Tuberc Respir Dis > Volume 88(3); 2025 > Article
Oya, Tanaka, and Soo: Integration of Liquid Biopsy for Optimal Management of Non-small Cell Lung Cancer

Abstract

Molecular profiling of tumors from patients plays a crucial role in precision oncology. While tumor tissue-based genomic testing remains the gold standard in clinical management of patients with non-small cell lung cancer, advances in genomic technologies, the analysis of various bodily fluids, mainly blood but also saliva, pleural/pericardial effusions, urine, and cerebrospinal fluid is now feasible and readily available. In this review, we will focus on the clinical application of circulating tumor DNA (ctDNA) in patients with non-small cell lung cancer in the setting of early-stage disease, locally advanced disease with attention to the potential of ctDNA in prognostication, risk stratification, minimal residual disease, and in advanced disease, its role in the detection of genomic markers and mechanisms of acquired resistance. The role of ctDNA and liquid biopsies in lung cancer screening will also be discussed.

Key Figure

Introduction

Molecular profiling of tumors from patients plays a crucial role in precision oncology [1] in which tumor tissue is traditionally used. There are however barriers associated with the use of tumor tissue such as the invasive nature of sample acquisition and its inability to fully capture tumor heterogeneity. Furthermore, given its invasive nature, conducting tumor biopsies to monitor for treatment tumor response, relapse and determination of resistance would be a major challenge. With considerable progress made in genomic technologies, the analysis of various bodily fluids is now feasible and readily available. The testing of biological fluids, mainly blood but also saliva, pleural/pericardial effusions, urine, and cerebrospinal fluid (CSF), for tumor derived components is known as liquid biopsy. The use of liquid biopsies, especially with blood, has provided increased sensitivity in diagnosis as well as a noninvasive and convenient approach for repeated sampling during treatment (Table 1) [2]. A liquid biopsy can be used to assess a range of tumor derived components including circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), tumor derived extracellular vesicles (EVs) or exosomes, circulating cell-free RNA (cfRNA), and tumor educated platelets (Figure 1).
In this review, we will focus on the clinical application of ctDNA in the management of non-small cell lung cancer (NSCLC). A review of other tumor derived components is beyond the scope of this manuscript and have been reviewed elsewhere [3-5].

Essentials of Liquid Biopsy Analysis

Biomarkers play an important role in the management of cancer patients. Among the various new blood-based biomarkers proposed for lung cancer, one of the most promising is the measurement of DNA from the tumor to the systemic circulation, i.e., circulating tumor DNA or ctDNA. This section will analyze the background on which to base our understanding of ctDNA

1. Definitions of liquid biopsy, ctDNA, and cfDNA

cfDNA refer to short DNA fragments with an average length of 120 to 160 bp that are present outside cells. ctDNA is the subset of cfDNA that are derived from tumor cells following apoptosis and/or necrosis and are usually <145 bp in length. ctDNA is highly diluted in plasma, accounting for approximately 0.1%-10% of the total cfDNA and its concentration is impacted by clearance and degradation by nuclease activity [6,7]. While tumor tissue biopsy is the gold standard in establishing the diagnosis of NSCLC, diagnostic rates vary by region and biopsy technique, but it is reported that over 40% of patients in the United States required repeat biopsies before diagnosis [8]. Furthermore, the failure rate of molecular characterization has been reported in approximately 30% of cases [9]. ctDNA can thus be an used as an alternative tool in the molecular genotyping of patients with NSCLC, especially in situations where tissue or time is limited [10,11].

2. Clinical significance of liquid biopsy, ctDNA, and cfDNA

Liquid biopsy testing of tumor DNA has some advantages over tissue-based DNA testing including less invasive, shorter turn around time and lower sample isolation costs. A liquid biopsy enables the assessment of quantitative changes in ctDNA which could be used for real time monitoring of therapeutic effects. In addition, detection of genetic mutations, changes in copy number and chromosomal rearrangement have the potential to be applied to detect resistance mechanisms or treatment efficacy [12,13]. On the other hand, an important limitation of liquid biopsy is the inability to assess the histology of the tumor and the risk of false negative results due to either insensitive ctDNA assays or inadequate ctDNA levels in circulation (Table 1).
ctDNA levels are influenced by the degree of shedding and clearance. Factors associated with shedding include tumor burden with early-stage disease are less likely to shed ctDNA, metabolic activity, histologic type and grade, lymph node involvement, tumor microenvironment, and location of metastasis [14]. Factors associated with ctDNA clearance includes soluble blood enzymes such as DNase I, protease factor VII-activating protease (FSAP) and factor H as well clearance by organs such as liver, spleen and kidneys [15]. The advantages and disadvantages of tissue biopsy and liquid biopsy are summarized in Table 1. Multiple applications exist for ctDNA including molecular genotyping of disease, screening, detection of measurable disease, treatment selection, treatment monitoring and the molecular characterization of resistance (Figure 1).

Clinical Applications of Liquid Biopsy in NSCLC

ctDNA is a plasma biomarker that can be used for detection, monitoring, and treatment evaluation in multiple type of cancers. In this section, we will discuss the clinical utility of ctDNA in lung cancer.

1. Identification of therapeutic targets for targeted anticancer therapy

Although driver mutation-based therapy is widely used in NSCLC, complete and timely tissue genotyping is difficult, and few patients have been examined for all eight genomic biomarkers recommended by professional guidelines (National Comprehensive Cancer Network [NCCN] Guidelines 2024) [16]. Molecular genotyping using ctDNA could overcome the tissue sampling barrier in advanced NSCLC.
The detection of plasma ctDNA in advanced NSCLC is prognostic and is associated with metabolic tumor volume, more frequent in extrapulmonary disease (74%) than intrapulmonary disease (40%) [17]. In the Noninvasive vs. Invasive Lung Evaluation (NILE) study, a prospective trial that compared the utility of plasma next generation sequencing (NGS) using Guardant360 versus tissue genotyping in patients with advanced NSCLC, the Guardant360 assay had a concordance of >98.2% and 100% positive predictive value versus tissue testing. In addition, the median turnaround time (TAT) for cfDNA was significantly faster than for tissue (9 days vs. 15 days, p<0.0001) [18]. In a prospective observational study comparing tissue genotyping with ctDNA genotyping, NGS results were significantly better for ctDNA at the first visit after biopsy for diagnosis (85% vs. 9%, p<0.01), especially in cases with specific driver gene mutations significantly shorter time to treatment initiation in cases (10 days vs. 19 days, p<0.01) [19].
Another important trial on this topic is the Blood First Assay Screening Trial, a Phase II/III umbrella trial. This study was designed to evaluate plasma-based NGS for the detection of actionable genetic alterations in patients with advanced NSCLC and to examine the efficacy of targeted and immunotherapy-based therapies using plasma results alone. Confirmed objective response rate (ORR) was 87.4% for alectinib in the anaplastic lymphoma kinase (ALK) cohort, better than existing prospective trials [20]. The ROS1 cohort also showed response rates and progression-free survival (PFS) comparable to tissue-based assessments [21].
Another potential application of ctDNA in advanced NSCLC is the evaluation of blood-based tumor mutation burden (bTMB). Tumor mutation burden (TMB), defined as the total number of non-synonymous somatic mutations in the coding region of a cancer genome, is a predictive biomarker of immune checkpoint inhibitor (ICI) in a range of solid tumours [22,23] and exome or large panel sequencing allow for the quantification of TMB. More recently, the U.S. Food and Drug Administration (FDA) approved pembrolizumab for the treatment of cancer patients with TMB >10 mutations per megabase (mut/Mb) [24] FDA. FDA approves pembrolizumab for adults and children with TMB-H solid tumors in 2020 [25]. A retrospective study of two large randomized trials, bTMB was a predictive biomarker for PFS in patients treated with atezolizumab [26]. In a single arm prospective study of first line atezolizumab (Blood First Assay Screening Trial [BFAST] study), investigator-assessed PFS in the bTMB ≥ 16 versus bTMB < 16 mut/Mb groups were not statistically significant but bTMB ≥16 was associated with higher ORR [27]. In the BFAST study, Cohort C compared first line atezolizumab versus platinum doublet chemotherapy in patients with a high TMB (>16 mut/Mb). Unfortunately the study did not meet its primary endpoint of investigator-assessed PFS (hazard ratio [HR], 0.77; 95% confidence interval [CI], 0.59 to 1.00; p=0.05). A major limitation of this study was programmed death-ligand 1 (PD-L1) expression was unknown [28]. The Evaluation of Blood tumor mutation burden for improved efficacy of atezolizumab (BUDDY) study showed that high cfDNA concentration at C0 (cutoff: 8.6 ng/mL) and cycle 4/baseline bTMB ratio >1 was significantly associated with PFS in atezolizumab treatment after platinum combination therapy [29]. Future research is required to define the optimal bTMB cutoff and standardization of assay technologies [30,31].

2. Study of drug resistance mechanisms through analysis of emerging mutations following drug administration

The characterization of resistance mutations with plasma ctDNA genotyping has been investigated extensively. In the AZD9291 (Osimertinib) Versus Platinum-Based Doublet-Chemotherapy in Locally Advanced or Metastatic Non-Small Cell Lung Cancer (AURA3) study, of the 564 patients for whom both plasma and tumor tissue was performed on the Cobas-based epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI) post-progression screening, 41% were ctDNA T790M positive and 64% were tissue EGFR T790M positive, for a ctDNA/tissue concordance rate of 61% [32]. Compared to the EGFR sensitive mutations L858R and exon19 deletion, the ctDNA-tissue concordance is lower for EGFR T790M. While sensitive mutations are clonal mutations and thus relatively homogeneous within the tumor, resistance genes such as T790M are subclonal mutations, and thus greater tumor heterogeneity is assumed to be the cause of the tissue-ctDNA discrepancy.
In the AZD9291 Versus Gefitinib or Erlotinib in Patients With Locally Advanced or Metastatic Non-small Cell Lung Cancer (FLAURA) study, 109 patients who were included in the osimertinib arm and had EGFR mutations detected by ctDNA at baseline had mesenchymal epithelial transition (MET) amplification at the time of resistance (amplification [n=17, 16%] and the EGFR C797S mutation [n=7, 6%]) was more frequent at the time of resistance [33]. Chinese Thoracic Oncology Group (CTONG1509), a phase III trial comparing erlotinib with erlotinib plus bevacizumab, also showed 57% (33/58) patients with known resistance mechanisms including EGFR T790M, oncogenic fusion, amplification, mitogen-activated protein kinase (MAPK)/phosphoinositide 3-kinase (PI3K) changes, and cell cycle gene changes at progression of disease [34]. Similarly, ctDNA has successfully detected resistance mutations after osimertinib as a second line [32,35,36].
The molecular mechanisms of resistance by ctDNA genotyping have also been described in patients after targeted therapy for BRAF V600E mutations, ALK rearrangement, MET exon14 skipping mutations, and ROS1 fusions [37-40].
The utility of ctDNA to monitor for plasma EGFR T790M acquired resistance in patients treated with a 1st- or 2nd- generation EGFR-TKIs. The authors found the median time of detectable plasma EGFR T790M was 2.2 months (range, 0.8 to 6) before clinical progressive disease [41].
This intriguing finding posits the question: does the early detection of acquired EGFR T790M resistance mutations improve outcomes with a change in therapeutic agent? To test this proposition, the Osimertinib Treatment on EGFR T790M Plasma Positive NSCLC Patients (APPLE) trial was designed to evaluate the feasibility of longitudinal plasma EGFR T790M monitoring performed intensively using the Cobas EGFR Mutation Test v2 and the best sequencing strategy from gefitinib to osimertinib (molecular progression by T790M positivity in plasma test or radiographic progression by Response Evaluation Criteria in Solid Tumors [RECIST] 1.1 criteria). In the trial arm B, which switches to osimertinib if T790M mutation is detected in plasma ctDNA, 17% of patients (eight of 47) switched to it before radiographic progression. However, overall survival (OS) was comparable between the group that switched early to osimertinib based on ctDNA detection and the group that switched after disease progression was confirmed by RECIST 1.1 criteria [42].

3. Monitoring residual tumor presence, tumor reduction, or recurrence following drug administration

It is well established clearance of ctDNA has been shown to be a prognostic factor in multiple clinical trials, including the FLAURA trial of osimertinib versus first generation EGFR-TKI for EGFR mutations [36], the A Study Of Lorlatinib Versus Crizotinib In First Line Treatment Of Patients With ALK-Positive NSCLC (CROWN) trial of lorlatinib versus crizotinib for ALK fusions [43], the Phase 1/2 Study of MRTX849 in Patients With Cancer Having a KRAS G12C Mutation (KRYSTAL-1) trial of adaglasib for KRAS G12C mutation [44], and the BRAF-targeted therapies (BRAF-TT) trial of targeted therapy against BRAF V600E mutation [37]. More recent phase III trials have reported on the association between ctDNA clearance status and outcomes. In the A Study of Amivantamab and Lazertinib Combination Therapy Versus Osimertinib in Locally Advanced or Metastatic Non-Small Cell Lung Cancer (MARIPOSA) trial examining the efficacy of 1st-line amivantamab plus lazertinib versus osimertinib in patients with EGFR mutated NSCLC, 78% of patients had available droplet digital polymerase chain reaction (ddPCR) testing for ctDNA at baseline. In the patients with ctDNA detected at baseline, the PFS for the group of amivantamab plus lazertinib and the group of osimertinib were 20.3 and 14.8 months, respectively (HR, 0.68), while, in the patients with no detectable ctDNA, the PFS for the group of amivantamab plus lazertinib and the group of osimertinib were 27.7 and 21.9 months, respectively (HR, 0.72). In addition, the therapeutic efficacies of amivantamab plus lazertinib were greater than that of osimertinib, regardless of ctDNA clearance at 9 weeks [45]. In an exploratory analysis of the FLAURA2 trial comparing the clinical efficacies of osimertinib plus chemotherapy to osimertinib, a better PFS for the group of osimertinib plus chemotherapy was observed in patients with positive ctDNA. Regardless of ctDNA clearance, the addition of chemotherapy to osimertinib provided a PFS advantage over osimertinib alone [46].

4. Study of dose escalation based on ctDNA status

To evaluate the role of treatment escalation based on plasma ctDNA status, a number of trials are ongoing. In a randomized phase II trial by Memorial Sloan Kettering Cancer Center (NCT04410796) where patients with advanced EGFR NSCLC without ctDNA clearance after one cycle of osimertinib are randomized to carboplatin and pemetrexed plus osimertinib versus continuation of osimertinib. In another ongoing study, patients with plasma EGFR mutations detected after one cycle of lazertinib are randomized to pemetrexed, carboplatin and lazertinib versus lazertinib alone (Lazertinib and Chemotherapy Combination in EGFR-mutant NSCLC Patients Without ctDNA Clearance After lead-in Lazertinib Monotherapy [CHAMELEON] trial, NCT06020989) in which chemotherapy is combined with lazertinib based plasma EGFR mutation status [47]. Furmonertinib Monotherapy and Combination Therapy in Advanced EGFR Mutant NSCLC With Uncleared ctDNA (FOCUS-C) is a randomized phase II where patients without ctDNA clearance with furmonertinib are randomized to furmonertinib or furmonertinib plus chemotherapy (pemetrexed and carboplatin) or furmonertinib plus chemotherapy plus bevacizumab (NCT05334277).

5. Role of ctDNA in oligometastatic disease

In advanced stage NSCLC, oligometastatic disease (OMD) has limited number of metastatic lesions (usually up to 5). The addition of local treatment is expected to prolong survival, but there is no clear consensus on its definition [48-50]. In 2020, European Society for Radiotherapy and Oncology/European Organisation for Research and Treatment of Cancer (ESTRO/EORTC) classified OMD into synchronous, metachronous, induced, persistence, and progression [51].
In a prospective study of ctDNA in patients with OMD, patients with ctDNA detected before local ablative radiotherapy had an mPFS of 5.4 months, significantly shorter than the 8.4 months for ctDNA negative patients (HR, 1.57; 95% CI, 1.15 to 2.13; p=0.004) [52], consistent with the notion detectable ctDNA is a prognostic factor.
More recently, a nonrandomized study was conducted to assess the feasibility of adaptive TKI de-escalation guided by ctDNA to achieve complete remission after local consolidative therapy (LCT) in patients with advanced NSCLC. The median PFS was 18.4 months and the authors concluded an adaptive de-escalation TKI approach was feasible in those with no lesions after LCT with undetectable ctDNA [53]. While these results are promising, potential challenges include the presence of ctDNA non-shedders and the use of a highly sensitive ctDNA assay is crucial. Furthermore, a randomized trial is required to confirm the validity of a de-escalation treatment strategy.

6. Role of minimal residual disease in early-stage NSCLC

Minimal Residual Disease (MRD) is a micro-tumor that remains in the body after or during cancer treatment. MRD was originally applied in hematologic malignancies and is a prognostic factor in acute leukemias and plays an important role in the choice of appropriate treatment. Quantitative polymerase chain reaction and multiparameter flow cytometry are typical methods used to detect MRD. In solid tumors, emerging data suggests blood-based ctDNA analysis is effective in detecting MRD [54-56]. Several prospective observational studies have reported MRD is associated with increased recurrence.
In a landmark analysis of plasma samples taken within 120 days of surgery detected ctDNA in 25% of patients, including 49% of all patients who experienced clinical recurrence [57]. In a prospective observational study of 261 patients with resected stage I to III NSCLC, longitudinal undetectable MRD was associated with decreased disease recurrence with a negative predictive value of 96.8%. These results suggests a potentially cured patient population in those with longitudinal undetectable MRD. Conversely, the positive predictive value of longitudinal detectable MRD was 89.1%, with a median lead time of 3.4 months [58].

1) Role of MRD in early-stage NSCLC treated with immune checkpint inhibitors

In the Neo-Adjuvant Immunotherapy (NADIM) II study, patients with negative ctDNA at baseline had significantly longer disease-free survival (DFS) and OS than those with positive ctDNA (DFS: HR, 0.38; 95% CI, 0.15 to 0.95; OS: HR, 0.22; 95% CI, 0.07 to 0.64) and pretreatment ctDNA added some prognostic information to that of the clinical stage [59].
In the Impower 010 trial of atezolizumab as adjuvant therapy after complete resection, of the 20% of patients (n=103) who had positive ctDNA postoperatively, 62% had ctDNA resolved after chemotherapy. Patients with ctDNA clearance in the best supportive care (BSC) group had prolonged DFS compared to patients with residual ctDNA. Adjuvant atezolizumab improved DFS regardless of ctDNA clearance status [60].
Furthermore, the CheckMate816 study showed better pathologic complete response rates and DFS in patients with ctDNA clearance pre and post-surgery [61]. A major limitation in clinical decision-making based on ctDNA is that ctDNA quantitation is not standardized and the concordance between the different platforms used to assess ctDNA quantity is rarely assessed.

2) Role of MRD in resected EGFR mutated NSCLC

In a Korean study of longitudinal monitoring of ctDNA as a biomarker of MRD in patients with resected stages I to IIIA EGFR-M+ NSCLC, approximately 20% of stage I patients were ctDNA positive preoperatively, 79% of stage IA and 78% of stage IB patients did not have ctDNA clearance postoperatively, suggesting EGFR mutated NSCLC is partly a systemic disease even at very early stages. In addition, in patients with early EGFR mutated NSCLC after surgery, continuous postoperative ctDNA monitoring detected MRD before radiological recurrence in 69% of patients with exon 19 deletion and in 20% with L858R mutation. This suggests that ctDNA may be useful for monitoring recurrence in EGFR mutated NSCLC [62].
In the same study, postoperative MRD status was prognostic. In stage IB-III patients grouped according to the presence of ctDNA at landmark time points: group A (baseline ctDNA negative), group B (baseline ctDNA positive, but postoperative MRD negative) and group C (baseline ctDNA positive and postoperative MRD positive), the 3-year DFS rates were 84%, 78%, and 50%, respectively [62].
In the AZD9291 Versus Placebo in Patients With Stage IB-IIIA Non-small Cell Lung Carcinoma, Following Complete Tumour Resection With or Without Adjuvant Chemotherapy (ADAURA) study patients with completely resected with stage II-III EGFR mutated NSCLC treated with 3 years of osimertinib was associated with a significant improvement in DFS and OS versus placebo and is approved in many countries [63]. Correlative ctDNA studies from the ADAURA study was presented recently. Baseline ctDNA detection was associated with a poorer prognosis in both the osimertinib and placebo groups. The presence of MRD were detected in 13% and 49% of patients treated with osimertinib and placebo, respectively. The presence of MRD accelerated the identification of recurrence by 4.7 months (95% CI, 2.2 to 5.6). In the osimertinib group, 8% had an MRD event during treatment and 17% had post treatment. More than half of post MRD events were observed within 12 months from the end of treatment [64].

7. Role of MRD in stage III NSCLC

In patients with locally advanced unresectable stage III NSCLC, the success of the A Global Study to Assess the Effects of MEDI4736 Following Concurrent Chemoradiation in Patients With Stage III Unresectable Non-Small Cell Lung Cancer (PACIFIC) trial led to the approval of durvalumab consolidation therapy after chemoradiotherapy (CRT). The 5-year PFS rate was 19.0% in the BSC group and 33.1% in the durvalumab group, and the 5-year survival rate was 33.4% in the BSC group compared to 42.9% in the durvalumab group [65].
In a retrospective study of ctDNA in patients NSCLC patients treated with CRT with or without consolidation ICI, cancer personalized profiling by deep sequencing (CAPP-Seq) ctDNA analysis of 218 samples from 65 patients undergoing CRT showed that patients with undetectable ctDNA after CRT had longer DFS, regardless of whether they had been treated with ICI. Of note, one MRD negative case died of G5 pneumonitis. Among patients with positive MRD, those who received consolidation ICI treatment had significantly longer DFS than those who did not. Early ctDNA responses identified patients who would benefit particularly from ICI treatment [66]. These results, albeit retrospective in nature, suggests consolidation ICI improves outcomes for NSCLC patients with MRD and ctDNA analysis may be useful in the personalization of consolidation therapy. Prospective randomized studies are required to confirm if personalization of consolidation ICI based on MRD status can be applied.
In the LAURA trial, osimertinib therapy after CRT prolonged DFS and OS in EGFR mutated locally advanced unresectable stage III NSCLC [67]. The length of treatment in adjuvant therapy is longer than in the advanced stage, and there is concern side effect including quality of life. Therefore, MRD may have a promising role in EGFR -positive early-stage NSCLC by (1) replacing or complementing pathologic stage as a criterion for selection of adjuvant TKI extension therapy; (2) reducing treatment and economic/resource burden; (3) improving prognosis; and (4) reducing frequency of imaging diagnosis.

8. Role of ctDNA in screening for early lung cancer

Randomized trials of lung cancer screening with low-dose computed tomography (LDCT) in selected high risk patient population has been shown to be effective in reducing lung cancer mortality [68,69]. Blood-based biomarkers may potential advantages over LDCT including ease of convenience, high-throughput, lower costs and lack of exposure for participants to ionizing radiation. However several challenges using ctDNA as a screening modality. First, the sensitivity may be inadequate due to limited ctDNA shedding or assay sensitivity. Plasma ctDNA mutations may be non-malignant or premalignant and their tissue of origin is often unknown. To overcome these barriers, screening in combination with positron emission tomography-computed tomography, protein biomarkers, methylation patterns, miRNAs, etc. is being considered.
In a study comparing the mutational profile of cfDNA in either apparent heathy subjects and patients with stage I or II colorectal, breast, lung, or ovarian cancer, somatic mutations were detected in the plasma of 71%, 59%, 59%, and 68%, respectively and sensitivity increased with stage of disease. However, approximately 16% of healthy controls had mutations in genes associated with clonal hematopoiesis of indeterminate potential [70].
The Circulating Cell-Free Genome Atlas Study (CCGA; NCT02889978) is a prospective, case-control, observational study that examined whether a blood-based multiple cancer early detection test combining targeted methylation of ctDNA and machine learning could detect cancer signals across multiple cancer types and predict cancer signal origin by detecting cancer signals across multiple cancer types. The sensitivity and specificity for all carcinomas were 51.5% and 99.5%, respectively, with a sensitivity of 75% for lung cancer For NSCLC, sensitivity increased with stage, with 21.9% for stage I, 79.5% for stage II, 90.7% for stage III, and 95.2% for stage IV [71]. The Cancer SEEK trial investigated the role of screening with mutated DNA and eight standard biomarkers in cfDNA and reported a sensitivity of 60% for resectable lung cancer [72]. In another study integrating genomic features with machine learning, termed 'lung cancer likelihood in plasma' (Lung-CLiP), this approach could discriminate early-stage lung cancer patients from risk-matched controls with a specificity of 96% [73].
Collectively, ctDNA assays have a high specificity for lung cancer but sensitivity is limited, especially for early-stage disease. Further prospective evaluation of blood-based biomarkers are ongoing to confirm its utility in lung cancer screening.

ctDNA beyond Blood

In addition to blood, ctDNA and other tumor derived components have been analyzed in non-blood biological fluids such as pleural effusions, saliva, urine, peritoneal fluid, CSF, and bronchoalveolar lavage (BAL) fluid.
Urine and saliva are even more noninvasive than blood. saliva is still underreported, but urine is reported to be potentially useful for screening and monitoring [74]. Examination of ctDNA using urine and blood samples in EGFR mutated NSCLC showed that EGFR detection using urinary ctDNA in the ddPCR assay was very consistent with results obtained from plasma DNA [75]. It was also noted that daily sampling of urine ctDNA after osimertinib initiation in EGFR-positive NSCLC may be useful for early assessment of patient response [76]. Furthermore, urinary methylation were analyzed for the relevant methylation DNA markers cysteine dioxygenase type 1 (CDO1), SRY-box transcription factor 17 (SOX17), and tachykinin precursor 1 (TAC1) in early-stage NSCLC, with urine CDO1 and SOX17 showing increased methylation levels in NSCLC patients compared to gender and age-matched controls [77].
The possibility that CSF may be useful in monitoring intracranial metastases is also reported [78,79]. The sensitivity of the CSF genotype to EGFR sensitizing mutations was 93.3% before osimertinib treatment for patients with NSCLC with leptomeningeal metastases (LM). And in cases of LM exacerbated by osimertinib, resistance mechanisms such as C797S mutation, MET dysregulation, and concurrent occurrence of tumor protein p53 (TP53) plus RB Transcriptional Corepressor 1 (RB1) were detected in the CSF, suggesting that it may be useful in detecting resistance [80]. EGFR mutated LM cases in which dynamic changes in CSF ctDNA correlate well with clinical therapy are also reported [81]. Prospective studies also showed that CSF and plasma tumor DNA levels were positively correlated, with cytology having a sensitivity of 81.8% and CSF-tDNA having a sensitivity of 91.7%, making it useful for leptomeningeal disease diagnosis. In patients who progressed on targeted therapy in this prospective study, resistance mutations such as EGFR T790M and MET amplification were common in peripheral blood, but rare in time-matched CSF, indicating differences in resistance mechanisms based on anatomic compartments, and CSF-tDNA may be useful for treatment selection against intracranial [82].
BAL fluid is a noninvasive diagnostic tool for NSCLC. The collection process is minimally invasive and can be repeated without significant risk to the patient, making it a promising option in the clinical management of NSCLC. ctDNA analysis using EV-derived DNA obtained in bronchoalveolar lavage fluid (BALF) reports a TAT of only 1 to 2 days and an overall concordance rate of 99.2% between tissue biopsy and EV-based BALF liquid biopsy, including EGFR mutation subtypes [83].

Conclusion

Although ctDNA is useful in predicting prognosis, it is still not well concordance with results from tissue samples currently and does not provide information on histology, so basically, tissue-based testing is preferred before treatment. However, ctDNA testing is still useful when more rapid TAT is needed clinically or when tissue samples are not available. Potential uses of ctDNA include screening, detection of measurable disease progression prior to imaging, treatment selection tools, monitoring, and detection of resistance. MRD and ctDNA monitoring is not yet recommended in clinical practice currently, but trials are underway to confirm its various usefulness. Patient selection for MRD-based adjuvant therapy will be most promising in recent years, when treatment is shifting to earlier stage.

Notes

Authors’ Contributions

Conceptualization: all authors. Methodology: all authors. Formal analysis: all authors. Data curation: all authors. Project administration: Soo RA. Validation: all authors. Investigation: all authors. Writing - original draft preparation: Oya Y, Tanaka I. Writing - review and editing: all authors. Approval of final manuscript: all authors.

Conflicts of Interest

Ross A. Soo has received honorarium from Abbvie, Amgen, AnHeart, Astrazeneca, Bayer, BMS, Boehringer Ingelheim, Daiichi Sankyo, GSK, J INTS BIO, Janssen, Lily, Merck, Merck Serono, Novartis, Pfizer, Puma, Roche, Sanofi, Taiho, Takeda, Thermo Fisher, Yuhan Corporation and Chugai and research funding from Astrazeneca, Boehringer Ingelheim, and Pfizer. Yuko Oya reports consulting/advisory roles: AstraZeneca; Honoraria: AstraZeneca, Chugai Pharmaceuticals, Bristol-Myers Squibb, Merck Sharp and Dohme, Eli Lilly, MSD, Takeda Pharmaceuticals, and Amgen. Ichidai Tanaka received research funding from Chugai Pharmaceutical Co. and reports consulting/advisory roles for AstraZeneca, Honoraria, AstraZeneca, Chugai Pharmaceuticals, Bristol-Myers Squibb, MSD K. K., Novartis Pharma K.K., Takeda Pharmaceuticals, Ono Pharmaceutical Co., and Daiichi Sankyo Company.

Funding

No funding to declare.

Fig. 1.
Clinical applications of circulating tumor DNA (ctDNA): screeening, diagnosis, molecular diagnosis, detection of resistance, and detection of residual disease. CSF: cerebrospinal fluid; CTC: circulating tumor cell.
trd-2024-0146f1.jpg
trd-2024-0146f2.jpg
Table 1.
Comparison of tissue biopsy and liquid biopsy
Advantage Disadvantage
Tissue biopsy High sensitivity High invasive
A wealth of evidence High costs
Clinically available Tumor heterogeneity cannot be assessed.
Liquid biopsy Less invasive Inability to assess the histology of the tumor
Shorter turn around time The risk of false negative
Lower costs Not validated enough
Quantitative changes in ctDNA can be evaluated Possible difficulty in differentiating clonal hematopoiesis from genetic abnormalities derived from tumor cells
Captures heterogeneity not captured by tissue specimens and provides a complete picture of the disease
Easy and repeatable sample collection

ctDNA: circulating tumor DNA.

References

1. Hodson R. Precision medicine. Nature 2016;537:S49.
crossref pmid pdf
2. Crowley E, Di Nicolantonio F, Loupakis F, Bardelli A. Liquid biopsy: monitoring cancer-genetics in the blood. Nat Rev Clin Oncol 2013;10:472-84.
crossref pmid pdf
3. Andrikou K, Rossi T, Verlicchi A, Priano I, Cravero P, Burgio MA, et al. Circulating tumour cells: detection and application in advanced non-small cell lung cancer. Int J Mol Sci 2023;24:16085.
crossref pmid pmc
4. Lohajova Behulova R, Bugalova A, Bugala J, Struharnanska E, Safranek M, Juras I. Circulating exosomal miRNAs as a promising diagnostic biomarker in cancer. Physiol Res 2023;72(S3):S193-207.
crossref pmid pmc
5. Wankhede D, Grover S, Hofman P. Circulating tumor cells as a predictive biomarker in resectable lung cancer: a systematic review and meta-analysis. Cancers (Basel) 2022;14:6112.
crossref pmid pmc
6. Diehl F, Schmidt K, Choti MA, Romans K, Goodman S, Li M, et al. Circulating mutant DNA to assess tumor dynamics. Nat Med 2008;14:985-90.
crossref pmid pmc pdf
7. Fleischhacker M, Schmidt B. Circulating nucleic acids (CNAs) and cancer: a survey. Biochim Biophys Acta 2007;1775:181-232.
crossref pmid
8. Zhang Y, Shi L, Simoff MJ, J Wagner O, Lavin J. Biopsy frequency and complications among lung cancer patients in the United States. Lung Cancer Manag 2020;9:LMT40.
crossref pmid pmc
9. Vanderlaan PA, Yamaguchi N, Folch E, Boucher DH, Kent MS, Gangadharan SP, et al. Success and failure rates of tumor genotyping techniques in routine pathological samples with non-small-cell lung cancer. Lung Cancer 2014;84:39-44.
crossref pmid pmc
10. Rolfo C, Mack PC, Scagliotti GV, Baas P, Barlesi F, Bivona TG, et al. Liquid biopsy for advanced Non-Small Cell Lung Cancer (NSCLC): a statement paper from the IASLC. J Thorac Oncol 2018;13:1248-68.
crossref pmid
11. 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;5:173-80.
crossref pmid
12. Lone SN, Nisar S, Masoodi T, Singh M, Rizwan A, Hashem S, et al. Liquid biopsy: a step closer to transform diagnosis, prognosis and future of cancer treatments. Mol Cancer 2022;21:79.
crossref pmid pmc pdf
13. Pascual J, Attard G, Bidard FC, Curigliano G, De Mattos-Arruda L, Diehn M, et al. ESMO recommendations on the use of circulating tumour DNA assays for patients with cancer: a report from the ESMO Precision Medicine Working Group. Ann Oncol 2022;33:750-68.
crossref pmid
14. Abbosh C, Birkbak NJ, Wilson GA, Jamal-Hanjani M, Constantin T, Salari R, et al. Phylogenetic ctDNA analysis depicts early-stage lung cancer evolution. Nature 2017;545:446-51.
pmid pmc
15. Vivancos A, Tabernero J. Circulating tumor DNA as a novel prognostic indicator. Nat Med 2022;28:2255-6.
crossref pmid pdf
16. Riely GJ, Wood DE, Ettinger DS, Aisner DL, Akerley W, Bauman JR, et al. Non-small cell lung cancer, version 4.2024, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw 2024;22:249-74.
pmid
17. Jee J, Lebow ES, Yeh R, Das JP, Namakydoust A, Paik PK, et al. Overall survival with circulating tumor DNA-guided therapy in advanced non-small-cell lung cancer. Nat Med 2022;28:2353-63.
pmid pmc
18. Leighl NB, Page RD, Raymond VM, Daniel DB, Divers SG, Reckamp KL, et al. Clinical utility of comprehensive cell-free DNA analysis to identify genomic biomarkers in patients with newly diagnosed metastatic non-small cell lung cancer. Clin Cancer Res 2019;25:4691-700.
crossref pmid pdf
19. Thompson JC, Aggarwal C, Wong J, Nimgaonkar V, Hwang WT, Andronov M, 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.
crossref pmid pmc
20. Dziadziuszko R, Mok T, Peters S, Han JY, Alatorre-Alexander J, Leighl N, 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;16:2040-50.
crossref pmid
21. Peters S, Gadgeel SM, Mok T, Nadal E, Kilickap S, Swalduz A, et al. Entrectinib in ROS1-positive advanced non-small cell lung cancer: the phase 2/3 BFAST trial. Nat Med 2024;30:1923-32.
crossref pmid pmc pdf
22. Chan TA, Yarchoan M, Jaffee E, Swanton C, Quezada SA, Stenzinger A, et al. Development of tumor mutation burden as an immunotherapy biomarker: utility for the oncology clinic. Ann Oncol 2019;30:44-56.
crossref pmid pmc
23. Samstein RM, Lee CH, Shoushtari AN, Hellmann MD, Shen R, Janjigian YY, et al. Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat Genet 2019;51:202-6.
pmid pmc
24. Marabelle A, Fakih M, Lopez J, Shah M, Shapira-Frommer R, Nakagawa K, et al. Association of tumour mutational burden with outcomes in patients with advanced solid tumours treated with pembrolizumab: prospective biomarker analysis of the multicohort, open-label, phase 2 KEYNOTE-158 study. Lancet Oncol 2020;21:1353-65.
crossref pmid
25. Marcus L, Fashoyin-Aje LA, Donoghue M, Yuan M, Rodriguez L, Gallagher PS, et al. FDA approval summary: pembrolizumab for the treatment of tumor mutational burden-high solid tumors. Clin Cancer Res 2021;27:4685-9.
crossref pmid pmc pdf
26. Gandara DR, Paul SM, Kowanetz M, Schleifman E, Zou W, Li Y, et al. Blood-based tumor mutational burden as a predictor of clinical benefit in non-small-cell lung cancer patients treated with atezolizumab. Nat Med 2018;24:1441-8.
crossref pmid pdf
27. Kim ES, Velcheti V, Mekhail T, Yun C, Shagan SM, Hu S, et al. Blood-based tumor mutational burden as a biomarker for atezolizumab in non-small cell lung cancer: the phase 2 B-F1RST trial. Nat Med 2022;28:939-45.
crossref pmid pmc pdf
28. Peters S, Dziadziuszko R, Morabito A, Felip E, Gadgeel SM, Cheema P, et al. Atezolizumab versus chemotherapy in advanced or metastatic NSCLC with high blood-based tumor mutational burden: primary analysis of BFAST cohort C randomized phase 3 trial. Nat Med 2022;28:1831-9.
crossref pmid pmc pdf
29. Park CK, Jun HR, Oh HJ, Lee JY, Cho HJ, Kim YC, et al. Evaluation of blood tumor mutation burden for the efficacy of second-line atezolizumab treatment in non-small cell lung cancer: BUDDY trial. Cells 2023;12:1246.
crossref pmid pmc
30. Merino DM, McShane LM, Fabrizio D, Funari V, Chen SJ, White JR, et al. Establishing guidelines to harmonize tumor mutational burden (TMB): in silico assessment of variation in TMB quantification across diagnostic platforms: phase I of the Friends of Cancer Research TMB Harmonization Project. J Immunother Cancer 2020;8:e000147.
crossref pmid pmc
31. Stenzinger A, Endris V, Budczies J, Merkelbach-Bruse S, Kazdal D, Dietmaier W, et al. Harmonization and standardization of panel-based tumor mutational burden measurement: real-world results and recommendations of the quality in pathology study. J Thorac Oncol 2020;15:1177-89.
crossref pmid
32. Mok TS, Wu YL, Ahn MJ, Garassino MC, Kim HR, Ramalingam SS, et al. Osimertinib or platinum-pemetrexed in EGFR T790M-positive lung cancer. N Engl J Med 2017;376:629-40.
crossref pmid pmc
33. Chmielecki J, Gray JE, Cheng Y, Ohe Y, Imamura F, Cho BC, et al. Candidate mechanisms of acquired resistance to first-line osimertinib in EGFR-mutated advanced non-small cell lung cancer. Nat Commun 2023;14:1070.
crossref pmid pmc pdf
34. Zhou Q, Xu CR, Cheng Y, Liu YP, Chen GY, Cui JW, et al. Bevacizumab plus erlotinib in Chinese patients with untreated, EGFR-mutated, advanced NSCLC (ARTEMIS-CTONG1509): a multicenter phase 3 study. Cancer Cell 2021;39:1279-91.
crossref pmid
35. Ang YL, Zhao X, Reungwetwattana T, Cho BC, Liao BC, Yeung R, et al. A phase II study of osimertinib in patients with advanced-stage non-small cell lung cancer following prior Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitor (EGFR TKI) therapy with EGFR and T790M mutations detected in plasma circulating tumour DNA (PLASMA Study). Cancers (Basel) 2023;15:4999.
crossref pmid pmc
36. Gray JE, Ahn MJ, Oxnard GR, Shepherd FA, Imamura F, Cheng Y, et al. Early clearance of plasma epidermal growth factor receptor mutations as a predictor of outcome on osimertinib in advanced non-small cell lung cancer; exploratory analysis from AURA3 and FLAURA. Clin Cancer Res 2023;29:3340-51.
crossref pmid pdf
37. Ortiz-Cuaran S, Mezquita L, Swalduz A, Aldea M, Mazieres J, Leonce C, et al. Circulating tumor DNA genomics reveal potential mechanisms of resistance to BRAF-targeted therapies in patients with BRAF-mutant metastatic non-small cell lung cancer. Clin Cancer Res 2020;26:6242-53.
crossref pmid pdf
38. Recondo G, Bahcall M, Spurr LF, Che J, Ricciuti B, Leonardi GC, et al. Molecular mechanisms of acquired resistance to MET tyrosine kinase inhibitors in patients with MET exon 14-mutant NSCLC. Clin Cancer Res 2020;26:2615-25.
crossref pmid pdf
39. Shaw AT, Solomon BJ, Besse B, Bauer TM, Lin CC, Soo RA, et al. ALK resistance mutations and efficacy of lorlatinib in advanced anaplastic lymphoma kinase-positive non-small-cell lung cancer. J Clin Oncol 2019;37:1370-9.
crossref pmid pmc
40. Dagogo-Jack I, Rooney M, Nagy RJ, Lin JJ, Chin E, Ferris LA, et al. Molecular analysis of plasma from patients with ROS1-positive NSCLC. J Thorac Oncol 2019;14:816-24.
crossref pmid pmc
41. Zheng D, Ye X, Zhang MZ, Sun Y, Wang JY, Ni J, et al. Plasma EGFR T790M ctDNA status is associated with clinical outcome in advanced NSCLC patients with acquired EGFR-TKI resistance. Sci Rep 2016;6:20913.
crossref pmid pmc pdf
42. Remon J, Besse B, Aix SP, Callejo A, Al-Rabi K, Bernabe R, et al. Overall survival from the EORTC LCG-1613 APPLE trial of osimertinib versus gefitinib followed by osimertinib in advanced EGFR-mutant non-small-cell lung cancer. J Clin Oncol 2024;42:1350-6.
crossref pmid
43. Soo RA, Martini JF, van der Wekken AJ, Teraoka S, Ferrara R, Shaw AT, et al. Early circulating tumor DNA dynamics and efficacy of lorlatinib in patients with treatment-naive, advanced, ALK-positive NSCLC. J Thorac Oncol 2023;18:1568-80.
crossref pmid
44. Paweletz CP, Heavey GA, Kuang Y, Durlacher E, Kheoh T, Chao RC, et al. Early changes in circulating cell-free KRAS G12C predict response to adagrasib in KRAS mutant non-small cell lung cancer patients. Clin Cancer Res 2023;29:3074-80.
crossref pmid pmc pdf
45. Felip E, Cho BC, Gutierrez V, Alip A, Besse B, Lu S, 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;35:805-16.
pmid
46. Janne PA, Kobayashi K, Robichaux J, Lee CK, Sugawara S, Yang TY, et al. Abstract CT017: FLAURA2: exploratory analysis of baseline (BL) and on-treatment plasma EGFRm dynamics in patients (pts) with EGFRm advanced NSCLC treated with first-line (1L) osimertinib (osi)±platinum-pemetrexed. Cancer Res 2024;84(7 Supplement):CT017.
crossref pdf
47. Desai A, Vazquez 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) 2024;16:940.
crossref pmid pmc
48. Dingemans AC, Hendriks LE, Berghmans T, Levy A, Hasan B, Faivre-Finn C, et al. Definition of synchronous oligometastatic non-small cell lung cancer: a consensus report. J Thorac Oncol 2019;14:2109-19.
pmid
49. Lievens Y, Guckenberger M, Gomez D, Hoyer M, Iyengar P, Kindts I, et al. Defining oligometastatic disease from a radiation oncology perspective: an ESTRO-ASTRO consensus document. Radiother Oncol 2020;148:157-66.
crossref pmid
50. Giaj-Levra N, Giaj-Levra M, Durieux V, Novello S, Besse B, Hasan B, et al. Defining synchronous oligometastatic non-small cell lung cancer: a systematic review. J Thorac Oncol 2019;14:2053-61.
crossref pmid
51. Guckenberger M, Lievens Y, Bouma AB, Collette L, Dekker A, deSouza NM, et al. Characterisation and classification of oligometastatic disease: a European Society for Radiotherapy and Oncology and European Organisation for Research and Treatment of Cancer consensus recommendation. Lancet Oncol 2020;21:e18-28.
crossref pmid
52. Semenkovich NP, Badiyan SN, Samson PP, Stowe HB, Wang YE, Star R, et al. Pre-radiotherapy ctDNA liquid biopsy for risk stratification of oligometastatic non-small cell lung cancer. NPJ Precis Oncol 2023;7:100.
crossref pmid pmc pdf
53. Dong S, Wang Z, Zhang JT, Yan B, Zhang C, Gao X, et al. Circulating tumor DNA-guided de-escalation targeted therapy for advanced non-small cell lung cancer: a non-randomized controlled trial. JAMA Oncol 2024;10:932-40.
crossref pmid
54. Prindull G. Minimal residual neoplastic disease: concept, pathogenesis, and supplementary therapeutic possibilities. Cancer Treat Rev 1986;13:177-94.
crossref pmid
55. Pui CH, Pei D, Coustan-Smith E, Jeha S, Cheng C, Bowman WP, et al. Clinical utility of sequential minimal residual disease measurements in the context of risk-based therapy in childhood acute lymphoblastic leukaemia: a prospective study. Lancet Oncol 2015;16:465-74.
crossref pmid pmc
56. Cescon DW, Bratman SV, Chan SM, Siu LL. Circulating tumor DNA and liquid biopsy in oncology. Nat Cancer 2020;1:276-90.
crossref pmid pdf
57. Abbosh C, Frankell AM, Harrison T, Kisistok J, Garnett A, Johnson L, et al. Tracking early lung cancer metastatic dissemination in TRACERx using ctDNA. Nature 2023;616:553-62.
pmid pmc
58. Zhang JT, Liu SY, Gao W, Liu SM, Yan HH, Ji L, et al. Longitudinal undetectable molecular residual disease defines potentially cured population in localized non-small cell lung cancer. Cancer Discov 2022;12:1690-701.
crossref pmid pmc pdf
59. Provencio M, Nadal E, Gonzalez-Larriba JL, Martinez-Marti A, Bernabe R, Bosch-Barrera J, et al. Perioperative nivolumab and chemotherapy in stage III non-small-cell lung cancer. N Engl J Med 2023;389:504-13.
pmid
60. Felip E, Srivastava M, Reck M, Wakelee H, Altorki NK, Vallieres E, et al. 1O IMpower010: ctDNA status in patients (pts) with resected NSCLC who received adjuvant chemotherapy (chemo) followed by atezolizumab (atezo) or best supportive care (BSC). Immuno-Oncol Tech 2022;16.
crossref
61. Forde PM, Spicer J, Lu S, Provencio M, Mitsudomi T, Awad MM, et al. Neoadjuvant nivolumab plus chemotherapy in resectable lung cancer. N Engl J Med 2022;386:1973-85.
pmid pmc
62. Jung HA, Ku BM, Kim YJ, Park S, Sun JM, Lee SH, 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;18:1199-208.
crossref pmid
63. Tsuboi M, Herbst RS, John T, Kato T, Majem M, Grohe C, et al. Overall survival with osimertinib in resected EGFR-mutated NSCLC. N Engl J Med 2023;389:137-47.
pmid
64. John T, Grohe C, Goldman JW, Kato T, Laktionov KK, Bonanno L, et al. Molecular residual disease (MRD) analysis from the ADAURA trial of adjuvant (adj) osimertinib in patients (pts) with resected EGFR-mutated (EGFRm) stage IB-IIIA non-small cell lung cancer (NSCLC). J Clin Oncol 2024;42(16 suppl):8005.
crossref
65. Spigel DR, Faivre-Finn C, Gray JE, Vicente D, Planchard D, Paz-Ares L, et al. Five-year survival outcomes from the PACIFIC trial: durvalumab after chemoradiotherapy in stage III non-small-cell lung cancer. J Clin Oncol 2022;40:1301-11.
crossref pmid pmc
66. Moding EJ, Liu Y, Nabet BY, Chabon JJ, Chaudhuri AA, Hui AB, et al. Circulating tumor DNA dynamics predict benefit from consolidation immunotherapy in locally advanced non-small cell lung cancer. Nat Cancer 2020;1:176-83.
crossref pmid pmc pdf
67. Lu S, Kato T, Dong X, Ahn MJ, Quang LV, Soparattanapaisarn N, et al. Osimertinib after chemoradiotherapy in stage III EGFR-mutated NSCLC. N Engl J Med 2024;391:585-97.
pmid
68. National Lung Screening Trial Research Team, Aberle DR, Adams AM, Berg CD, Black WC, Clapp JD, et al. Reduced lung-cancer mortality with low-dose computed tomographic screening. N Engl J Med 2011;365:395-409.
crossref pmid pmc
69. de Koning HJ, van der Aalst CM, de Jong PA, Scholten ET, Nackaerts K, Heuvelmans MA, et al. Reduced lung-cancer mortality with volume CT screening in a randomized trial. N Engl J Med 2020;382:503-13.
pmid
70. Phallen J, Sausen M, Adleff V, Leal A, Hruban C, White J, et al. Direct detection of early-stage cancers using circulating tumor DNA. Sci Transl Med 2017;9:eaan2415.
crossref pmid pmc
71. Klein EA, Richards D, Cohn A, Tummala M, Lapham R, Cosgrove D, et al. Clinical validation of a targeted methylation-based multi-cancer early detection test using an independent validation set. Ann Oncol 2021;32:1167-77.
crossref pmid
72. Cohen JD, Li L, Wang Y, Thoburn C, Afsari B, Danilova L, et al. Detection and localization of surgically resectable cancers with a multi-analyte blood test. Science 2018;359:926-30.
crossref pmid pmc
73. Chabon JJ, Hamilton EG, Kurtz DM, Esfahani MS, Moding EJ, Stehr H, et al. Integrating genomic features for non-invasive early lung cancer detection. Nature 2020;580:245-51.
pmid pmc
74. Su YH, Wang M, Brenner DE, Ng A, Melkonyan H, Umansky S, et al. Human urine contains small, 150 to 250 nucleotide-sized, soluble DNA derived from the circulation and may be useful in the detection of colorectal cancer. J Mol Diagn 2004;6:101-7.
crossref pmid pmc
75. Li F, Huang J, Ji D, Meng Q, Wang C, Chen S, et al. Utility of urinary circulating tumor DNA for EGFR mutation detection in different stages of non-small cell lung cancer patients. Clin Transl Oncol 2017;19:1283-91.
crossref pmid pdf
76. Husain H, Melnikova VO, Kosco K, Woodward B, More S, Pingle SC, et al. Monitoring daily dynamics of early tumor response to targeted therapy by detecting circulating tumor DNA in urine. Clin Cancer Res 2017;23:4716-23.
crossref pmid pmc pdf
77. Wever BM, Bach S, Tibbesma M, Ter Braak TJ, Wajon D, Dickhoff C, et al. Detection of non-metastatic non-small-cell lung cancer in urine by methylation-specific PCR analysis: a feasibility study. Lung Cancer 2022;170:156-64.
crossref pmid
78. Pentsova EI, Shah RH, Tang J, Boire A, You D, Briggs S, et al. Evaluating cancer of the central nervous system through next-generation sequencing of cerebrospinal fluid. J Clin Oncol 2016;34:2404-15.
crossref pmid pmc
79. Wijetunga NA, Goglia AG, Weinhold N, Berger MF, Cislo M, Higginson DS, et al. Dynamic mutational landscape of cerebrospinal fluid circulating tumor DNA and predictors of survival after proton craniospinal irradiation for leptomeningeal metastases. Clin Cancer Res 2023;29:775-83.
crossref pmid pmc pdf
80. Zheng MM, Li YS, Tu HY, Jiang BY, Yang JJ, Zhou Q, et al. Genotyping of cerebrospinal fluid associated with osimertinib response and resistance for leptomeningeal metastases in EGFR-mutated NSCLC. J Thorac Oncol 2021;16:250-8.
crossref pmid
81. Bai K, Chen X, Qi X, Zhang Y, Zou Y, Li J, et al. Cerebrospinal fluid circulating tumour DNA genotyping and survival analysis in lung adenocarcinoma with leptomeningeal metastases. J Neurooncol 2023;165:149-60.
crossref pmid pmc pdf
82. Azad TD, Nanjo S, Jin MC, Chabon JJ, Kurtz DM, Chaudhuri AA, et al. Quantification of cerebrospinal fluid tumor DNA in lung cancer patients with suspected leptomeningeal carcinomatosis. NPJ Precis Oncol 2024;8:121.
crossref pmid pmc pdf
83. Kim IA, Hur JY, Kim HJ, Kim WS, Lee KY. A prospective phase 2 study of expeditious EGFR genotyping and immediate therapeutic initiation through extracellular vesicles (EV)-based bronchoalveolar lavage fluid (BALF) liquid biopsy in advanced NSCLC patients. Transl Lung Cancer Res 2023;12:1425-35.
crossref pmid pmc


ABOUT
ARTICLE & TOPICS
Article category

Browse all articles >

Topics

Browse all articles >

BROWSE ARTICLES
FOR CONTRIBUTORS
Editorial Office
101-605, 58, Banpo-daero, Seocho-gu (Seocho-dong, Seocho Art-Xi), Seoul 06652, Korea
Tel: +82-2-575-3825, +82-2-576-5347    Fax: +82-2-572-6683    E-mail: katrdsubmit@lungkorea.org                

Copyright © 2025 by The Korean Academy of Tuberculosis and Respiratory Diseases. All rights reserved.

Developed in M2PI

Close layer
prev next