S.; Helbekkmo, N.; Aanerud, M.; et al. ctDNA-based MRD detection in unresectable NSCLC undergoing curatively intended chemoradiotherapy and durvalumab. J. Clin. Oncol. 2025, 43, 8011. [Google Scholar] [CrossRef]
  83. Ernst, S.M.; van Marion, R.; Atmodimedjo, P.N.; de Jonge, E.; Mathijssen, R.H.J.; Paats, M.S.; de Bruijn, P.; Koolen, S.L.; von der Thüsen, J.H.; Aerts, J.G.J.V.; et al. Clinical utility of circulating tumor DNA in patients with advanced KRASG12C-mutated NSCLC treated with sotorasib. J. Thorac. Oncol. 2024, 19, 995–1006. [Google Scholar] [CrossRef]
  84. Yamaguchi, O.; Kasahara, N.; Soda, H.; Imai, H.; Naruse, I.; Yamaguchi, H.; Itai, M.; Taguchi, K.; Uchida, M.; Sunaga, N.; et al. Predictive significance of circulating tumor DNA against patients with T790M-positive EGFR-mutant NSCLC receiving osimertinib. Sci. Rep. 2023, 13, 20848. [Google Scholar] [CrossRef]
  85. Shaw, A.T.; Solomon, B.J.; Besse, B.; Bauer, T.M.; Lin, C.C.; Soo, R.A.; Riely, G.J.; Ou, S.H.I.; Clancy, J.S.; Li, S.; et al. ALK Resistance Mutations and Efficacy of Lorlatinib in Advanced Anaplastic Lymphoma Kinase-Positive Non-Small-Cell Lung Cancer. J. Clin. Oncol. 2019, 37, 1370–1379. [Google Scholar] [CrossRef]
  86. Vendrell, J.A.; Quantin, X.; Aussel, A.; Solassol, I.; Serre, I.; Solassol, J. EGFR-dependent mechanisms of resistance to osimertinib determined by ctDNA NGS analysis identify patients with better outcome. Transl. Lung Cancer Res. 2021, 10, 4084–4094. [Google Scholar] [CrossRef]
  87. Remon, J.; Besse, B.; Aix, S.P.; Callejo, A.; Al-Rabi, K.; Bernabe, R.; Greillier, L.; Majem, M.; Reguart, N.; Monnet, I.; et al. Osimertinib treatment based on plasma T790M monitoring in patients with EGFR-mutant non-small-cell lung cancer (NSCLC): EORTC Lung Cancer Group 1613 APPLE phase II randomized clinical trial. Ann. Oncol. 2023, 34, 468–476. [Google Scholar] [CrossRef]
  88. Murtaza, M.; Dawson, S.-J.; Tsui, D.W.Y.; Gale, D.; Forshew, T.; Piskorz, A.M.; Parkinson, C.; Chin, S.-F.; Kingsbury, Z.; Wong, A.S.C.; et al. Non-Invasive Analysis of Acquired Resistance to Cancer Therapy by Sequencing of Plasma DNA. Nature 2013, 497, 108–112. [Google Scholar] [CrossRef]
  89. Gridelli, C.; Nuccio, E.; Casaluce, F. The Role of Osimertinib in Stage I–II Non-Small-Cell Lung Cancer with Activating EGFR Mutation. Targets 2025, 3, 20. [Google Scholar] [CrossRef]
  90. Zhang, E.W.; Dagogo-Jack, I.; Kuo, A.; Rooney, M.M.; Shaw, A.T.; Digumarthy, S.R. Association between circulating tumor DNA burden and disease burden in patients with ALK-positive lung cancer. Cancer 2020, 126, 4473–4484. [Google Scholar] [CrossRef]
  91. Dziadziuszko, R.; Mok, T.; Peters, S.; Han, J.Y.; Alatorre-Alexander, J.; Leighl, N.; Sriuranpong, V.; Pérol, M.; de Castro Junior, G.; Nadal, E.; et al. Blood First Assay Screening Trial (BFAST) in treatment-naive advanced or metastatic NSCLC: Initial Results of the Phase 2 ALK-Positive Cohort. J. Thorac. Oncol. 2021, 16, 2040–2050. [Google Scholar] [CrossRef]
  92. Dagogo-Jack, I.; Brannon, A.R.; Ferris, L.A.; Campbell, C.D.; Lin, J.J.; Schultz, K.R.; Ackil, J.; Stevens, S.; Dardaei, L.; Yoda, S.; et al. Tracking the evolution of resistance to ALK Tyrosine Kinase inhibitors through longitudinal analysis of circulating tumor DNA. JCO Precis. Oncol. 2018, 2, 1–14. [Google Scholar] [CrossRef]
  93. Heeke, S.; Gandhi, S.; Tran, H.T.; Lam, V.K.; Byers, L.A.; Gibbons, D.L.; Gay, C.M.; Altan, M.; Antonoff, M.B.; Le, X.; et al. Longitudinal Tracking of ALK-Rearranged NSCLC From Plasma Using Circulating Tumor RNA and Circulating Tumor DNA. JTO Clin. Res. Rep. 2025, 6, 100795. [Google Scholar] [CrossRef]
  94. Chmielecki, J.; Gray, J.E.; Cheng, Y.; Ohe, Y.; Imamura, F.; Cho, B.C.; Lin, M.-C.; Majem, M.; Shah, R.; Rukazenkov, Y. Can-didate mechanisms of acquired resistance to first-line osimertinib in EGFR-mutated advanced non-small cell lung cancer. Nat. Commun. 2023, 14, 1070. [Google Scholar] [CrossRef]
  95. Sivapalan, L.; Murray, J.C.; Canzoniero, J.V.; Landon, B.; Jackson, J.; Scott, S.; Lam, V.; Levy, B.P.; Sausen, M.; Anagnostou, V. Liquid biopsy approaches to capture tumor evolution and clinical outcomes during cancer immunotherapy. J. Immunother. Cancer 2023, 11, e005924. [Google Scholar] [CrossRef]
  96. Planchard, D.; Loriot, Y.; Andre, F.; Gobert, A.; Auger, N.; Lacroix, L.; Soria, J. EGFR-independent mechanisms of acquired resistance to AZD9291 in EGFR T790M-positive NSCLC patients. Ann. Oncol. 2015, 26, 2073–2078. [Google Scholar] [CrossRef]
  97. Leonetti, A.; Sharma, S.; Minari, R.; Perego, P.; Giovannetti, E.; Tiseo, M. Resistance mechanisms to osimertinib in EGFR-mutated non-small cell lung cancer. Br. J. Cancer 2019, 121, 725–737. [Google Scholar] [CrossRef]
Figure 1. The prevalence of various actionable genomic alterations in adenocarcinoma. Of note, the prevalence of EGFR exon 19 deletions and EGFR Exon 20 mutation L858R occurs at a higher rate in ~50% of the Asian population, and may vary amongst different populations.
Figure 1. The prevalence of various actionable genomic alterations in adenocarcinoma. Of note, the prevalence of EGFR exon 19 deletions and EGFR Exon 20 mutation L858R occurs at a higher rate in ~50% of the Asian population, and may vary amongst different populations.
Cancers 17 03327 g001
Table 1. Key studies following or utilizing ctDNA in resectable NSCLC.
Table 1. Key studies following or utilizing ctDNA in resectable NSCLC.
StudyStudy Design, Stage, and/or PopulationOutcome
Guo et al., 2016 [55]Prospective; all stages ctDNA from stage IA and IB patients had more dramatic decrease post-op compared with mutation frequency in more advanced cancer pts
Abbosh et al., 2017 [13]Prospective; IIA–IIIB Tumor volume correlated with the mean plasma VAF of clonal SNVs in ctDNA-positive NSCLCs
Chaudhuri et al., 2017 [11]Retrospective; IB–IIIB, Posttreatment ctDNA detection preceded radiographic progression in 72% of patients by a median of 5.2 months
Chen et al., 2019 [39]Prospective; I–IIIA Rapid decrease in ctDNA occurred after radical tumor resection. Median ctDNA half-life was 35 min. RFS in pts with detectable ctDNA was shorter than in those without
Waldeck et al., 2022 [56]Prospective; IA–IIIB Of ctDNA-negative pts, 33% experienced relapse post-op. Positive ctDNA in early post-op plasma samples was associated with shorter PFS and OS
Gale et al., 2022 [57]Prospective; IA–IIIB ctDNA detection had clinical specificity >98.5% and preceded clinical recurrence by a median of 212.5 days
Abbosh et al., 2023 [53]Prospective; II–III Results showed that 3- to 6-monthly ctDNA surveillance identified impending disease relapse in 20% of landmark-negative patients
Jung et al., 2023 [54]Prospective; I-IIIA EGFR mutant Of patients with baseline +ctDNA, 76% had clearance at 4 weeks after surgery. MRD was detected before radiological recurrence in 69% of patients with exon 19 deletion and in 20% with L858R mutation
Provencio et al., 2022 [47]Phase II trial; IIIA or IIIB ctDNA levels were associated with differences in PFS/OS; 67% of the experimental group and 44% of the control group were ctDNA-negative after neoadjuvant treatment.
Oh et al., 2024 [58]Retrospective; I–IV Pts with ctDNA+ were more likely to experience recurrence compared to ctDNA- patients. ctDNA+ was associated with poorer RFS than persistently ctDNA- patients
Ohara et al., 2020 [59]Prospective; II–III ctDNA+ pts had disease recurrence within median of 9.1 months and shorter RFS/OS than those without detectable ctDNA. Three pts who had CNS-only metastases did not have detectable ctDNA
Tan et al., 2024 [60]Retrospective; I–III ctDNA detection preop was associated with shorter RFS. ctDNA positivity preceded radiological findings by a median lead time of 2.8 months
Herbst et al., 2022 [51]Phase III post hoc; IB–IIIA; EGFR mutant MRD+ had clinical sensitivity of 65% and specificity of 95% and preceded a DFS event by a median of 4.7 months
Rosenlund et al., 2025 [61]Prospective; I–III Detectable ctDNA post-treatment was significantly associated with increased risk of tumor recurrence and shorter RFS. Post RT/CRT, ctDNA detection was significantly linked to shorter RFS in MRD analysis
Schuurbiers et al., 2025 [62]Retrospective; I–III Before treatment, ctDNA was detected in 48% LEMA and 51% LUCID pts. ctDNA detection after treatment was associated with shorter recurrence-free survival and overall survival
Table 2. Outlines of key studies on ctDNA and outcomes in advanced and metastatic NSCLC.
Table 2. Outlines of key studies on ctDNA and outcomes in advanced and metastatic NSCLC.
StudyStudy Design, Stage, and/or PopulationOutcome
Liu et al., 2024 [74]Retrospective; Metastatic HER2-mutant HER2 mutations tracked longitudinally correlated with treatment response. Pts with detectable BL ctDNA had shorter OS
Song et al., 2020 [77]Prospective; IIIB to IV Higher ctDNA at BL was associated with shorter OS. ctDNA- was associated with longer PFS/OS regardless of type of treatment
Paik et al., 2020 [78]Phase II; advanced/metastatic MET exon 14 skipping mutationA total of 67% of patients had a molecular cfDNA response; of those, 71% had a radiographic response. ctDNA and imaging PD were highly concordant
Anagnostou et al., 2023 [75]Phase II; advanced/metastatic PD-L1 expression level of ≥1%Median time to ctDNA response was 2.1 months; pts with MR had longer PFS and OS
Jun et al., 2024 [79]Phase II post hoc; III ctDNA+ predicted inferior PFS after completion of CRT and at the end of CPI
Moding et al., 2020 [69]Stage IIB-IIIB NSCLCPts who were ctDNA- after CRT had better outcomes regardless of CICI. Patients with MRD+ after CRT who received CICI had significantly better outcomes than pts who did not receive CICI
Pan et al., 2023 [80]Stage IIB- IIICLongitudinal undetectable MRD was found in 20.1% of patients. The 2-year PFS of these pts was 88.4%
Yang et al., 2022 [81]Stage III NSCLCctDNA collected 1 month post-CRT/RT was the optimal choice to predict pts’ PFS and OS, and the dynamic change in ctDNA was closely associated with clinical outcomes
Horndalsveen et al., 2025 [82] (abstract)Phase II; III Preliminary OS data shows that detectable ctDNA during the first four months post-CRT significantly increased the odds of death within 24 months
Ernst et al., 2024 [83]Prospective; advanced KRASG12C-Mutated Pretreatment, KRASG12C ctDNA was detected in 76% of pts. The disease control rate was significantly higher in those with a molecular response
Yamaguchi et al., 2023 [84]Prospective; III-IVDetection of T790M at PD after osimertinib initiation was a significant independent prognostic factor for predicting shorter prognosis
Shaw et al., 2019 [85]Phase II; ALK mutationPFS was similar in pts with and without ALK mutations on plasma genotyping but longer in patients with ALK mutations on tissue genotyping
Vendrell et al., 2021 [86]Prospective; IV Pts who developed an EGFR-dependent mechanism of resistance responded longer to osimertinib than EGFR-independent resistant pts
Gray et al., 2024 [9]Phase III post hoc; advanced stagectDNA PD preceded or co-occurred with RECIST-defined PD: 64% in FLAURA and 56% in AURA3
Remon et al., 2023 [87]Prospective; EGFR mutantMolecular PD before RECIST PD led to an earlier switch to osimertinib in 17% of patients with satisfactory PFS/OS outcomes
Table 3. Active clinical trials using ctDNA in lung cancer, divided into resectable, non-resectable, and screening/stratification categories. All are active unless otherwise indicated by an asterisk.
Table 3. Active clinical trials using ctDNA in lung cancer, divided into resectable, non-resectable, and screening/stratification categories. All are active unless otherwise indicated by an asterisk.
Trial/Study (with NCT if Available)Study TypeObjective/Goal
Resectable
NCT06358222ObservationalDevelop tumor-naïve MRD panel to predict nodal disease
NCT06426511Phase II Personalize consolidation toripalimab based on MRD
NCT06979661Early Phase I Use HaystackTM MRD to guide post-op RT/systemic therapy
NCT05536505Phase IIDe-escalate in MRD−; treat MRD+ with icotinib/osimertinib
NCT04317534 Phase IIObservation vs. pembrolizumab in stage I with ctDNA correlatives
NCT04712877 ObservationalValidate biomarker-integrated peri-operative care pathways including ctDNA
*NCT05079022Phase I/IITest benefits of adjuvant furmonertinib specifically in MRD+ stage I
*NCT04385368 Phase IIIDetermine if adding durvalumab to adjuvant chemotherapy improves DFS in MRD+ post-resection
*NCT04642469Phase IIITest durvalumab vs. placebo in patients who become MRD+ during surveillance after curative therapy
NCT06053099Prospective cohortDefine prognostic and mechanistic correlates of relapse during/after adjuvant osimertinib
*NCT04037150ObservationalEvaluate peri-operative ctDNA as a surveillance tool
NCT06323148 Phase III Test adjuvant osimertinib specifically in MRD+
NCT05457049ObservationalEvaluate therapy-free surveillance if MRD remains undetectable
NCT05059444 ObservationalValidate a novel commercial ctDNA assay for surveillance
*NCT04351555Phase III Evaluate benefits of MRD in neoadjuvant osimertinib ± chemo vs. chemo
NCT04966663Phase IITest adjuvant therapy in ctDNA+ vs. observation
NCT05167604ObservationalExplore MRD status and correlation with outcomes after adjuvant chemo in early NSCLC
NCT04367311Phase IIAssess intensified adjuvant chemo + atezolizumab for post-op ctDNA+ pts
*NCT04625699Phase IIEvaluate peri-operative IO doublet for patients who develop detectable ctDNA after standard adjuvant therapy
NCT05236114 ObservationalMap optimal peri-op timepoints for ctDNA MRD
NCT04638582Phase II Define benefits of preop pembrolizumab with ctDNA correlatives in early NSCLC
NCT05254782ObservationalQuantify detection rate and prognostic value of peri-operative ctDNA
NCT05460195Phase II Peri-op sintilimab and anlotinib based on MRD
NCT04585477Phase IIctDNA-adapted adjuvant immunotherapy strategies
Advanced/Metastatic
NCT04841811 Phase IIIEvaluate almonertinib in unresectable stage III EGFRm NSCLC; evaluate dynamic MRD-guided maintenance therapy with almonertinib
*NCT06020989 Phase II Determine if early chemo add-on for ctDNA-positive patients improves outcomes vs. lazertinib alone
NCT04912687 ObservationalImplement and evaluate baseline ctDNA testing at initial diagnosis
NCT05334277 Phase IITest ctDNA-guided escalation after furmonertinib induction
*NCT03865511 Phase IICharacterize on-treatment resistance using paired biopsies and ctDNA with osimertinib
NCT04737382 ObservationalDefine concordance and clinical utility of tumor NGS vs. plasma ctDNA
NCT05281406 Phase IIAssess benefit of adding platinum + pemetrexed for early ctDNA persistence on 1 L osimertinib
NCT05598528 ObservationalIdentify predictors of primary resistance to 3rd-gen EGFR TKIs
NCT05257967ObservationalQuantify incremental value of CSF ctDNA in leptomeningeal disease
NCT05401110 Phase IDetermine safety/activity of carotuximab + osimertinib; follow ctDNA+
NCT05534113Phase IIEvaluate sequential envafolimab after ctDNA clearance on almonertinib
NCT05813522Phase IIAssess high-dose furmonertinib efficacy in LM and validate CSF ctDNA monitoring
NCT04585490Phase IIIPersonalize consolidation durvalumab after CRT using ctDNA
NCT05198154 Observational Anticipate progression using ctDNA in long-responders
Screening/Risk Stratification
*NCT05117840ObservationalEvaluate blood-based GuardantLUNAR-2 screening assay
NCT06163846ObservationalIntegrate imaging and liquid biopsy for stratification
*NCT05020275 Observational Map resistance to inform subsequent therapy
NCT03774758 ObservationalValidate ctDNA signature for risk stratification
*NCT02194738Screening (ALCHEMIST)Test resected NSCLC for genetic mutations to facilitate accrual to randomized adjuvant studies of adjuvant treatment trials for resected NSCLC
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.

Share and Cite

MDPI and ACS Style

Dhillon, P.; Du, S.; Cheng, H. Circulating Tumor DNA as a Prognostic and Predictive Biomarker in Lung Cancer. Cancers 2025, 17, 3327. https://doi.org/10.3390/cancers17203327

AMA Style

Dhillon P, Du S, Cheng H. Circulating Tumor DNA as a Prognostic and Predictive Biomarker in Lung Cancer. Cancers. 2025; 17(20):3327. https://doi.org/10.3390/cancers17203327

Chicago/Turabian Style

Dhillon, Puneet, Simo Du, and Haiying Cheng. 2025. "Circulating Tumor DNA as a Prognostic and Predictive Biomarker in Lung Cancer" Cancers 17, no. 20: 3327. https://doi.org/10.3390/cancers17203327

APA Style

Dhillon, P., Du, S., & Cheng, H. (2025). Circulating Tumor DNA as a Prognostic and Predictive Biomarker in Lung Cancer. Cancers, 17(20), 3327. https://doi.org/10.3390/cancers17203327

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop