Current and future applications of liquid biopsy in non-small-cell lung cancer—a narrative review
Review Article

Current and future applications of liquid biopsy in non-small-cell lung cancer—a narrative review

Bartłomiej Tomasik1#, Marcin Skrzypski1#, Michał Bieńkowski2, Rafał Dziadziuszko1, Jacek Jassem1^

1Department of Oncology and Radiotherapy, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland; 2Department of Pathomorphology, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland

Contributions: (I) Conception and design: B Tomasik, M Bieńkowski, J Jassem; (II) Administrative support: J Jassem, R Dziadziuszko; (III) Provision of study materials or patients: B Tomasik, M Skrzypski, M Bieńkowski; (IV) Collection and assembly of data: B Tomasik, M Skrzypski, M Bieńkowski; (V) Data analysis and interpretation: All authors; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

#These authors contributed equally to this work.

^ORCID: 0000-0002-8875-6747.

Correspondence to: Prof. Jacek Jassem, MD, PhD. Department of Oncology and Radiotherapy, Medical University of Gdańsk, Smoluchowskiego 17 St., 80-214 Gdańsk, Poland. Email: jacek.jassem@gumed.edu.pl.

Background and Objective: Lung cancer remains the leading cause of cancer-related mortality and constitutes a significant societal burden. Recent advancements in targeted therapies and immunotherapy have considerably broadened therapeutic options in lung cancer, particularly in non-small-cell lung cancer (NSCLC). However, these novel methods necessitate sophisticated molecular diagnostics. Liquid biopsy, which refers to the cytological and molecular analysis of cancer markers shed by the tumor into the body fluids, may offer an attractive diagnostic tool at the individual patient level. This approach is particularly relevant for lung cancer, as the anatomical location of tumor lesions frequently makes them inaccessible for tissue biopsy. Apart from minimal invasiveness, the major advantages of liquid biopsy include better reflection of the tumor clonal heterogeneity (spatial heterogeneity), the possibility of sequential sampling, and real-time monitoring of tumor load and its evolving mutational status (temporal heterogeneity).

Methods: This article reviews the available data in this field, current applications, and future perspectives in accordance with the Narrative Review reporting rules.

Key Content and Findings: We discuss the most used approaches, i.e., circulating DNA and tumor cells, but also emerging liquid biopsy techniques, such as plasma DNA methylation, plasma metabolites and RNA, extracellular vesicles, and tumor-educated platelets in NSCLC. Finally, we highlight the current limitations of liquid biopsy techniques hampering their clinical applications.

Conclusions: Due to their advantages, liquid biopsy-based approaches have recently gained immense interest in oncology. Potential applications of this method include early detection, informing precision medicine-based individualized treatment, and real-time monitoring of disease evolution and treatment. The development of next-generation sequencing has vastly extended genetic profiling, thus enabling better identification of druggable alterations. However, the clinical application of liquid biopsy techniques is still limited due to their suboptimal specificity and sensitivity, lack of standardization, and relatively high costs. Addressing these issues may allow further integration of liquid biopsies in the routine clinical setting, thus making a profound and permanent change in NSCLC management.

Keywords: Circulating tumor DNA (ctDNA); circulating tumor cells (CTC); liquid biopsy; non-small-cell lung cancer (NSCLC)


Submitted Oct 12, 2022. Accepted for publication Feb 17, 2023. Published online Mar 09, 2023.

doi: 10.21037/tlcr-22-742


Introduction

With an estimated 1.8 million deaths accounting for 18% of global cancer mortality, lung cancer remains the leading cause of cancer death (1). During the past two decades, the application of targeted therapies and, more recently, immune checkpoint inhibitors (ICIs) has resulted in a spectacular improvement in treatment efficacy in lung cancer. However, this progress mainly refers to non-small-cell lung cancer (NSCLC). Modern management of NSCLC using precision medicine methods necessitates sophisticated molecular diagnostics. Genetic testing is currently a routine procedure for many therapeutic targets, such as activating mutations in epidermal growth factor receptor (EGFR), HER2, BRAF, KRAS, MET exon 14 skipping; rearrangements of anaplastic lymphoma kinase (ALK), NTRK, RET, and ROS proto-oncogene1 (ROS1) (2,3). New-generation assays also allow quantification of tumor mutational burden (TMB), which is associated with response to ICIs (4,5).

NSCLC presents a large intratumor heterogeneity and genomic instability. Acquiring molecular changes and treatment-induced clonal selection leads to genetic evolution and resistance to systemic therapies (6-8).

Tissue biopsy remains the gold standard in pathological NSCLC diagnosis, tissue genotyping, and informing treatment (9-11). However, the limitation of this approach lies in its inability to address the clonal heterogeneity of this malignancy (11). More importantly, the increasing number of treatment-guiding biomarkers usually necessitates obtaining many tumor biopsies and repeated sampling to tailor subsequent treatments. This is often not feasible or risky due to the anatomical location of the primary or metastatic lesions or the worsening general condition of the patient (12).

Liquid biopsy, which refers to the cytological and molecular analysis of cancer markers shed by the tumor into the body fluids, is a minimally invasive and easily repeatable test. The most common source for liquid biopsy is blood, as it provides the largest array of biological analytes, such as circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), tumor-educated platelets (TEPs), circulating exosomes, DNA methylation, and metabolomic and proteomic markers (Figure 1). In addition to convenience, ease of access and minimal invasiveness, the major advantages of liquid biopsy include better reflection of the tumor clonal heterogeneity (spatial heterogeneity), the possibility of sequential sampling, and real-time monitoring of tumor load and its evolving mutational status (temporal heterogeneity). Supplementing the standard tissue evaluation with liquid biopsy was also shown to save system costs and increase the number of patients administered appropriate targeted therapies (13-15). Finally, a liquid biopsy may significantly shorten turnaround times at primary diagnosis and at progression (16-18).

Figure 1 Liquid biopsy materials in NSCLC. Tumor derivates include circulating tumor DNA, circulating tumor cells, extracellular vesicles, and tumor-educated platelets. NSCLC, non-small-cell lung cancer.

The first clinical application of liquid biopsy techniques in NSCLC included testing for EGFR mutational status (19,20). Within the past decade, liquid biopsy-based approaches have gained immense interest in oncology, manifested by thousands of publications. A milestone in their widespread use was the development of high-throughput sequencing methods, in particular next-generation sequencing (NGS). This method has considerably broadened genetic profiling, thereby allowing for better identification of druggable alterations and their evolution throughout the disease course (21).

Potential clinical applications of liquid biopsy in NSCLC include early detection, informing precision medicine-based individualized treatment, and real-time monitoring of disease evolution and treatment (21).

Thus far, the only circulating NSCLC biomarker in routine clinical use is ctDNA, which was approved in 2016 for selecting patients for targeted therapies (21,22). Other liquid biopsy sources are in various phases of clinical development. This review summarizes current knowledge on liquid biopsy in NSCLC and its clinical applications, with a particular focus on emerging techniques. We present the following article in accordance with the Narrative Review reporting checklist (available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-22-742/rc).


Methods

In this review, PubMed/MEDLINE, Embase and Google Scholar databases were searched between the date of database inception and December 30, 2022. Only articles in English were evaluated. Details are listed in Table 1. The figures were created using BioRender.com.

Table 1

The search strategy summary

Items Specification
Date of search Last update 01/01/2023
Databases and other sources searched PubMed/MEDLINE, Embase, Google Scholar
Search terms used “non-small-cell lung cancer”, “liquid biopsy”, “circulating DNA” and “circulating tumour cells”
Timeframe Databases were searched between the date of database inception and December 30, 2022
Inclusion and exclusion criteria Only articles in English were evaluated, only full texts were evaluated
Selection process Each step of the study was conducted by two independent researchers (BT and MB) with disputes resolved by discussion

ctDNA

In 1948, Mandel and Metais first described the presence of short DNA fragments (termed cell-free DNA, cfDNA) in the blood (23). In healthy individuals, they mainly originate from the hematopoietic cells and reflect their clonal heterogeneity (24,25). In 1977, Leon et al. reported an increase in cfDNA content in cancer patients (26), which was later attributed to neoplastic cell death (27). However, the interest in ctDNA remained modest until it boomed following the development of PCR-based techniques. Importantly, ctDNA has a very short half-life, thus making it an ideal biomarker for disease monitoring (28,29).

The pre-analytical factors that may influence the accuracy of ctDNA evaluation remain poorly described. One aspect of the general consensus is the use of plasma over serum samples to prevent unnecessary leukocyte lysis and, thus, genomic DNA contamination (30,31). Among the numerous available cfDNA extraction methods, neither has been proven superior with significant interlaboratory variability (32,33). In addition, the timing of sample collection is crucial, as numerous factors (including physical exercise, inflammatory conditions, concurrent treatment, and time of the day) may affect cfDNA levels (11,34). Therefore, no unanimously agreed protocol for sample collection has not yet been defined, and procedural uniformity within a study becomes vital (11). Another important aspect of ctDNA testing is clonal hematopoiesis of indeterminate potential (CHIP), i.e., somatic gene variants, common in aging human hematopoietic stem cells. CHIP is generally defined by any variant allele frequency (VAF) of at least 2% (35). Some of these variants are relatively frequent in solid malignancies. As these variants are also detectable in cfDNA, they are a relevant source of biological noise in liquid profiling. Hence, to avoid false positive results, it is advisable to couple cfDNA testing with whole blood sequencing as a control (36). This might however impact time- and cost-effectiveness. Hence, novel approaches involving machine learning are currently under investigation (37).

It is also essential to distinguish between tumor-informed and tumor-agnostic strategies. The former involves tumor specimen sequencing to create a tumor-specific signature. Identified aberrations may then be tracked using polymerase chain reaction (PCR)-based techniques or NGS panels. Focusing on already-known alterations allows for deeper coverage, making these assays more sensitive. A tumor-informed probe serving as an internal control also increases specificity by reducing background noise from not tumor-derived clonal populations, e.g., CHIP. However, this approach has a long turnaround time and requires high tissue quality. In turn, a tumor-agnostic strategy works across various cancer types sharing the same targetable abnormality. This approach is faster, cheaper, and does not necessitate sequencing the primary tumor, but at a price of lower sensitivity (38).

In metastatic NSCLC, ctDNA testing already plays a vital role in clinical practice, allowing the detection of tumor-derived somatic aberrations from plasma to inform treatment decisions (39). Although tissue is still the gold standard for the initial NSCLC genotyping, complementary ctDNA testing may provide valuable insights into possible targets missed by tissue biopsy. Liquid biopsy may have particular value for patients with a discordant clinical history or a high probability of intratumor heterogeneity. Complementary ctDNA testing may also be helpful when tumor biopsy is difficult, or the sample amount is insufficient (11,40). This approach is now endorsed by the European Medicines Agency (EMA), the International Association for the Study of Lung Cancer, and the National Comprehensive Cancer Network (11,39,41,42). Plasma and tissue genotyping seem complementary, with 20% of targetable variants detected in blood but not in tissue (43). In addition, plasma-based assays allow for a shorter turnaround time (44).

In 2020, the US Food and Drug Administration (FDA) and EMA approved the use of two ctDNA assays, Guardant360 and FoundationOne Liquid CDx, to identify genomic alterations in patients with advanced-stage solid malignancies (45,46). Both are hybrid capture-based NGS assays that detect variants in 73 and 311 genes, respectively (47,48). Detecting EGFR variants in ctDNA from peripheral blood is already widely used (Figure 2). Two platforms, the cobas® EGFR mutation test v2 (Roche, Basel, Switzerland) and the TheraScreen EGFR RGQ PCR Kit (Qiagen, Hilden, Germany), have been approved by both the EMA and FDA for the testing of EGFR variants in liquid biopsies (49). Many studies have shown high concordance in EGFR status between ctDNA and tissue biopsies (50,51). The role of liquid biopsy is particularly compelling at disease progression, as it may allow tracking mechanisms of resistance. For example, in EGFR-mutated NSCLC, approximately 30% of T790M-positive tissues are missed by plasma, and a similar proportion of cfDNA T790M-positive results would test negative in tissues, making these two approaches complementary (52). This is of particular importance because the clinical gain from osimertinib, a third-generation EGFR tyrosine kinase inhibitor, has been demonstrated in both plasma-positive and tissue-positive groups (52). The feasibility of using ctDNA as a surrogate for tumor biopsy was shown in the EURTAC trial, which provided evidence that progression-free survival (PFS), overall survival (OS), and response to therapy are associated with the type of EGFR variants in ctDNA (53). Unlike single variants, molecular aberrations, such as fusions, copy number variations, and some focal amplifications, appear more difficult to detect in blood than in tissue (54). Nevertheless, genomic profiling of ctDNA has also been demonstrated to be a reliable and useful method for genetic profiling and monitoring tumor evolution in ALK-rearranged NSCLCs (55-59). Two ctDNA assays, FoundationOne® Liquid CDx and Guardant360® CDx (Guardant Health), have been approved by the FDA for testing this aberration and found clinical applications (40).

Figure 2 Overview of current and potential applications of liquid biopsy in non-small-cell lung cancer. EGFR, epidermal growth factor receptor; EML4, echinoderm microtubule-associated protein-like 4; ALK, anaplastic lymphoma kinase.

High levels of ctDNA are also linked to worse survival; hence, ctDNA can contribute to estimating prognosis (Figure 2) (60). In addition, the persistence of ctDNA after surgical treatment of early-stage NSCLC may be indicative of disease recurrence and can identify patients for further intervention (61). ctDNA detection in advanced NSCLC is reasonably reliable, although sensitivity in early-stage tumors remains low, usually not exceeding 50% (62,63). Nevertheless, plasma ctDNA could probably be a valuable adjunct to other screening methods, such as low-dose computed tomography (LDCT), and is an approach under evaluation in the ongoing SUMMIT study (NCT03934866).

While approved assays can be applied to plasma samples from patients with advanced NSCLC, they do not perform very well for minimal residual disease (MRD) monitoring. With increasing attention and progress toward the earlier detection of solid tumors, the relevance of detecting MRD and early recurrence may transform the optimal treatment paradigm through more timely interventions (Figures 2,3). The concept of MRD has been introduced in hematology, where the presence of residual malignant cells following treatment was shown to be associated with poorer prognosis and earlier relapse (64). Nowadays, MRD-guided therapy has become widely introduced in hematology, and MRD itself is used as a surrogate efficacy-response biomarker to accelerate drug development (65). This approach has also been successfully tested in stage II colon cancer, where the evaluation of ctDNA after surgery led to reduced adjuvant chemotherapy use without compromising recurrence-free survival (66). Similar to MRD testing in hematology, ctDNA may have utility for residual disease detection following curative-intent treatment and during the surveillance period for relapse (Table 2). Although commercially available assays have not yet achieved regulatory approval, several trials evaluating the role of ctDNA in the personalized treatment of patients with NSCLC are ongoing (Table 2).

Figure 3 Hypothetical disease control available when guiding treatment using the following approaches: (A) physical examination, (B) imaging, (C) liquid biopsy-based assays, and (D) liquid biopsy-based screening for minimal residual disease after curative local treatment.

Table 2

Studies investigating the role of ctDNA in MRD evaluation and surveillance in NSCLC patients

Study N Stage Treatment Sensitivity, % Specificity, % Assay
ctDNA performance to detect MRD
   Chaudhuri et al., 2017 (67) 32 IB–IIIB CRT or RT and/or surgery ± CHT 94 100 CAPP-Seq
   Abbosh et al., 2017 (68) 24 IA–IIIB Surgery ± CHT ± PORT 36 90 Signatera
   Chen et al., 2019 (69) 25 IIB–IIIB Surgery ± CHT ± PORT 44 88 cSMART
   Zviran et al., 2020 (70) 22 IA–III Surgery ± CHT and RT 100 71 MRDetect
ctDNA performance to detect disease in the surveillance setting
   Chaudhuri et al., 2017 (67) 37 IB–IIIB CRT or RT and/or surgery ± CHT 100 100 CAPP-Seq
   Abbosh et al., 2017 (68) 24 IA–IIIB Surgery ± CHT 93 70 Signatera
   Abbosh et al., 2020 (71) 78 I–III Surgery ± CHT 82 96 ArcherDx

ctDNA, circulating tumor DNA; MRD, minimal residual disease; NSCLC, non-small-cell lung cancer; CRT, chemoradiotherapy; RT, radiotherapy; CHT, chemotherapy; PORT, postoperative radiotherapy.

As ICIs become increasingly common in the treatment of NSCLC, the potential of ctDNA to guide and follow the response to immune therapy is also being evaluated (Figure 2) (72). Good concordance of ctDNA NGS with tissue NGS enables the estimation of TMB (73). A high blood-based TMB (bTMB) was associated with improved response to ICIs in the POPLAR and OAK trials (73). However, the test outcome is highly dependent on the gene panel (73). Recently, B-F1RST, an open-label, phase 2 trial that evaluated bTMB as a predictive biomarker for first-line atezolizumab monotherapy in locally advanced or metastatic stage IIIB–IVB NSCLC, showed that bTMB ≥16 was associated with longer OS (74). Additionally, the overall response rate (ORR) improved with increasing bTMB cutoffs.

Although higher TMB is generally associated with clinical benefit from ICIs, the clinically relevant bTMB cutoff values are lacking. Clinical trials have measured TMB using Whole Exome Sequencing (WES) or the FoundationOne CDx assay. Later on, several targeted panels appeared on the market, providing TMB estimates in a time- and cost-effective manner (75). A recent initiative of the Friends of Cancer Research TMB Harmonization Project showed substantial variability between assays. The same group developed and made publicly available a software tool that could promote reproducibility and comparability across various assays (76).

ctDNA has also been proposed as an ICI monitoring tool to discriminate between pseudo-progression and true progression (77). Rapid decreases in ctDNA levels after initiating first-line pembrolizumab with or without chemotherapy were associated with significantly higher response rates and longer PFS and OS (78).

The optimal post-treatment time point for ctDNA MRD assessment remains unclear. Addressing this question is of utmost importance and requires initiatives to harmonize clinical trials measuring ctDNA. An example of such an initiative is the ctMoniTR harmonization effort of post-ICI ctDNA data in advanced NSCLC. The evidence from prospective studies is currently scarce [NILE trial, NCT03615443 (44)], and most current data on the clinical utility of plasma ctDNA genotyping come from retrospective observational studies (79). Also, it is still unknown whether an increase in plasma ctDNA levels alone should lead to treatment modification. The APPLE-EORTC trial, a randomized, open-label, multicenter, 3-arm, phase II study of advanced EGFR-positive NSCLC, addressed this issue (80,81). A recent study showed that serial monitoring of ctDNA T790M status by Cobas v2.0 PCR test in patients with advanced EGFR mutant-NSCLC treated with first-generation EGFR inhibitors is feasible and allows for an earlier switch to second-line EGFR inhibition (81). However, the study evaluated only T790M-driven acquired resistance; therefore, extrapolation of its results to other clinical scenarios is disputable. More studies with appropriate sample size and multiple post-treatment time points are needed to improve the understanding of ctDNA MRD and its predictive role in NSCLC. A summary of major ongoing trials is presented in Table 3.

Table 3

Ongoing clinical trials investigating the role of ctDNA in personalized treatment of NSCLC

Trial acronym/NCT # Phase NSCLC stage Primary endpoint Primary treatment Moment of ctDNA measurement ctDNA (+) intervention ctDNA (−) intervention Planned enrollment Recruitment status Type of assay
MERMAID-1 (NCT04385368) III II–III DFS in MRD + analysis set Surgery After surgery Durvalumab + SoC CHT vs. placebo + SoC CHT N/A 332 Recruiting ArcherDx
MERMAID-2 (NCT04642469) III II–III DFS in the PD-L1 TC ≥1% analysis set Surgery +/− neoadjuvant or adjuvant treatment Surgery +/− neoadjuvant or adjuvant treatment Durvalumab vs. placebo N/A 284 Active, not recruiting ArcherDx
NCT04585490 III III Change in ctDNA level following CHT CRT After CRT 4 cycles of CHT (platinum-based doublet) + durvalumab (1,500 mg IV every 21 days for 1 year) SoC durvalumab (10 mg/kg every 2 weeks, or equivalent, for 1 year) 48 Recruiting Avenio
NCT04585477 II I–III Decrease in ctDNA level Surgery or definitive SBRT After surgery or SBRT 12 cycles of durvalumab SoC and no treatment 80 Recruiting Avenio
SCION (NCT04944173) II I–IIA ORR at 18 months SBRT + 4 cycles of durvalumab After SBRT + 4 cycles durvalumab Additional 8 cycles of durvalumab No further treatment 94 Not yet recruiting Avenio

ctDNA, circulating tumor DNA; NSCLC, non-small-cell lung cancer; DFS, disease-free survival; MRD, minimal residual disease; SoC, standard of care; CHT, chemotherapy; PD-L1, programmed death ligand 1; TC, tumor cell; N/A, not applicable; CRT, chemoradiotherapy; IV, intravenous; SBRT, stereotactic body radiotherapy; ORR, overall response rate.


CTCs

CTCs play an important role in metastasis formation and represent an intermediate stage of this process (82). Despite their rare occurrence (approximately 1–10 per 10 mL of blood), they are accessible through a simple and non-invasive sampling of body fluids. Only approximately 0.02% of CTCs can survive the adverse environment, which includes continuous mobility, immune attack, mechanical shear forces, and oxidative stress (83). CTCs may occur as single cells, clusters of several cells, or circulating tumor microemboli consisting of huge numbers of cells aggregated together with other cells, including hematopoietic cells, platelets, and stromal cells (84). The clustered forms are more stable in the bloodstream and have a greater metastatic capacity than single CTCs (85). The most analyzed source of CTCs is pulmonary vein blood because their counts are higher there than in peripheral vessels (86,87). Numerous methods of CTC detection can include those based on microfluidics and the physical properties of the cells, in particular their size, the functional or immune assays. The most frequently applied approaches usually employ epithelial cell enrichment (with EpCAM antibodies) (88), leukocyte depletion (with CD45 antibodies) (89), or both (90). In contrast to ctDNA, CTC analysis necessitates sample collection and processing specifically matched to the isolation method and downstream applications. Major characteristics and the comparison between ctDNA/cfDNA and CTCs are presented in Table 4.

Table 4

Characteristics and comparison of ctDNA/cfDNA and CTCs

Feature ctDNA/cfDNA CTCs
Collection Easier More complex
Isolation Easier More complex
Cell culture Impossible Possible
Therapy monitoring Changes in levels may reflect response/resistance/relapse
Genomic analyses Possible Possible
Transcriptomic analyses Impossible Possible
Protein analyses Impossible Possible
Functional analyses Impossible Possible
Methylation analyses Possible Possible
FISH analyses Impossible Possible
Single-cell analyses Impossible Possible
Chromosomal analyses Impossible Possible
Challenges Potential affecting by treatment- or stress-induced cell death CTC heterogeneity
Potential confounding by CHIP
Small levels or quantities in circulation
Sampling bias
Low signal-to-noise ratio in early-stage disease
Cost Several hundred to several thousand USD (depending on the panel used) Several hundred dollars to one thousand USD (CellSearch®)

ctDNA, circulating tumor DNA; cfDNA, cell-free DNA; CTCs, circulating tumor cells; FISH, fluorescent in situ hybridization; CHIP, clonal hematopoiesis of indeterminate potential; USD, United States dollar.

CTCs as a screening tool

Blood-based biomarkers may act as a standalone screening tool (Figure 2) or in addition to other screening methods, such as LDCT in NSCLC (91). In 2006, a study using a 17-gene array found CTCs in 90% of NSCLC patients at different disease stages and in 6% of healthy controls (92). However, the performance of this tool was associated with cancer stage and was the lowest in stage I disease. Other studies using the folate receptor transcript as a marker to identify CTCs have shown promising sensitivity and specificity (93,94). However, this approach was also hampered by lower performance in the early stages of NSCLC. A study including patients with chronic obstructive pulmonary disease (COPD), which often coexists with NSCLC, identified CTCs in 3% (5 out of 168 patients) of the group (95). During follow-up with annual CT imaging, lung nodules were found one to four years after CTC detection, and CTCs were undetectable after the surgery. Another study from China showed the in vivo CTC detection strategy to be characterized by 53% sensitivity and 90% specificity for diagnosing early-stage NSCLC (96). However, since the evaluated cohort was small, the study was underpowered and inconclusive. On the other hand, a prospective study done on 614 COPD patients treated in 21 French university centers showed only 26% sensitivity of CTC detection for NSCLC, bringing into question the utility of this method in NSCLC screening (97).

CTCs for the evaluation of cancer prognosis

The prognostic value of CTCs in NSCLC has been extensively studied. As mentioned earlier, the number of CTCs generally correlates with the NSCLC burden, and the detection rate varies greatly from 15% to 100% (98-110). Hence, the utility of CTC assessment is highest in more advanced tumors. A decrease in CTC counts after treatment may indicate cancer remission, while an increase may herald cancer progression. Changing CTC counts have been associated with shorter disease-free survival (DFS), PFS, and OS; however, the optimal threshold of detectable CTCs remains to be determined (111-117).

A prospective study including stage IV NSCLC identified CTCs in most subjects (118). The absolute CTC counts were not associated with the prognosis, whereas changes in CTC counts were predictive of OS. The largest study of CTCs in advanced NSCLC included 550 patients through multicenter European collaboration (119). This study confirmed the independent prognostic value of CTCs for PFS and OS. The detection of CTCs was associated with worse PFS (≥2 CTCs: HR =1.72, P<0.001; ≥5 CTCs: HR =2.21, P<0.001) and OS (≥2 CTCs: HR =2.18, P<0.001; ≥5 CTCs: HR =2.75, P<0.001). Importantly, CTC counts added to clinicopathological predictive models significantly improved their performance. In addition, the presence of circulating tumor microemboli coupled with clinical and imaging data significantly improved diagnostic accuracy in early-stage NSCLC (120). Similar findings were reported in small-cell lung cancer (SCLC). The analysis of CTCs collected from patients enrolled within CONVERT trial showed that the threshold ≥15 CTCs was associated with worse OS independent of all other factors (26.7 months in the group <15 CTCs and 5.9 months in the group ≥15 CTC) (121).

CTCs for guiding treatment

A study using telomerase-based CTC assay in patients with early-stage NSCLC managed with stereotactic body radiotherapy (SBRT) showed that higher pretreatment CTCs (≥5 cells/mL) and persistent CTCs after SBRT were associated with increased risk of regional and distant recurrence (122). This CTC assay may identify subsets of patients who can maximally benefit from adjuvant systemic therapy and enables early detection of recurrence or progression.

CTCs may also be useful in monitoring patients harboring oncogenic driver variants, such as EGFR-activating mutations or ALK fusion rearrangements. In 2008, Maheswaran et al. reported the detection of EGFR mutations in CTCs isolated from 27 NSCLC patients (123). The clinical utility of single CTC sequencing in ALK-rearranged NSCLCs was demonstrated by Pailler et al. (124). The receptor tyrosine kinase-KRAS pathway (EGFR, KRAS, BRAF genes) and TP53 pathways were recurrently mutated in the CTCs of crizotinib-resistant patients. Another study from this group identified ROS1 rearrangements in the CTCs of four NSCLC patients (125). Gorges et al. identified KRAS variants in CTCs that are potentially relevant to treatment decisions (126).

The role of CTCs has also been evaluated in the real-time monitoring of immune cell activation. The presence of PD-L1(+) CTCs was associated with poor prognosis in patients with advanced NSCLC (127). However, the data regarding concordance between tumor tissue and CTCs are conflicting (127,128). Most of these studies were small, making their results inconclusive.

Although monitoring for CTCs may provide predictive information, the lack of a standardized methodology for CTC enrichment and detection has hampered its uptake by the clinical community. Ongoing initiatives within the framework of collaborative groups, such as CANCER-ID, a European consortium of 38 partners from 13 countries, may better define the clinical utility of liquid biopsies (129).


Emerging technologies and strategies for liquid biopsy

Given the suboptimal sensitivity of DNA variants detection in cfDNA for the early diagnosis of NSCLC (130), new liquid biopsy methods, e.g., DNA methylation, metabolomic, proteomic or RNA markers, or new marker bio-reservoirs, such as tumor educated platelets (TEPs), are being explored. Plasma metabolites might be particularly useful as companion diagnostics for drugs targeting cancer metabolic vulnerabilities. TEPs are thought to provide a unique pool of markers and have the potential for early cancer detection and monitoring after treatment.

Plasma DNA methylation

The low performance of plasma cfDNA for mutation-based cancer detection is due to the limited number of specific cancer variants, affecting only a handful of genomic locations. Moreover, the amount of ctDNA is relatively low compared with the non-tumor cfDNA, resulting in a low signal-to-noise ratio. These problems are further confounded by the variants arising concomitantly in hematopoietic cells, namely aging-related clonal hematopoiesis variants that may mimic cancer-associated mutations (25,131). In contrast, the cfDNA methylation spans nearly 30 million sites known as CpG sites across the human genome, which enables the specific selection of CpG sites not shared by other cancer locations (132). Cancer-specific methylation patterns have been established and can be inferred from plasma DNA with the advance of bioinformatics methods (133). The large-scale comparisons between cfDNA sequencing and cfDNA whole genome bisulfite sequencing have shown the superior sensitivity of cancer detection of the latter (134).

The recently developed Galleri targeted-methylation multi-cancer early detection (MCED) test showed an overall sensitivity of 52% for detecting malignancy across more than 50 cancer types (135). At 99.5% specificity, the sensitivity for stage I–III cancer detection was 68% in 12 pre-specified cancers and 75% in lung cancer. However, the sensitivity of stage I NSCLC detection was only 22%, compared with 80% and 91% in stages II and III, respectively. The overall accuracy of the primary tumor site determination in true positives was 89%, which is suitable for informing care after the positive screening test result. This test was assessed prospectively in an epidemiological interventional Pathfinder study (136). Healthy individuals with positive test underwent test-directed diagnostic procedures toward a cancer diagnosis. In this single-arm study, the primary endpoints included the time required to establish clinical cancer diagnosis following a positive MCED blood test and the number and types of diagnostic tests used. The test detected a cancer signal in 1.4% of 6,621 individuals 50 years or older not known to have cancer. Cancer was confirmed in 38% and 43% of those with a positive test using earlier and refined versions, respectively. The median time to confirm or exclude a tumor was 79 days among participants with a positive screening test, and 73% of subjects obtained the diagnostic resolution within three months. Overall, adding MCED testing to the standard screening doubled the cancer detection rates. The Galleri test is currently being assessed in England in a prospective trial of 140,000 participants (137).

Recently, genome-wide cfDNA methylation profiling was found to provide specific patterns for SCLC (138). Additionally, the levels of tumor methylation detected in cfDNA correlated with OS. SCLC comprises several molecular subtypes with differential therapeutic vulnerabilities (139). For example, ASCL1 drives a phenotype susceptible to BCL2 apoptosis regulator and δ-like canonical Notch ligand 3 inhibitors, whereas the NEUROD1-driven subtype is sensitive to Aurora kinase inhibitors (140,141). The cfDNA SCLC-methylation patterns may differentiate between these molecular subtypes, potentially allowing for monitoring the dynamics of the molecular make-up along the disease progression (138).

Plasma metabolites

In multiple cancer types, the cell metabolism is altered to initiate or promote cancerogenesis or support the demands of high proliferation (142). Specific cancer phenotypes characterized by persistent dysregulation of metabolic pathways could be leveraged for cancer diagnosis with liquid biopsy (141). Sensitive detection techniques, such as liquid chromatography-mass spectroscopy (LC-MS), have enabled the interrogation of plasma for a vast array of metabolites (142). Moreover, metabolite addiction typical of certain cancers may result in metabolic vulnerabilities constituting potential targets for therapeutic interventions (143,144). For example, NSCLC with mutational activation of Nrf2, an antioxidant and detoxification transcription factor, becomes dependent on glutamine, which could be exploited therapeutically using glutaminase inhibitors or G6PD inhibitors (145,146). Thus, the plasma metabolites may be considered to be potential predictive markers of response to metabolically targeted anticancer therapies.

In a study of 25 early-stage NSCLC patients, the major serum metabolic alterations included increased levels of ketone bodies and lactate and decreased levels of glucose, lipids, choline phospholipid metabolites, TMAO, and betaine, compared with matched healthy controls (59). The levels of glutamine, glutamate, asparagine, aspartate, tyrosine histidine, cysteine, isoleucine, and leucine were increased in the serum of cancer patients, whereas the levels of tryptophan and methionine were reduced. In another study of 110 NSCLC patients and 43 healthy controls, targeted metabolomic analysis with LC-MS, a specific combination of six metabolic biomarkers enabled discrimination between stage I NSCLC patients and healthy individuals with 98% sensitivity and 100% specificity. An exhaustive summary of studies regarding NSCLC was presented earlier (147).

Cancers of different lineages may utilize some metabolites differently. For example, KRAS activation and Trp53 inactivation result in formation of pancreatic ductal adenocarcinoma and NSCLC. However, despite sharing the same initiating events, the plasma branched-chain amino acid levels are elevated in pancreatic cancer and normal in NSCLC, as its growth relies on branched-chain amino acid metabolism (148). The analysis also revealed distinct metabolic signatures for lung adenocarcinoma and squamous cell carcinoma (149).

Glioma and acute myeloid leukemia cells with IDH1 mutation constitutively produce an oncometabolite D-2-hydroxyglutarate (D-2-HG) with the diminished production of its normal IDH1 product, i.e., α-ketoglutarate (αKG) (150-153). The decreased amounts of αKG in cancer cells release hypoxia-inducible factor 1 (HIF-1), which leads to widespread pro-oncogenic consequences and contributes to the progression of these malignancies. Mutations in the IDH-1 and IDH-2 genes occur in majority of malignant gliomas (60–90%), 10–20% of acute myeloid leukemias, and up to approximately 1% of lung adenocarcinomas (154,155). Significantly elevated levels of D-2-HG in cells, tissues, plasma, and urine from cancers with somatic variants in IDH may indicate the presence of respective malignancies. This oncometabolite is present in negligible amounts in non-IDH mutant cells (156). The tissue levels of D-2-HG were reported to be increased in lung adenocarcinoma compared with normal lung parenchyma (157).

Plasma metabolomics offers the potential for developing clinically relevant liquid-biopsy solutions for cancer detection and prediction of therapy benefit. However, NSCLCs display intratumor metabolic heterogeneity in nutrient utilization, which may be challenging for the development of markers and therapies (158). The plasma metabolomics markers need large-scale validations before their clinical utility is ascertained.

Another potential source of material from body fluids in NSCLC is urine. LC-MS analysis of urine samples collected from patients with NSCLC showed increased levels of certain amino acids, including tyrosine, tryptophan, and phenylalanine (159). Modified nucleosides, regarded as indicators for the whole-body turnover of RNAs, are excreted in abnormal amounts in the urine of patients with various malignancies, including NSCLC (160).

TEPs

TEPs can sequester approximately 5500 RNA biomarkers and are considered a promising biosource for cancer detection (161,162). The mechanism for platelet education in tumors remains largely elusive. Platelet pre-mRNA in cancer patients can undergo premature splicing and translation, which results in their activation, likely enhancing the thrombo-embolic state. Platelets alter their RNA content upon cancer-associated cues, however, overall, the RNA transcripts enriched in TEPs are ontologically associated with platelet activity and platelet vesicles. Platelet RNA profiles discriminate between patients with localized and metastatic disease and the healthy individuals with an accuracy of 84–96%. Notably, this seems possible with only minute amounts of platelet RNA (100–500 picograms) extracted from routinely used volumes of blood samples. Machine learning–based classification algorithms were found to predict the site of the primary tumor with 71% accuracy, which may potentially define the molecular subtype of NSCLC (163). In a follow-up study, the accuracy of TEP-based detection of early- and advanced-stage NSCLC was 81% and 88%, respectively, independent of patient age, smoking habits, whole-blood storage time, and various inflammatory conditions (163). However, further large-scale validation showed lower sensitivity of TEP-based assays in NSCLC (50%, 70%, 63%, and 77% for stages I, II, III, and IV, respectively), at the 99% specificity (161). However, the assay specificity dropped to an average of 78% if the controls included individuals with symptomatic inflammatory and cardiovascular diseases and benign tumors. This limits the potential application of current pan-cancer TEPs-based tests to only asymptomatic individuals. Hence, the TEP-based algorithms still require refinement.

TEPs have also been shown to sequester tumor-derived EML4–ALK fusion transcripts that displayed lowered titer upon successful crizotinib therapy in a patient with NSCLC (162). The platelet lifespan is around 7 to 10 days, and tumor-derived transcript can accumulate in the TEPs and be protected from plasma RNases. Therefore, TEP RNA analysis may enable higher sensitivity of detection and, thus, more accurate monitoring of response to treatment.

The performance of TEPs-based detection of cancer is somewhat lower than ctDNA/protein-based tests (69–98% sensitivity at 99% specificity for Cancer Seek and 80–85% sensitivity at 99% specificity for Cancer Radar) (161,163-165). The clinical usefulness of TEPs as a biomarker warrant further investigation.

Extracellular membrane vesicles (ExCeMVs)

ExCeMVs comprise a pool of small vesicles released by cells as a part of normal or pathological cell processes. These vesicles include apoptotic and necrotic bodies and exosomes (166). Exosomes, ranging from 40 to 160 nanometers, are actively released from living cells (167). ExCeMVs contain various molecules, such as lipids, proteins, and RNA, and are considered a mode of intercellular communication, contributing to a wide range of biological processes, including cancer (167,168). Alterations in exosomal cargo content can serve as diagnostic and prognostic biomarkers. As an example, a panel of three exosomal proteins (CD151, CD171, and tetraspanin 8) has been shown to be a promising diagnostic marker in NSCLC (169), whereas elevated levels of exosomal membrane–bound protein NY-ESO-1 have been associated with poor prognosis (165). Interestingly, exosomal content may also prompt metastasis formation. Amphiregulin carried by exosomes derived from NSCLC may induce EGFR pathway activation in pre-osteoclasts, which results in the expression of receptor-activator-of-nuclear-factor-kappa-B-ligand (RANKL). RANKL, in turn, increases the expression levels of several proteolytic enzymes, triggering a vicious circle in osteolytic bone metastases. Expression levels of exosomal ZEB1 are associated with significantly higher resistance to cisplatin and gemcitabine and may inform treatment decisions (170). Interestingly, exosomes from T790M-positive cells can induce resistance to gefitinib in sensitive cells via activation of the PI3K/AKT signaling pathway (171), whereas exosomal transfer of wild-type EGFR may confer osimertinib resistance (172). In turn, exosomal transfer of miR-7 may restore gefitinib sensitivity in previously resistant cells (173).

Importantly, interrogation of exosome RNA/DNA content was shown to improve the detection of T790M resistance mutations to the first and second generation of EGFR TKIs. For example, the sensitivity and specificity of the exosome-based T790M mutation detection combined with ctDNA detection were 92% and 89%, respectively, with ExoDx Lung test, compared to 58% and 80%, respectively, with the FDA-approved cfDNA test (cobas EGFR Mutation Test v2, Roche) (174). Interestingly, the exosome-based test showed an unparalleled performance for the detection of T790M resistance mutation in the case of intrathoracic disease that is difficult to detect by liquid biopsy, with sensitivity and specificity of 88% and 94% for disease stages M0/M1a and M1b, respectively. Such combined exosome/cfDNA platform was also shown to generate approximately ten-fold more copies of EGFR-activating mutations compared to cfDNA-based BEAMing analysis and a higher clinical sensitivity (175). The dual approach addressing two biological processes of the tumor, with living cells shedding the exosomes and cfDNA being released by necrotic/apoptotic cells, yields better sensitivity and may facilitate earlier detection of the developing resistance to TKIs.

Circulating cell-free RNA (cfRNA)

Several earlier reports suggested that alterations in the plasma cfRNA can be detected in cancer patients; however, this biosource was considered less stable and thus less robust than cfDNA pool (176,177). However, a recent comprehensive study showed that samples collected in cfDNA-preserving tubes stored for up to 48 hours at room temperature allow for reliable plasma cfRNA assessment (178). This study also showed that cfRNA is specific for cancer origin and identified 15 mRNA transcripts potentially useful for detecting breast and lung cancers. Another recent study interrogated cfRNA as a potential biosource specific for multiple myeloma and liver cancer detection (179). Identified biomarkers included liver- or bone marrow-specific mRNAs related to cell-cycle processes. The levels of cfRNAs were increasing from the lowest in noncancerous states, intermediate in precancerous conditions, to highest in cancer. Further, the plasma cfRNA turned out to contain biomarkers providing the cue for cancer origin, which likely recruit from the tumor microenvironment and may reflect the healthy tissue response to the tumor. The plasma volume required for RNA extraction was 3 mL, without the extra steps of extracellular vesicles extraction.

Another interesting category of cfRNAs are cell-free microRNAs (cf-miRNAs), characterized by high stability even in harsh conditions (180). A systematic review, including early-stage NSCLC, showed a sensitivity exceeding 80% for miR-223, miR-20a, miR-448, and miR-145 and a specificity exceeding 90% for miR-628-3p, miR-29c, miR-210, and miR-1244 (181). In a cohort study including more than 3,000 patients, a panel of cf-miRNAs identified patients with lung cancer with 91.4% accuracy, 82.8% sensitivity, and 93.5% specificity (182). The prognostic value of cf-miRNAs was assessed in a study including 192 NSCLC patients (99 adenocarcinomas and 83 squamous cell carcinomas) (183). Of the 68 miRNAs analyzed, the most predictive for the outcome was cf-miR-126, with low expression predicting poor prognosis. This finding was confirmed in the meta-analysis including 1,012 patients (184).

Currently, the clinical usefulness of cfRNA for cancer detection requires further validation, particularly in screening for early-stage disease in the background of concomitant disorders, e.g., precancerous states or COPD. The targeted cfRNA appears to be a promising analyte to enhance the sensitivity of cfDNA-based cancer detection, particularly in low-shedding tumors. Lastly, a multianalyte test comprising both cfRNA and cfDNA may improve tissue of origin determination, which is vital to pan-cancer screening approaches.


Conclusions

Liquid biopsy has revolutionized the oncology field after overcoming several of the limitations of traditional tissue biopsy techniques. This innovative non-invasive approach has the unquestionable potential to optimize NSCLC management. Currently, it constitutes a valuable diagnostic tool for identifying druggable molecular alterations. New applications may include early detection, real-time monitoring, and the evaluation of spatial and temporal NSCLC heterogeneity.

We discussed the two most commonly used approaches, ctDNA/cfDNA and CTCs, and emerging liquid biopsy techniques, such as plasma DNA methylation, plasma cfRNA and metabolites, extracellular vesicles, and platelets. We also highlighted the limitations hampering clinical applications of liquid biopsies. We summarized the most up-to-date results of ongoing clinical trials and presented studies whose results may shortly impact clinical practice. The greatest hopes lie in MRD assessment, which may guide adjuvant therapies and allow early relapse detection and faster initiation of salvage treatments. However, the clinical application of liquid biopsies is still limited due to their suboptimal specificity and sensitivity, lack of standardization, and relatively high costs. Addressing these issues may allow further integration of liquid biopsies in the routine clinical setting, thus making a profound and permanent change in NSCLC management.


Acknowledgments

Funding: None.


Footnote

Reporting Checklist: The authors have completed the Narrative Review reporting checklist. Available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-22-742/rc

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-22-742/coif). JJ declares advisory roles for AstraZeneca, BMS, MSD and Exact Sciences, and presentation for Roche (not compensated). RD declares advisory roles for AstraZeneca, Roche, Boehringer-Ingelheim, MSD, Amgen, Pfizer, Bristol-Myers Squibb, Karyopharm, Bayer and FoundationMedicine. The other authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Sung H, Ferlay J, Siegel RL, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin 2021;71:209-49. [Crossref] [PubMed]
  2. Barlesi F, Mazieres J, Merlio JP, et al. Routine molecular profiling of patients with advanced non-small-cell lung cancer: results of a 1-year nationwide programme of the French Cooperative Thoracic Intergroup (IFCT). Lancet 2016;387:1415-26. [Crossref] [PubMed]
  3. Lindeman NI, Cagle PT, Aisner DL, et al. Updated Molecular Testing Guideline for the Selection of Lung Cancer Patients for Treatment With Targeted Tyrosine Kinase Inhibitors: Guideline From the College of American Pathologists, the International Association for the Study of Lung Cancer, and the Association for Molecular Pathology. Arch Pathol Lab Med 2018;142:321-46. [Crossref] [PubMed]
  4. Galvano A, Gristina V, Malapelle U, et al. The prognostic impact of tumor mutational burden (TMB) in the first-line management of advanced non-oncogene addicted non-small-cell lung cancer (NSCLC): a systematic review and meta-analysis of randomized controlled trials. ESMO Open 2021;6:100124. [Crossref] [PubMed]
  5. Ricciuti B, Wang X, Alessi JV, et al. Association of High Tumor Mutation Burden in Non-Small Cell Lung Cancers With Increased Immune Infiltration and Improved Clinical Outcomes of PD-L1 Blockade Across PD-L1 Expression Levels. JAMA Oncol 2022;8:1160-8. [Crossref] [PubMed]
  6. Jamal-Hanjani M, Wilson GA, McGranahan N, et al. Tracking the Evolution of Non-Small-Cell Lung Cancer. N Engl J Med 2017;376:2109-21. [Crossref] [PubMed]
  7. de Bruin EC, McGranahan N, Mitter R, et al. Spatial and temporal diversity in genomic instability processes defines lung cancer evolution. Science 2014;346:251-6. [Crossref] [PubMed]
  8. Zhang J, Fujimoto J, Zhang J, et al. Intratumor heterogeneity in localized lung adenocarcinomas delineated by multiregion sequencing. Science 2014;346:256-9. [Crossref] [PubMed]
  9. Planchard D, Popat S, Kerr K, et al. Metastatic non-small cell lung cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2018;29:iv192-237. [Crossref] [PubMed]
  10. Postmus PE, Kerr KM, Oudkerk M, et al. Early and locally advanced non-small-cell lung cancer (NSCLC): ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2017;28:iv1-iv21. [Crossref] [PubMed]
  11. Pascual J, Attard G, Bidard FC, 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] [PubMed]
  12. Redig AJ, Costa DB, Taibi M, et al. Prospective Study of Repeated Biopsy Feasibility and Acquired Resistance at Disease Progression in Patients With Advanced EGFR Mutant Lung Cancer Treated With Erlotinib in a Phase 2 Trial. JAMA Oncol 2016;2:1240-2. [Crossref] [PubMed]
  13. Ezeife DA, Spackman E, Juergens RA, et al. The economic value of liquid biopsy for genomic profiling in advanced non-small cell lung cancer. Ther Adv Med Oncol 2022;14:17588359221112696. [Crossref] [PubMed]
  14. Englmeier F, Bleckmann A, Brückl W, et al. Clinical benefit and cost-effectiveness analysis of liquid biopsy application in patients with advanced non-small cell lung cancer (NSCLC): a modelling approach. J Cancer Res Clin Oncol 2022; Epub ahead of print. [Crossref] [PubMed]
  15. Harvey MJ, Cunningham R, Sawchyn B, et al. Budget Impact Analysis of Comprehensive Genomic Profiling in Patients With Advanced Non-Small-Cell Lung Cancer. JCO Precis Oncol 2021;5:1611-24. [Crossref] [PubMed]
  16. Thompson JC, Yee SS, Troxel AB, et al. Detection of Therapeutically Targetable Driver and Resistance Mutations in Lung Cancer Patients by Next-Generation Sequencing of Cell-Free Circulating Tumor DNA. Clin Cancer Res 2016;22:5772-82. [Crossref] [PubMed]
  17. Garcia-Pardo M, Czarnecka K, Law JH, et al. Plasma-first: accelerating lung cancer diagnosis and molecular profiling through liquid biopsy. Ther Adv Med Oncol 2022;14:17588359221126151. [Crossref] [PubMed]
  18. Cui W, Milner-Watts C, O'Sullivan H, et al. Up-front cell-free DNA next generation sequencing improves target identification in UK first line advanced non-small cell lung cancer (NSCLC) patients. Eur J Cancer 2022;171:44-54. [Crossref] [PubMed]
  19. Martins I, Ribeiro IP, Jorge J, et al. Liquid Biopsies: Applications for Cancer Diagnosis and Monitoring. Genes (Basel) 2021;12:349. [Crossref] [PubMed]
  20. Ignatiadis M, Sledge GW, Jeffrey SS. Liquid biopsy enters the clinic - implementation issues and future challenges. Nat Rev Clin Oncol 2021;18:297-312. [Crossref] [PubMed]
  21. Bonanno L, Dal Maso A, Pavan A, et al. Liquid biopsy and non-small cell lung cancer: are we looking at the tip of the iceberg? Br J Cancer 2022;127:383-93. [Crossref] [PubMed]
  22. FDA. FDA approves first blood test to detect gene mutation associated with non-small cell lung cancer. Accessed January 14, 2023. Available online: https://www.fda.gov/news-events/press-announcements/fda-approves-first-blood-test-detect-gene-mutation-associated-non-small-cell-lung-cancer
  23. Mandel P, Metais P. Nuclear Acids In Human Blood Plasma. C R Seances Soc Biol Fil. 1948;142:241-3. [PubMed]
  24. Moss J, Magenheim J, Neiman D, et al. Comprehensive human cell-type methylation atlas reveals origins of circulating cell-free DNA in health and disease. Nat Commun 2018;9:5068. [Crossref] [PubMed]
  25. Razavi P, Li BT, Brown DN, et al. High-intensity sequencing reveals the sources of plasma circulating cell-free DNA variants. Nat Med 2019;25:1928-37. [Crossref] [PubMed]
  26. Leon SA, Shapiro B, Sklaroff DM, et al. Free DNA in the serum of cancer patients and the effect of therapy. Cancer Res 1977;37:646-50. [PubMed]
  27. Jahr S, Hentze H, Englisch S, et al. DNA fragments in the blood plasma of cancer patients: quantitations and evidence for their origin from apoptotic and necrotic cells. Cancer Res 2001;61:1659-65. [PubMed]
  28. Diehl F, Schmidt K, Choti MA, et al. Circulating mutant DNA to assess tumor dynamics. Nat Med 2008;14:985-90. [Crossref] [PubMed]
  29. Muhanna N, Di Grappa MA, Chan HHL, et al. Cell-Free DNA Kinetics in a Pre-Clinical Model of Head and Neck Cancer. Sci Rep 2017;7:16723. [Crossref] [PubMed]
  30. Pittella-Silva F, Chin YM, Chan HT, et al. Plasma or Serum: Which Is Preferable for Mutation Detection in Liquid Biopsy? Clin Chem 2020;66:946-57. [Crossref] [PubMed]
  31. Bronkhorst AJ, Ungerer V, Holdenrieder S. The emerging role of cell-free DNA as a molecular marker for cancer management. Biomol Detect Quantif 2019;17:100087. [Crossref] [PubMed]
  32. Lampignano R, Neumann MHD, Weber S, et al. Multicenter Evaluation of Circulating Cell-Free DNA Extraction and Downstream Analyses for the Development of Standardized (Pre)analytical Work Flows. Clin Chem 2020;66:149-60. [Crossref] [PubMed]
  33. Markus H, Contente-Cuomo T, Farooq M, et al. Evaluation of pre-analytical factors affecting plasma DNA analysis. Sci Rep 2018;8:7375. [Crossref] [PubMed]
  34. Stawski R, Walczak K, Kosielski P, et al. Repeated bouts of exhaustive exercise increase circulating cell free nuclear and mitochondrial DNA without development of tolerance in healthy men. PLoS One 2017;12:e0178216. [Crossref] [PubMed]
  35. Young AL, Challen GA, Birmann BM, et al. Clonal haematopoiesis harbouring AML-associated mutations is ubiquitous in healthy adults. Nat Commun 2016;7:12484. [Crossref] [PubMed]
  36. Chan HT, Chin YM, Nakamura Y, et al. Clonal Hematopoiesis in Liquid Biopsy: From Biological Noise to Valuable Clinical Implications. Cancers (Basel) 2020;12:2277. [Crossref] [PubMed]
  37. Chabon JJ, Hamilton EG, Kurtz DM, et al. Integrating genomic features for non-invasive early lung cancer detection. Nature 2020;580:245-51. [Crossref] [PubMed]
  38. Moding EJ, Nabet BY, Alizadeh AA, et al. Detecting Liquid Remnants of Solid Tumors: Circulating Tumor DNA Minimal Residual Disease. Cancer Discov 2021;11:2968-86. [Crossref] [PubMed]
  39. Rolfo C, Mack PC, Scagliotti GV, 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] [PubMed]
  40. Heitzer E, van den Broek D, Denis MG, et al. Recommendations for a practical implementation of circulating tumor DNA mutation testing in metastatic non-small-cell lung cancer. ESMO Open 2022;7:100399. [Crossref] [PubMed]
  41. National Comprehensive Cancer Network Guidelines. Non-Small Cell Lung Cancer (Version 1.2023). Accessed January 14, 2023. Available online: https://www.nccn.org/professionals/physician_gls/pdf/nscl.pdf
  42. García-Pardo M, Makarem M, Li JJN, et al. Integrating circulating-free DNA (cfDNA) analysis into clinical practice: opportunities and challenges. Br J Cancer 2022;127:592-602. [Crossref] [PubMed]
  43. Aggarwal C, Thompson JC, Black TA, 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] [PubMed]
  44. Leighl NB, Page RD, Raymond VM, 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] [PubMed]
  45. FDA. FDA Approves First Liquid Biopsy Next-Generation Sequencing Companion Diagnostic Test. Accessed September 18, 2022. Available online: https://www.fda.gov/news-events/press-announcements/fda-approves-first-liquid-biopsy-next-generation-sequencing-companion-diagnostic-test ().
  46. FDA. FDA approves liquid biopsy NGS companion diagnostic test for multiple cancers and biomarkers. Accessed September 18, 2022. Available online: https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-liquid-biopsy-ngs-companion-diagnostic-test-multiple-cancers-and-biomarkers
  47. The Guardant360®. Assay Specifications. Accessed September 18, 2022. Available online: https://guardant360cdx.com/
  48. FoundationOne® Liquid CDx. Accessed September 18, 2022. Available online: https://www.foundationmedicine.com/test/foundationone-liquid-cdx
  49. Di Capua D, Bracken-Clarke D, Ronan K, et al. The Liquid Biopsy for Lung Cancer: State of the Art, Limitations and Future Developments. Cancers (Basel) 2021;13:3923. [Crossref] [PubMed]
  50. Weber B, Meldgaard P, Hager H, et al. Detection of EGFR mutations in plasma and biopsies from non-small cell lung cancer patients by allele-specific PCR assays. BMC Cancer 2014;14:294. [Crossref] [PubMed]
  51. Goldman JW, Noor ZS, Remon J, et al. Are liquid biopsies a surrogate for tissue EGFR testing? Ann Oncol 2018;29:i38-46. [Crossref] [PubMed]
  52. Oxnard GR, Thress KS, Alden RS, et al. Association Between Plasma Genotyping and Outcomes of Treatment With Osimertinib (AZD9291) in Advanced Non-Small-Cell Lung Cancer. J Clin Oncol 2016;34:3375-82. [Crossref] [PubMed]
  53. Karachaliou N, Mayo-de las Casas C, Queralt C, et al. Association of EGFR L858R Mutation in Circulating Free DNA With Survival in the EURTAC Trial. JAMA Oncol 2015;1:149-57. [Crossref] [PubMed]
  54. Hofman P. Detecting Resistance to Therapeutic ALK Inhibitors in Tumor Tissue and Liquid Biopsy Markers: An Update to a Clinical Routine Practice. Cells 2021;10:168. [Crossref] [PubMed]
  55. Dagogo-Jack I, Brannon AR, Ferris LA, et al. Tracking the Evolution of Resistance to ALK Tyrosine Kinase Inhibitors through Longitudinal Analysis of Circulating Tumor DNA. JCO Precis Oncol 2018;2018:PO.17.00160.
  56. Dagogo-Jack I, Rooney M, Lin JJ, et al. Treatment with Next-Generation ALK Inhibitors Fuels Plasma ALK Mutation Diversity. Clin Cancer Res 2019;25:6662-70. [Crossref] [PubMed]
  57. Horn L, Whisenant JG, Wakelee H, et al. Monitoring Therapeutic Response and Resistance: Analysis of Circulating Tumor DNA in Patients With ALK+ Lung Cancer. J Thorac Oncol 2019;14:1901-11. [Crossref] [PubMed]
  58. Madsen AT, Winther-Larsen A, McCulloch T, et al. Genomic Profiling of Circulating Tumor DNA Predicts Outcome and Demonstrates Tumor Evolution in ALK-Positive Non-Small Cell Lung Cancer Patients. Cancers (Basel) 2020;12:947. [Crossref] [PubMed]
  59. Zhang X, Zhu X, Wang C, et al. Non-targeted and targeted metabolomics approaches to diagnosing lung cancer and predicting patient prognosis. Oncotarget 2016;7:63437-48. [Crossref] [PubMed]
  60. Goldberg SB, Narayan A, Kole AJ, et al. Early Assessment of Lung Cancer Immunotherapy Response via Circulating Tumor DNA. Clin Cancer Res 2018;24:1872-80. [Crossref] [PubMed]
  61. Gale D, Heider K, Ruiz-Valdepenas A, et al. Residual ctDNA after treatment predicts early relapse in patients with early-stage non-small cell lung cancer. Ann Oncol 2022;33:500-10. [Crossref] [PubMed]
  62. Bettegowda C, Sausen M, Leary RJ, et al. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci Transl Med 2014;6:224ra24. [Crossref] [PubMed]
  63. Phallen J, Sausen M, Adleff V, et al. Direct detection of early-stage cancers using circulating tumor DNA. Sci Transl Med 2017;9:eaan2415. [Crossref] [PubMed]
  64. Schuurhuis GJ, Heuser M, Freeman S, et al. Minimal/measurable residual disease in AML: a consensus document from the European LeukemiaNet MRD Working Party. Blood 2018;131:1275-91. [Crossref] [PubMed]
  65. Heuser M, Freeman SD, Ossenkoppele GJ, et al. 2021 Update on MRD in acute myeloid leukemia: a consensus document from the European LeukemiaNet MRD Working Party. Blood 2021;138:2753-67. [Crossref] [PubMed]
  66. Tie J, Cohen JD, Lahouel K, et al. Circulating Tumor DNA Analysis Guiding Adjuvant Therapy in Stage II Colon Cancer. N Engl J Med 2022;386:2261-72. [Crossref] [PubMed]
  67. Chaudhuri AA, Chabon JJ, Lovejoy AF, et al. Early Detection of Molecular Residual Disease in Localized Lung Cancer by Circulating Tumor DNA Profiling. Cancer Discov 2017;7:1394-403. [Crossref] [PubMed]
  68. Abbosh C, Birkbak NJ, Wilson GA, et al. Phylogenetic ctDNA analysis depicts early-stage lung cancer evolution. Nature 2017;545:446-51. [Crossref] [PubMed]
  69. Chen K, Zhao H, Shi Y, et al. Perioperative Dynamic Changes in Circulating Tumor DNA in Patients with Lung Cancer (DYNAMIC). Clin Cancer Res 2019;25:7058-67. [Crossref] [PubMed]
  70. Zviran A, Schulman RC, Shah M, et al. Genome-wide cell-free DNA mutational integration enables ultra-sensitive cancer monitoring. Nat Med 2020;26:1114-24. [Crossref] [PubMed]
  71. Abbosh C, Frankell A, Garnett A, et al. Abstract CT023: Phylogenetic tracking and minimal residual disease detection using ctDNA in early-stage NSCLC: A lung TRACERx study. Cancer Res 2020;80:CT023. [Crossref]
  72. Cabel L, Proudhon C, Romano E, et al. Clinical potential of circulating tumour DNA in patients receiving anticancer immunotherapy. Nat Rev Clin Oncol 2018;15:639-50. [Crossref] [PubMed]
  73. Gandara DR, Paul SM, Kowanetz M, 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] [PubMed]
  74. Kim ES, Velcheti V, Mekhail T, 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] [PubMed]
  75. Merino DM, McShane LM, Fabrizio D, 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] [PubMed]
  76. Vega DM, Yee LM, McShane LM, et al. Aligning tumor mutational burden (TMB) quantification across diagnostic platforms: phase II of the Friends of Cancer Research TMB Harmonization Project. Ann Oncol 2021;32:1626-36. [Crossref] [PubMed]
  77. Guibert N, Mazieres J, Delaunay M, et al. Monitoring of KRAS-mutated ctDNA to discriminate pseudo-progression from true progression during anti-PD-1 treatment of lung adenocarcinoma. Oncotarget 2017;8:38056-60. [Crossref] [PubMed]
  78. Ricciuti B, Jones G, Severgnini M, et al. Early plasma circulating tumor DNA (ctDNA) changes predict response to first-line pembrolizumab-based therapy in non-small cell lung cancer (NSCLC). J Immunother Cancer 2021;9:e001504. [Crossref] [PubMed]
  79. Gray J, Thompson JC, Carpenter EL, et al. Plasma Cell-Free DNA Genotyping: From an Emerging Concept to a Standard-of-Care Tool in Metastatic Non-Small Cell Lung Cancer. Oncologist 2021;26:e1812-21. [Crossref] [PubMed]
  80. Remon J, Menis J, Hasan B, et al. The APPLE Trial: Feasibility and Activity of AZD9291 (Osimertinib) Treatment on Positive PLasma T790M in EGFR-mutant NSCLC Patients. EORTC 1613. Clin Lung Cancer 2017;18:583-8. [Crossref] [PubMed]
  81. Masip JR, Besse B, Aix SP, et al. LBA51 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 2022;33:S1419. [Crossref]
  82. Dotan E, Cohen SJ, Alpaugh KR, et al. Circulating tumor cells: evolving evidence and future challenges. Oncologist 2009;14:1070-82. [Crossref] [PubMed]
  83. Alix-Panabières C, Pantel K. Challenges in circulating tumour cell research. Nat Rev Cancer 2014;14:623-31. [Crossref] [PubMed]
  84. Szczerba BM, Castro-Giner F, Vetter M, et al. Neutrophils escort circulating tumour cells to enable cell cycle progression. Nature 2019;566:553-7. [Crossref] [PubMed]
  85. Au SH, Storey BD, Moore JC, et al. Clusters of circulating tumor cells traverse capillary-sized vessels. Proc Natl Acad Sci U S A 2016;113:4947-52. [Crossref] [PubMed]
  86. Reddy RM, Murlidhar V, Zhao L, et al. Pulmonary venous blood sampling significantly increases the yield of circulating tumor cells in early-stage lung cancer. J Thorac Cardiovasc Surg 2016;151:852-8. [Crossref] [PubMed]
  87. Okumura Y, Tanaka F, Yoneda K, et al. Circulating tumor cells in pulmonary venous blood of primary lung cancer patients. Ann Thorac Surg 2009;87:1669-75. [Crossref] [PubMed]
  88. Allard WJ, Matera J, Miller MC, et al. Tumor cells circulate in the peripheral blood of all major carcinomas but not in healthy subjects or patients with nonmalignant diseases. Clin Cancer Res 2004;10:6897-904. [Crossref] [PubMed]
  89. Alix-Panabières C. EPISPOT assay: detection of viable DTCs/CTCs in solid tumor patients. Recent Results Cancer Res 2012;195:69-76. [Crossref] [PubMed]
  90. Shields CW 4th, Reyes CD, López GP. Microfluidic cell sorting: a review of the advances in the separation of cells from debulking to rare cell isolation. Lab Chip 2015;15:1230-49. [Crossref] [PubMed]
  91. Jonas DE, Reuland DS, Reddy SM, et al. Screening for Lung Cancer With Low-Dose Computed Tomography: Updated Evidence Report and Systematic Review for the US Preventive Services Task Force. JAMA 2021;325:971-87. [Crossref] [PubMed]
  92. Sheu CC, Yu YP, Tsai JR, et al. Development of a membrane array-based multimarker assay for detection of circulating cancer cells in patients with non-small cell lung cancer. Int J Cancer 2006;119:1419-26. [Crossref] [PubMed]
  93. Yu Y, Chen Z, Dong J, et al. Folate receptor-positive circulating tumor cells as a novel diagnostic biomarker in non-small cell lung cancer. Transl Oncol 2013;6:697-702. [Crossref] [PubMed]
  94. Wang L, Wu C, Qiao L, et al. Clinical Significance of Folate Receptor-positive Circulating Tumor Cells Detected by Ligand-targeted Polymerase Chain Reaction in Lung Cancer. J Cancer. 2017;8:104-10. [Crossref] [PubMed]
  95. Ilie M, Hofman V, Long-Mira E, et al. "Sentinel" circulating tumor cells allow early diagnosis of lung cancer in patients with chronic obstructive pulmonary disease. PLoS One 2014;9:e111597. [Crossref] [PubMed]
  96. Duan GC, Zhang XP, Wang HE, et al. Circulating Tumor Cells as a Screening and Diagnostic Marker for Early-Stage Non-Small Cell Lung Cancer. Onco Targets Ther 2020;13:1931-9. [Crossref] [PubMed]
  97. Marquette CH, Boutros J, Benzaquen J, et al. Circulating tumour cells as a potential biomarker for lung cancer screening: a prospective cohort study. Lancet Respir Med 2020;8:709-16. [Crossref] [PubMed]
  98. Das M, Riess JW, Frankel P, et al. ERCC1 expression in circulating tumor cells (CTCs) using a novel detection platform correlates with progression-free survival (PFS) in patients with metastatic non-small-cell lung cancer (NSCLC) receiving platinum chemotherapy. Lung Cancer 2012;77:421-6. [Crossref] [PubMed]
  99. Devriese LA, Bosma AJ, van de Heuvel MM, et al. Circulating tumor cell detection in advanced non-small cell lung cancer patients by multi-marker QPCR analysis. Lung Cancer 2012;75:242-7. [Crossref] [PubMed]
  100. Hofman V, Long E, Ilie M, et al. Morphological analysis of circulating tumour cells in patients undergoing surgery for non-small cell lung carcinoma using the isolation by size of epithelial tumour cell (ISET) method. Cytopathology 2012;23:30-8. [Crossref] [PubMed]
  101. Krebs MG, Hou JM, Sloane R, et al. Analysis of circulating tumor cells in patients with non-small cell lung cancer using epithelial marker-dependent and -independent approaches. J Thorac Oncol 2012;7:306-15. [Crossref] [PubMed]
  102. Saucedo-Zeni N, Mewes S, Niestroj R, et al. A novel method for the in vivo isolation of circulating tumor cells from peripheral blood of cancer patients using a functionalized and structured medical wire. Int J Oncol 2012;41:1241-50. [PubMed]
  103. Wendel M, Bazhenova L, Boshuizen R, et al. Fluid biopsy for circulating tumor cell identification in patients with early-and late-stage non-small cell lung cancer: a glimpse into lung cancer biology. Phys Biol 2012;9:016005. [Crossref] [PubMed]
  104. Funaki S, Sawabata N, Abulaiti A, et al. Significance of tumour vessel invasion in determining the morphology of isolated tumour cells in the pulmonary vein in non-small-cell lung cancer. Eur J Cardiothorac Surg 2013;43:1126-30. [Crossref] [PubMed]
  105. Hosokawa M, Kenmotsu H, Koh Y, et al. Size-based isolation of circulating tumor cells in lung cancer patients using a microcavity array system. PLoS One 2013;8:e67466. [Crossref] [PubMed]
  106. Earhart CM, Hughes CE, Gaster RS, et al. Isolation and mutational analysis of circulating tumor cells from lung cancer patients with magnetic sifters and biochips. Lab Chip 2014;14:78-88. [Crossref] [PubMed]
  107. Chudasama D, Rice A, Soppa G, et al. Circulating tumour cells in patients with lung cancer undergoing endobronchial cryotherapy. Cryobiology 2015;71:161-3. [Crossref] [PubMed]
  108. Hanssen A, Wagner J, Gorges TM, et al. Characterization of different CTC subpopulations in non-small cell lung cancer. Sci Rep 2016;6:28010. [Crossref] [PubMed]
  109. Chudasama D, Burnside N, Beeson J, et al. Perioperative detection of circulating tumour cells in patients with lung cancer. Oncol Lett 2017;14:1281-6. [Crossref] [PubMed]
  110. Chudasama D, Barr J, Beeson J, et al. Detection of Circulating Tumour Cells and Survival of Patients with Non-small Cell Lung Cancer. Anticancer Res 2017;37:169-73. [Crossref] [PubMed]
  111. Krebs MG, Sloane R, Priest L, et al. Evaluation and prognostic significance of circulating tumor cells in patients with non-small-cell lung cancer. J Clin Oncol 2011;29:1556-63. [Crossref] [PubMed]
  112. Qi Y, Wang W. Clinical significance of circulating tumor cells in squamous cell lung cancer patients. Cancer Biomark 2017;18:161-7. [Crossref] [PubMed]
  113. Coco S, Alama A, Vanni I, et al. Circulating Cell-Free DNA and Circulating Tumor Cells as Prognostic and Predictive Biomarkers in Advanced Non-Small Cell Lung Cancer Patients Treated with First-Line Chemotherapy. Int J Mol Sci 2017;18:1035. [Crossref] [PubMed]
  114. Tong B, Xu Y, Zhao J, et al. Prognostic role of circulating tumor cells in patients with EGFR-mutated or ALK-rearranged non-small cell lung cancer. Thorac Cancer 2018;9:640-5. [Crossref] [PubMed]
  115. Dong J, Zhu D, Tang X, et al. Detection of Circulating Tumor Cell Molecular Subtype in Pulmonary Vein Predicting Prognosis of Stage I-III Non-small Cell Lung Cancer Patients. Front Oncol 2019;9:1139. [Crossref] [PubMed]
  116. Wei T, Zhu D, Yang Y, et al. The application of nano-enrichment in CTC detection and the clinical significance of CTCs in non-small cell lung cancer (NSCLC) treatment. PLoS One 2019;14:e0219129. [Crossref] [PubMed]
  117. Castello A, Carbone FG, Rossi S, et al. Circulating Tumor Cells and Metabolic Parameters in NSCLC Patients Treated with Checkpoint Inhibitors. Cancers (Basel) 2020;12:487. [Crossref] [PubMed]
  118. Shishido SN, Carlsson A, Nieva J, et al. Circulating tumor cells as a response monitor in stage IV non-small cell lung cancer. J Transl Med 2019;17:294. [Crossref] [PubMed]
  119. Lindsay CR, Blackhall FH, Carmel A, et al. EPAC-lung: pooled analysis of circulating tumour cells in advanced non-small cell lung cancer. Eur J Cancer 2019;117:60-8. [Crossref] [PubMed]
  120. Carlsson A, Nair VS, Luttgen MS, et al. Circulating tumor microemboli diagnostics for patients with non-small-cell lung cancer. J Thorac Oncol 2014;9:1111-9. [Crossref] [PubMed]
  121. Tay RY, Fernández-Gutiérrez F, Foy V, et al. Prognostic value of circulating tumour cells in limited-stage small-cell lung cancer: analysis of the concurrent once-daily versus twice-daily radiotherapy (CONVERT) randomised controlled trial. Ann Oncol 2019;30:1114-20. [Crossref] [PubMed]
  122. Frick MA, Feigenberg SJ, Jean-Baptiste SR, et al. Circulating Tumor Cells Are Associated with Recurrent Disease in Patients with Early-Stage Non-Small Cell Lung Cancer Treated with Stereotactic Body Radiotherapy. Clin Cancer Res 2020;26:2372-80. [Crossref] [PubMed]
  123. Maheswaran S, Sequist LV, Nagrath S, et al. Detection of mutations in EGFR in circulating lung-cancer cells. N Engl J Med 2008;359:366-77. [Crossref] [PubMed]
  124. Pailler E, Faugeroux V, Oulhen M, et al. Acquired Resistance Mutations to ALK Inhibitors Identified by Single Circulating Tumor Cell Sequencing in ALK-Rearranged Non-Small-Cell Lung Cancer. Clin Cancer Res 2019;25:6671-82. [Crossref] [PubMed]
  125. Pailler E, Auger N, Lindsay CR, et al. High level of chromosomal instability in circulating tumor cells of ROS1-rearranged non-small-cell lung cancer. Ann Oncol 2015;26:1408-15. [Crossref] [PubMed]
  126. Gorges TM, Penkalla N, Schalk T, et al. Enumeration and Molecular Characterization of Tumor Cells in Lung Cancer Patients Using a Novel In Vivo Device for Capturing Circulating Tumor Cells. Clin Cancer Res 2016;22:2197-206. [Crossref] [PubMed]
  127. Sinoquet L, Jacot W, Gauthier L, et al. Programmed Cell Death Ligand 1-Expressing Circulating Tumor Cells: A New Prognostic Biomarker in Non-Small Cell Lung Cancer. Clin Chem 2021;67:1503-12. [Crossref] [PubMed]
  128. Ilié M, Szafer-Glusman E, Hofman V, et al. Detection of PD-L1 in circulating tumor cells and white blood cells from patients with advanced non-small-cell lung cancer. Ann Oncol 2018;29:193-9. [Crossref] [PubMed]
  129. CANCER-ID – Innovation in Medicine. Accessed September 18, 2022. Available online: https://www.cancer-id.eu/
  130. Heitzer E, Haque IS, Roberts CES, et al. Current and future perspectives of liquid biopsies in genomics-driven oncology. Nat Rev Genet 2019;20:71-88. [Crossref] [PubMed]
  131. Hu Y, Ulrich BC, Supplee J, et al. False-Positive Plasma Genotyping Due to Clonal Hematopoiesis. Clin Cancer Res 2018;24:4437-43. [Crossref] [PubMed]
  132. Yan H, Guan Q, He J, et al. Individualized analysis reveals CpG sites with methylation aberrations in almost all lung adenocarcinoma tissues. J Transl Med 2017;15:26. [Crossref] [PubMed]
  133. Guo S, Diep D, Plongthongkum N, et al. Identification of methylation haplotype blocks aids in deconvolution of heterogeneous tissue samples and tumor tissue-of-origin mapping from plasma DNA. Nat Genet 2017;49:635-42. [Crossref] [PubMed]
  134. Liu MC, Oxnard GR, Klein EA, et al. Sensitive and specific multi-cancer detection and localization using methylation signatures in cell-free DNA. Ann Oncol 2020;31:745-59. [Crossref] [PubMed]
  135. Klein EA, Richards D, Cohn A, 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] [PubMed]
  136. Nadauld LD, McDonnell CH 3rd, Beer TM, et al. The PATHFINDER Study: Assessment of the Implementation of an Investigational Multi-Cancer Early Detection Test into Clinical Practice. Cancers (Basel) 2021;13:3501. [Crossref] [PubMed]
  137. Swanton C, Neal RD, Johnson PWM, et al. NHS-Galleri Trial Design: Equitable study recruitment tactics for targeted population-level screening with a multi-cancer early detection (MCED) test. J Clinic Oncol 2022;40:TPS6606. [Crossref]
  138. Chemi F, Pearce SP, Clipson A, et al. cfDNA methylome profiling for detection and subtyping of small cell lung cancers. Nat Cancer 2022;3:1260-70. [Crossref] [PubMed]
  139. Gay CM, Stewart CA, Park EM, et al. Patterns of transcription factor programs and immune pathway activation define four major subtypes of SCLC with distinct therapeutic vulnerabilities. Cancer Cell 2021;39:346-360.e7. [Crossref] [PubMed]
  140. Mollaoglu G, Guthrie MR, Böhm S, et al. MYC Drives Progression of Small Cell Lung Cancer to a Variant Neuroendocrine Subtype with Vulnerability to Aurora Kinase Inhibition. Cancer Cell 2017;31:270-85. [Crossref] [PubMed]
  141. Augustyn A, Borromeo M, Wang T, et al. ASCL1 is a lineage oncogene providing therapeutic targets for high-grade neuroendocrine lung cancers. Proc Natl Acad Sci U S A 2014;111:14788-93. [Crossref] [PubMed]
  142. Schmidt DR, Patel R, Kirsch DG, et al. Metabolomics in cancer research and emerging applications in clinical oncology. CA Cancer J Clin 2021;71:333-58. [Crossref] [PubMed]
  143. Sinkala M, Mulder N, Patrick Martin D. Metabolic gene alterations impact the clinical aggressiveness and drug responses of 32 human cancers. Commun Biol 2019;2:414. [Crossref] [PubMed]
  144. Hu J, Locasale JW, Bielas JH, et al. Heterogeneity of tumor-induced gene expression changes in the human metabolic network. Nat Biotechnol 2013;31:522-9. [Crossref] [PubMed]
  145. Ding H, Chen Z, Wu K, et al. Activation of the NRF2 antioxidant program sensitizes tumors to G6PD inhibition. Sci Adv 2021;7:eabk1023. [Crossref] [PubMed]
  146. Fox DB, Garcia NMG, McKinney BJ, et al. NRF2 activation promotes the recurrence of dormant tumour cells through regulation of redox and nucleotide metabolism. Nat Metab 2020;2:318-34. [Crossref] [PubMed]
  147. Madama D, Martins R, Pires AS, et al. Metabolomic Profiling in Lung Cancer: A Systematic Review. Metabolites 2021;11:630. [Crossref] [PubMed]
  148. Mayers JR, Torrence ME, Danai LV, et al. Tissue of origin dictates branched-chain amino acid metabolism in mutant Kras-driven cancers. Science 2016;353:1161-5. [Crossref] [PubMed]
  149. Rocha CM, Barros AS, Goodfellow BJ, et al. NMR metabolomics of human lung tumours reveals distinct metabolic signatures for adenocarcinoma and squamous cell carcinoma. Carcinogenesis 2015;36:68-75. [Crossref] [PubMed]
  150. Losman JA, Kaelin WG Jr. What a difference a hydroxyl makes: mutant IDH, (R)-2-hydroxyglutarate, and cancer. Genes Dev 2013;27:836-52. [Crossref] [PubMed]
  151. Janin M, Mylonas E, Saada V, et al. Serum 2-hydroxyglutarate production in IDH1- and IDH2-mutated de novo acute myeloid leukemia: a study by the Acute Leukemia French Association group. J Clin Oncol 2014;32:297-305. [Crossref] [PubMed]
  152. Dang L, White DW, Gross S, et al. Cancer-associated IDH1 mutations produce 2-hydroxyglutarate. Nature 2009;462:739-44. [Crossref] [PubMed]
  153. Gross S, Cairns RA, Minden MD, et al. Cancer-associated metabolite 2-hydroxyglutarate accumulates in acute myelogenous leukemia with isocitrate dehydrogenase 1 and 2 mutations. J Exp Med 2010;207:339-44. [Crossref] [PubMed]
  154. Cairns RA, Mak TW. Oncogenic isocitrate dehydrogenase mutations: mechanisms, models, and clinical opportunities. Cancer Discov 2013;3:730-41. [Crossref] [PubMed]
  155. Zehir A, Benayed R, Shah RH, et al. Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients. Nat Med 2017;23:703-13. [Crossref] [PubMed]
  156. Lombardi G, Corona G, Bellu L, et al. Diagnostic value of plasma and urinary 2-hydroxyglutarate to identify patients with isocitrate dehydrogenase-mutated glioma. Oncologist 2015;20:562-7. [Crossref] [PubMed]
  157. Wikoff WR, Grapov D, Fahrmann JF, et al. Metabolomic markers of altered nucleotide metabolism in early stage adenocarcinoma. Cancer Prev Res (Phila) 2015;8:410-8. [Crossref] [PubMed]
  158. Hensley CT, Faubert B, Yuan Q, et al. Metabolic Heterogeneity in Human Lung Tumors. Cell 2016;164:681-94. [Crossref] [PubMed]
  159. An Z, Chen Y, Zhang R, et al. Integrated ionization approach for RRLC-MS/MS-based metabonomics: finding potential biomarkers for lung cancer. J Proteome Res 2010;9:4071-81. [Crossref] [PubMed]
  160. Seidel A, Seidel P, Manuwald O, et al. Modified nucleosides as biomarkers for early cancer diagnose in exposed populations. Environ Toxicol 2015;30:956-67. [Crossref] [PubMed]
  161. In 't Veld SGJG, Arkani M, Post E, et al. Detection and localization of early- and late-stage cancers using platelet RNA. Cancer Cell 2022;40:999-1009.e6. [Crossref] [PubMed]
  162. Nilsson RJ, Karachaliou N, Berenguer J, et al. Rearranged EML4-ALK fusion transcripts sequester in circulating blood platelets and enable blood-based crizotinib response monitoring in non-small-cell lung cancer. Oncotarget 2016;7:1066-75. [Crossref] [PubMed]
  163. Best MG, Sol N, In 't Veld SGJG, et al. Swarm Intelligence-Enhanced Detection of Non-Small-Cell Lung Cancer Using Tumor-Educated Platelets. Cancer Cell 2017;32:238-252.e9.
  164. Lennon AM, Buchanan AH, Kinde I, et al. Feasibility of blood testing combined with PET-CT to screen for cancer and guide intervention. Science 2020;369:eabb9601. [Crossref] [PubMed]
  165. Stackpole M, Zeng W, Li S, et al. Abstract 24: Multi-feature ensemble learning on cell-free dna for accurately detecting and locating cancer. Cancer Res 2021;81:24. [Crossref]
  166. Ratajczak MZ, Ratajczak J. Extracellular microvesicles/exosomes: discovery, disbelief, acceptance, and the future? Leukemia 2020;34:3126-35. [Crossref] [PubMed]
  167. Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes. Science 2020;367:eaau6977. [Crossref] [PubMed]
  168. Enderle D, Spiel A, Coticchia CM, et al. Characterization of RNA from Exosomes and Other Extracellular Vesicles Isolated by a Novel Spin Column-Based Method. PLoS One 2015;10:e0136133. [Crossref] [PubMed]
  169. Sandfeld-Paulsen B, Jakobsen KR, Bæk R, et al. Exosomal Proteins as Diagnostic Biomarkers in Lung Cancer. J Thorac Oncol 2016;11:1701-10. [Crossref] [PubMed]
  170. Lobb RJ, van Amerongen R, Wiegmans A, et al. Exosomes derived from mesenchymal non-small cell lung cancer cells promote chemoresistance. Int J Cancer 2017;141:614-20. [Crossref] [PubMed]
  171. Liu X, Jiang T, Li X, et al. Exosomes transmit T790M mutation-induced resistance in EGFR-mutant NSCLC by activating PI3K/AKT signalling pathway. J Cell Mol Med 2020;24:1529-40. [Crossref] [PubMed]
  172. Wu S, Luo M, To KKW, et al. Intercellular transfer of exosomal wild type EGFR triggers osimertinib resistance in non-small cell lung cancer. Mol Cancer 2021;20:17. [Crossref] [PubMed]
  173. Chen R, Qian Z, Xu X, et al. Exosomes-transmitted miR-7 reverses gefitinib resistance by targeting YAP in non-small-cell lung cancer. Pharmacol Res 2021;165:105442. [Crossref] [PubMed]
  174. Castellanos-Rizaldos E, Grimm DG, Tadigotla V, et al. Exosome-Based Detection of EGFR T790M in Plasma from Non-Small Cell Lung Cancer Patients. Clin Cancer Res 2018;24:2944-50. [Crossref] [PubMed]
  175. Krug AK, Enderle D, Karlovich C, et al. Improved EGFR mutation detection using combined exosomal RNA and circulating tumor DNA in NSCLC patient plasma. Ann Oncol 2018;29:700-6. [Crossref] [PubMed]
  176. Kopreski MS, Benko FA, Gocke CD. Circulating RNA as a tumor marker: detection of 5T4 mRNA in breast and lung cancer patient serum. Ann N Y Acad Sci 2001;945:172-8. [Crossref] [PubMed]
  177. Chang JW, Shih CL, Wang CL, et al. Transcriptomic Analysis in Liquid Biopsy Identifies Circulating PCTAIRE-1 mRNA as a Biomarker in NSCLC. Cancer Genomics Proteomics 2020;17:91-100. [Crossref] [PubMed]
  178. Larson MH, Pan W, Kim HJ, et al. A comprehensive characterization of the cell-free transcriptome reveals tissue- and subtype-specific biomarkers for cancer detection. Nat Commun 2021;12:2357. [Crossref] [PubMed]
  179. Roskams-Hieter B, Kim HJ, Anur P, et al. Plasma cell-free RNA profiling distinguishes cancers from pre-malignant conditions in solid and hematologic malignancies. NPJ Precis Oncol 2022;6:28. [Crossref] [PubMed]
  180. Glinge C, Clauss S, Boddum K, et al. Stability of Circulating Blood-Based MicroRNAs - Pre-Analytic Methodological Considerations. PLoS One 2017;12:e0167969. [Crossref] [PubMed]
  181. Moretti F, D'Antona P, Finardi E, et al. Systematic review and critique of circulating miRNAs as biomarkers of stage I-II non-small cell lung cancer. Oncotarget 2017;8:94980-96. [Crossref] [PubMed]
  182. Fehlmann T, Kahraman M, Ludwig N, et al. Evaluating the Use of Circulating MicroRNA Profiles for Lung Cancer Detection in Symptomatic Patients. JAMA Oncol 2020;6:714-23. [Crossref] [PubMed]
  183. Ulivi P, Petracci E, Marisi G, et al. Prognostic Role of Circulating miRNAs in Early-Stage Non-Small Cell Lung Cancer. J Clin Med 2019;8:131. [Crossref] [PubMed]
  184. Sun L, Zhou H, Yang Y, et al. Meta-analysis of diagnostic and prognostic value of miR-126 in non-small cell lung cancer. Biosci Rep 2020;40:BSR20200349. [Crossref] [PubMed]
Cite this article as: Tomasik B, Skrzypski M, Bieńkowski M, Dziadziuszko R, Jassem J. Current and future applications of liquid biopsy in non-small-cell lung cancer—a narrative review. Transl Lung Cancer Res 2023;12(3):594-614. doi: 10.21037/tlcr-22-742

Download Citation