Abstract
Non-small cell lung cancer (NSCLC) is the leading cause of cancer-related death in both men and women. The ability of cancer cells to break-off from the primary tumor and spread to distant organs is the main cause of death of cancer patients. The detection of circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA) is a considerable part of liquid biopsy, which contributes to the diagnosis, treatment and prognosis, and especially to identify the targetable mutations of NSCLC. This review is to discuss the detection and application of CTC and ctDNA in the diagnosis, prognostic evaluation and guiding targeted therapy of NSCLC.
Keywords: Non-small cell lung cancer (NSCLC), circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), liquid biopsy, targeted therapy
Introduction
Lung cancer, with 80-85% being non-small cell lung cancer (NSCLC), is the leading cause of cancer-related death in both men and women [1-3]. More than one-third of all newly diagnosed lung cancers occurred in China, making it a large social and economic burden. According to the annual report on the status of cancer in China, in total, 651,053 patients were newly diagnosed with lung cancer in 2011, including 441,364 men and 209,689 women [4-6].
The ability of cancer cells to break-off from the primary tumor and spread to distant organs is the main cause of death in cancer patients. The detection of circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA) serving as liquid biopsies is novel developed technology [7]. Quantification of genetic mutations using CTCs and ctDNA may provide a noninvasive means for early cancer detection and disease monitoring. Compared with tissue biopsy, liquid biopsies are far less invasive and more repeatable [7-9]. Furthermore, because of the high degree of heterogeneity among tumor cells in different regions, liquid biopsy can provide more comprehensive tumor mutation analysis results [10]. CTCs and ctDNA as the cornerstones of liquid biopsy diagnostics have their own advantages and can complement each other. CTCs analysis can provide the information of cancer DNA, RNA and protein, whereas ctDNA can only provide genetic abnormality information. These information can help clinicians to identify drug resistance mutations, minimum residual disease after treatment, treatment progress. Although the CTCs have more information available, ctDNA is easier to save and can be used for a longer time [7,9]. In the development of personalized medicine, CTCs and ctDNA play a crucial part in the diagnosis, treatment and prognosis of non-small cell lung cancer [11-13].
Detection of CTCs and ctDNA in NSCLC
Detection of CTCs for NSCLC
CTCs are tumor cells shedding from either primary tumors or its metastases that circulate in the peripheral blood [14]. These cells can be divided into single CTCs and CTC clusters, with the latter being defined as groups of tumor cells (exceed two or three cells, varied among previous researches) that march in the bloodstream [15] (Figure 1A). In the early stage of tumor formation, tumor cells can be extensively shed into the circulatory system [14]. The CTCs can be detected in the blood of patients with only primary tumor and no metastasis, and the correlation between CTCs count and cancer prognosis is observed [16]. Clinical trials and clinical data have shown that CTCs can still be detected in cancer patients after their primary tumors have been removed for several months or even years, indicating that tumor cells can be re-disseminated from the metastases into the blood [9,17].
Figure 1.

Formation mechanism of CTCs and ctDNA. A: Circulating tumor cells (CTCs) are shed from primary tumors or its metastases; CTC clusters can also be generated by single CTCs aggregation or proliferation; B: Circulating tumor DNA (ctDNA) is released by necrotic cells; C: Active cells (including CTC) actively secrete DNA; D: CtDNA is released by apoptotic cells.
Currently, the CellSearch system (Janssen Diagnostics Company, USA) is the only FDA-approved assay for CTCs detection. Since the CellSearch system detects CTCs based on their expression of epithelial biomarkers which are likely to be lost in CTCs due to epithelial-mesenchymal transition (EMT) during circulating, the detection rate for CTCs is limited [18]. Many other strategies are proposed and developed. Herringbone-chip or “HB-Chip” creates an enhanced platform for CTCs insulation, which increases the capture rate of CTCs [19]. A microcavity array (MCA) system has been developed by Hosokawa M et al. and was demonstrated to have higher detection sensitivity for NSCLC than the CellSearch system [20]. An approach named isolation by size of epithelial tumor cell (ISET) can capture all the circulating non-haematological cells (CNHCs) and characterize as the CTCs among them [21]. Also many flow cytometry and chip based systems have been developed, including FISHMAN-R, On-chip Sort (On-Chip Biotechnologies) and microfluidic chips [22-24].
Detection of ctDNA for NSCLC
The cell-free DNA (cfDNA) level in the blood increases due to various pathological processes. Both benign and malignant lesions may release their DNA into the blood through apoptosis and necrosis, forming cfDNA (Figure 1B and 1D). Although cfDNA may be elevated in a variety of pathological processes, cancer patients have a greater increase than those patients with benign lesions [25]. Higher concentrations of cfDNA often were measured in the plasma of cancer patients, referring as circulating tumor DNA (ctDNA). Another view about the origin of cfDNA is that cells actively secrete DNA, which has nothing to do with apoptosis or necrosis (Figure 1C). Since all cells release cfDNA, the cfDNA in the blood of cancer patients may originate from the cancer cells or other non-cancerous cells [26]. Some scholars believe the two mechanisms mentioned above are both the main sources of cfDNA, and they are characterized as passive and active respectively [27].
CtDNA detection technology has went through many development periods, from the conventional karyotype analysis to the various molecular cytogenetic methods such as spectral karyotyping, chromosome-based comparative genomic hybridization (CGH) and fluorescence in situ hybridization (FISH), and to the modern technologies including microarray-based CGH and single nucleotide polymorphism (SNP) analysis and next-generation sequencing (NGS) [28]. Currently, detecting ctDNA is clinically practical. By sending ctDNA and tumor tissue of the same patients to NGS at the same time, researchers found that the concordance rate for clinically actionable DNA alteration detection in the two samples can reach 54.9% with a sensitivity of 53.2% and a specificity of 75.0% [29]. NGS has been widely used in clinics as routine for personalized therapy, and it can detect multiple mutations at the same time [30,31]. Many PCR-based assays are also potential methods for detecting genotype mutations in plasma, such as microfluidics digital PCR and droplet digital PCR (ddPCR) which have all been applied to the detection of EGFR mutations in NSCLC [32,33].
CTCs and ctDNA can be used as the diagnostic biomarkers of NSCLC
CTCs and ctDNA are emerging as noninvasive multifunctional biomarkers in liquid biopsy. Lung cancer is the most common cause of cancer-related death worldwide [34], and the five-year survival rate of lung cancer varies from 4% to 17% depending on geographic and stage differences [35]. The lack of clinical symptoms in early stage is the crucial reason for this extremely low survival rate of lung cancer. In addition, physicians never practice a biopsy in the patients without any clinical symptom. Early lung cancer patients have far better prognosis versus advanced patients. For these reasons, exploring and researching biomarkers for early diagnosis of lung cancer is an inevitable trend [36,37].
CTCs as the diagnostic biomarkers of NSCLC
Tanaka F et al. found that the quantity of CTCs in patients with lung cancer was much higher than those without malignant disease, and increased CTCs significantly associated with the progression or metastasis of lung cancer. Therefore, CTCs counts may be useful for the diagnosis and metastasis of primary lung cancer [38].
Ilie M et al. examined the presence of CTCs in 245 patients without cancer including 168 chronic obstructive pulmonary disease (COPD) patients and 77 subjects without COPD by using ISET technology, CTCs were detected in 5 COPD patients. These patients are all diagnosed with pulmonary nodules in the one to four years after the detection of CTCs, and the pathological diagnosis after surgery was confirmed as early stage lung cancer. Neither recurrence nor CTCs were found at 16 months after operation because of early diagnosis and intervention. This result preliminarily confirmed the potential of CTCs testing for the early diagnosis of lung cancer in high-risk patients [39]. Many studies have demonstrated that Folate Receptor-positive CTCs can be used as a biomarker for the early diagnosis and progress monitoring in NSCLC patients [40,41].
ctDNA as the diagnostic biomarkers of NSCLC
Some studies have found that circulating DNA in plasma of NSCLC patients is higher than normal subjects. By sequencing specific gene regions with PCR, the proportion of tumor derived DNA in all ctDNA can be determined. Also, some studies found that NSCLC patients with lymph node metastasis or distance metastasis had higher circulating DNA levels, and those patients with higher circulating DNA levels had less overall survival time. These findings indicated that circulating DNA can act as a potential diagnostic marker for NSCLC [42-44].
At the present stage, the diagnosis and treatment of cancer is greatly depending on molecular detection of tumor-drived genes, but limited tumor tissue has made it difficult to obtain molecular detection data. The proposal to replace tumor DNA with ctDNA for molecular phenotype determing has gradually been taken seriously. EGFR mutation status is an important biomarker for NSCLC targeted therapy. It is, therefore, helpful to have an early diagnosis about mutation type for receiving optimal targeted therapy. Studies have shown that ctDNA is capable of detecting epidermal growth factor receptor (EGFR) mutations in ctDNA of patients with NSCLC [45-47].
CTCs and ctDNA can act as prognostic biomarkers of NSCLC
Although the prognosis of NSCLC patients has been greatly improved with the development of targeted therapies, the overall prognosis of NSCLC remains poor. Blood circulation markers can be an effective indicator about the prognosis and treatment response of lung cancer patients [48,49].
CTCs as prognostic biomarkers of NSCLC
As a minimally invasive test, CTCs can be conveniently acquired for many times, thus can help patients to customize individual treatment plans, monitor treatment efficacy and observe patients’ prognosis [51]. Krebs et al. conducted a single-center prospective study, detecting CTCs in untreated patients with late stage NSCLC. CTCs number detected in stage IV patients is significantly higher than in stage III patients. Moreover, the CTCs counts are positively related to overall survival (OS), suggesting CTCs can act as a novel prognostic factor of NSCLC [52]. Spiliotaki et al. have also demonstrated that monitoring the proliferation and apoptosis in CTCs can be a useful tool for long-term follow-up of cancer patients [53].
Multivariate analyses showed that the number of CTCs is significantly related to shorter disease-free survival (DFS) and progression-free survival (PFS), indicating CTCs can be an independent prognostic factor and an efficacy predictor for lung cancer [54,55]. CTC clusters are more invasive than single CTCs, and patients who can be detected with clustered CTCs have a worse prognosis than patients with single CTCs only [56,57].
ctDNA as prognostic biomarkers of NSCLC
A study in India demonstrated that the EGFR mutation status detected in tissue biopsies and ctDNA were highly consistent, ctDNA not only can be used as an important biomarker for cancer prognosis, but also it could be used for early diagnosis and treatment response assessment [58]. The concentration of ctDNA has a strong positive relation with cancer prognosis, with ctDNA level increasing when cancer progresses and declining after effective treatment [13]. Molecular tests are commonly used as companion diagnostics to help adjust treatment regimens and improve prognosis. The ctDNA can also be used to dynamically monitor EGFR mutations to improve patient care [59].
Another research conducted a prospective trial to measure ctDNA and tumor volume with PET-CT in advanced NSCLC patients and tracked their prognosis. Results revealed that ctDNA level has high correlation with OS but has no correlation with tumor burden [60]. Sirera et al. tracked the EGFR mutation status in plasma ctDNA in 45 NSCLC patients who are receiving EGFR targeted therapy. They found that EGFR mutation status in ctDNA changed in 26 cases, and the T790M negative to positive alteration group has a shorter PFS [61]. Another large-scale research also proved that high ctDNA level is an independent poor prognostic biomarker for advanced NSCLC, and it may be useful for improving the prognosis [62].
Detection of CTC and ctDNA can be used to guide the targeted therapy of NSCLC
In recent years, more and more attention has been paid to precision medicine, and cancer treatment is also more inclined to molecular targeted therapy which can improve the prognosis of cancer patients and enhance PFS and overall survival (OS) [63,64]. The key to targeted therapy is to find targetable activating mutations. Tissue biopsy is gradually replaced by liquid biopsy for mutation detection. Detectable genetic alterations of NSCLC patients include not only common mutations such as EGFR, ALK, and ROS-1 but rare driver mutations like MET amplification, RET rearrangement, and HER-2 insertion. These mutations are the widely used therapeutic target to treat patients with NSCLC [65]. Some assessments based on the results of targeted therapy for patients with NSCLC suggest that CTC and ctDNA analysis are effective means for identifying genotyping of tumors, especially if tissue biopsy is not feasible [66-68]. They are also important non-invasive tools for assessing early treatment response and monitoring mutation status in real time [69-71]. And ctDNA is more sensitive than CTC in predicting and monitoring the treatment effectiveness for mutation harboring lung cancer [72].
CTC in the targeted therapy of NSCLC
The use of CTC counts in targeted therapy of NSCLC is very common. 43 cases of NSCLC patients with EGFR mutation or ALK rearrangement were classified by CTC counts. Then the prognostic analysis showed that baseline CTC counts can be used as predictive prognosis biomarkers for EGFR mutations and ALK rearrangement of NSCLC, and can better guide patients in drug therapy and monitor patient’s prognosis in targeted therapy [73]. Chang et al. developed a parallel flow micro-aperture chip system for detection of CTCs and found that CTC counts in untreated patients in statically higher than treated patients with NSCLC (detailed comparison) [74].
In addition, some researchers compared the number of CTCs in patients with NSCLC and those without lung cancer finding that CTCs can only be found in NSCLC group and CTCs positive detection rate obviously rises as the pathological stage increases from I to IV. Comparing pre-treatment and post chemotherapy CTCs counts, they found that CTCs counts decreased significantly after two courses of chemotherapy. The decreasing in CTCs counts are also positively relative to the treatment efficacy [75,76]. Other investigators have developed a microfluidic device to enrich CTCs, which not only facilitates the detection of genetic mutations in individual cells, but also dynamically monitors the genetic aberrations of patients during treatment and explores possible drug-resistant mutations in patients [77].
ctDNA in the targeted therapy of NSCLC
The detection of ctDNA can be qualitative or quantitative. Qualitative testing is mainly used to detect genetic subtypes (EGFR, ALK, KRAS, ROS1, et al.) of NSCLC patients. Therefore, liquid biopsies utilizing ctDNA testing represent a powerful approach to detect cancer genotype and monitor the development of resistance [78,79]. Thompson’s team used NGS to compare ctDNA with tissue genome sequence, demonstrating that ctDNA NGS was feasible for the detection of targeted driver and resistance mutations in NSCLC patients [80]. Particularly, for patients with unusable or inadequate tumor tissue, ctDNA detection may be the first choice for detecting drive mutations and resistance mutations (especially forthe EGFR mutation) [81,82].
Quantitative testing, on the other hand, is used to predict the cancer response and progression and to assess efficacy and prognosis [83]. The concentration of ctDNA is related to the response and disease progression, reflecting its ability to act as a biomarker [84]. Quantitative ctDNA detection commonly used digital PCR to quantify ctDNA mutations in plasma. The German researchers used digital PCR to quantify EGFR and KRAS mutations in circulating DNA, and monitored changes in the level of DNA mutations in the plasma of the subjects over time, and confirmed the correlation between ctDNA concentration and tumor progression [85]. Other researches also confirmed these results [86,87] (Table 1).
Table 1.
Compare the differences between circulating tumor cell (CTCs) and circulating tumor DNA (ctDNA)
| Comparison | CTCs | ctDNA |
|---|---|---|
| Definition | Tumor cells are released into the blood from primary tumors and/or metastatic sites [9] | High concentration of cell-free DNA in tumor patients [24] |
| Common detection technology | CellSearch method (Janssen Diagnostics Company, USA) [17]/FISHMAN-R/On-chip Sort [20,21] | Conventional approach (karyotype analysis) |
| Molecular cytogenetic methods (spectral karyotyping, CGH and FISH) | ||
| Modern technologies (microarray-based CGH, SNP, NGS) [26] | ||
| PCR-based methods (microfluidics digital PCR, ddPCR [30,31] | ||
| Information type providing by analysis | DNA, RNA and protein [7,9] | DNA [7,9] |
| As diagnostic biomarkers | CTC counts contributes to the judgment of benign and malignant lung diseases [36,37] | Prone to the diagnosis of molecular subtypes of lung cancer [40-45] |
| As prognostic biomarkers | Assess the change of patient’s condition by monitoring the proliferation and apoptosis changes of CTCs [51] | CtDNA A can be used as an independent prognostic marker [13,56,58] |
| The number of CTCs is significantly related to DFS and PFS [52,53] | Different mutation status of the same subtype has a different prognosis [57] | |
| Roles of CTC and ctDNA in targeted therapy | Baseline CTC counts can be used to predict and monitor the efficacy of targeted therapies [69] | Monitor mutation status to guide the selection of targeted drugs [65-67] |
Conclusions and futures
CTCs and ctDNA detecting are gradually developed to play roles in the diagnosis, treatment, and prognosis assessment of NSCLC. More importantly, CTCs and ctDNA are used to detect the mutation status and gene copy number of the EGFR gene in NSCLC patients which could benefit patients by providing a more convenient and dynamic monitoring for targeted therapy and improving prognosis.
With the development of liquid biopsy, CTCs and ctDNA detection have also received increasing attention in other body fluids (such as urine, cerebrospinal fluid) besides blood, and will better help diagnose and treat NSCLC and prolong the PFS of patients. In the future, further studies would be concentrated on the following aspects: (1) developing more convenient and applicable CTCs and ctDNA detection methods (2) applying CTCs and ctDNA detection in other body fluids samples (3) discovering more driver mutations using CTCs and ctDNA (4) finding more targeted drugs with higher sensitivity and specificity by tracking CTCs and ctDNA (5) further exploration of the relationship between CTCs and ctDNA detection and targeted therapy.
Acknowledgements
The work was supported by grants from The National Natural Sciences Foundations of China (No. 81472773, 81773218, 81703009 and 81272566).
Disclosure of conflict of interest
None.
References
- 1.Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394–424. doi: 10.3322/caac.21492. [DOI] [PubMed] [Google Scholar]
- 2.Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA Cancer J Clin. 2015;65:87–108. doi: 10.3322/caac.21262. [DOI] [PubMed] [Google Scholar]
- 3.Ahmad A. “Lung cancer and personalized medicine current knowledge and therapies”. Springer International Publishing; 2015. [Google Scholar]
- 4.Chen W, Zheng R, Zeng H, Zhang S, He J. Annual report on status of cancer in China, 2011. Chin J Cancer Res. 2015;27:2–12. doi: 10.3978/j.issn.1000-9604.2015.01.06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Pan R, Zhu M, Yu C, Lv J, Guo Y, Bian Z, Yang L, Chen Y, Hu Z, Chen Z, Li L, Shen H China Kadoorie Biobank Collaborative Group. Cancer incidence and mortality: a cohort study in China, 2008-2013. Int J Cancer. 2017;141:1315–1323. doi: 10.1002/ijc.30825. [DOI] [PubMed] [Google Scholar]
- 6.Chen W, Zheng R, Zhang S, Zeng H, Xia C, Zuo T, Yang Z, Zou X, He J. Cancer incidence and mortality in China, 2013. Cancer Lett. 2017;401:63–71. doi: 10.1016/j.canlet.2017.04.024. [DOI] [PubMed] [Google Scholar]
- 7.Lowes LE, Bratman SV, Dittamore R, Done S, Kelley SO, Mai S, Morin RD, Wyatt AW, Allan AL. Circulating tumor cells (CTC) and cell-free DNA (cfDNA) workshop 2016: scientific opportunities and logistics for cancer clinical trial incorporation. Int J Mol Sci. 2016;17 doi: 10.3390/ijms17091505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Sundaresan TK, Sequist LV, Heymach JV, Riely GJ, Jänne PA, Koch WH, Sullivan JP, Fox DB, Maher R, Muzikansky A, Webb A, Tran HT, Giri U, Fleisher M, Yu HA, Wei W, Johnson BE, Barber TA, Walsh JR, Engelman JA, Stott SL, Kapur R, Maheswaran S, Toner M, Haber DA. Detection of T790M, the acquired resistance EGFR mutation, by tumor biopsy versus noninvasive blood-based analyses. Clin Cancer Res. 2016;22:1103–1110. doi: 10.1158/1078-0432.CCR-15-1031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Alix-Panabières C, Pantel K. Clinical applications of circulating tumor cells and circulating tumor DNA as liquid biopsy. Cancer Discov. 2016;6:479–491. doi: 10.1158/2159-8290.CD-15-1483. [DOI] [PubMed] [Google Scholar]
- 10.Gerlinger M, Rowan AJ, Horswell S, Math M, Larkin J, Endesfelder D, Gronroos E, Martinez P, Matthews N, Stewart A, Tarpey P, Varela I, Phillimore B, Begum S, McDonald NQ, Butler A, Jones D, Raine K, Latimer C, Santos CR, Nohadani M, Eklund AC, Spencer-Dene B, Clark G, Pickering L, Stamp G, Gore M, Szallasi Z, Downward J, Futreal PA, Swanton C. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med. 2012;366:883–892. doi: 10.1056/NEJMoa1113205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Isobe K, Hata Y, Kobayashi K, Hirota N, Sato K, Sano G, Sugino K, Sakamoto S, Takai Y, Shibuya K, Takagi K, Homma S. Clinical significance of circulating tumor cells and free DNA in non-small cell lung cancer. Anticancer Res. 2012;32:3339–3344. [PubMed] [Google Scholar]
- 12.Maheswaran S, Sequist LV, Nagrath S, Ulkus L, Brannigan B, Collura CV, Inserra E, Diederichs S, Iafrate AJ, Bell DW, Digumarthy S, Muzikansky A, Irimia D, Settleman J, Tompkins RG, Lynch TJ, Toner M, Haber DA. Detection of mutations in EGFR in circulating lung-cancer cells. N Engl J Med. 2008;359:366–377. doi: 10.1056/NEJMoa0800668. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Jiang T, Ren S, Zhou C. Role of circulating-tumor DNA analysis in non-small cell lung cancer. Lung Cancer. 2015;90:128–134. doi: 10.1016/j.lungcan.2015.09.013. [DOI] [PubMed] [Google Scholar]
- 14.Kim MY, Oskarsson T, Acharyya S, Nguyen DX, Zhang XH, Norton L, Massagué J. Tumor self-seeding by circulating cancer cells. Cell. 2009;139:1315–1326. doi: 10.1016/j.cell.2009.11.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Hong Y, Fang F, Zhang Q. Circulating tumor cell clusters: what we know and what we expect (Review) Int J Oncol. 2016;49:2206–2216. doi: 10.3892/ijo.2016.3747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Pantel K, Alix-Panabières C. Real-time liquid biopsy in cancer patients: fact or fiction? Cancer Res. 2013;73:6384–6388. doi: 10.1158/0008-5472.CAN-13-2030. [DOI] [PubMed] [Google Scholar]
- 17.Meng S, Tripathy D, Frenkel EP, Shete S, Naftalis EZ, Huth JF, Beitsch PD, Leitch M, Hoover S, Euhus D, Haley B, Morrison L, Fleming TP, Herlyn D, Terstappen LW, Fehm T, Tucker TF, Lane N, Wang J, Uhr JW. Circulating tumor cells in patients with breast cancer dormancy. Clin Cancer Res. 2004;10:8152–8162. doi: 10.1158/1078-0432.CCR-04-1110. [DOI] [PubMed] [Google Scholar]
- 18.Ilie M, Hofman V, Long E, Bordone O, Selva E, Washetine K, Marquette CH, Hofman P. Current challenges for detection of circulating tumor cells and cell-free circulating nucleic acids, and their characterization in non-small cell lung carcinoma patients. What is the best blood substrate for personalized medicine? Ann Transl Med. 2014;2:107. doi: 10.3978/j.issn.2305-5839.2014.08.11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Stott SL, Hsu CH, Tsukrov DI, Yu M, Miyamoto DT, Waltman BA, Rothenberg SM, Shah AM, Smas ME, Korir GK, Floyd FP Jr, Gilman AJ, Lord JB, Winokur D, Springer S, Irimia D, Nagrath S, Sequist LV, Lee RJ, Isselbacher KJ, Maheswaran S, Haber DA, Toner M. Isolation of circulating tumor cells using a microvortex-generating herringbone-chip. Proc Natl Acad Sci U S A. 2010;107:18392–18397. doi: 10.1073/pnas.1012539107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Hosokawa M, Kenmotsu H, Koh Y, Yoshino T, Yoshikawa T, Naito T, Takahashi T, Murakami H, Nakamura Y, Tsuya A, Shukuya T, Ono A, Akamatsu H, Watanabe R, Ono S, Mori K, Kanbara H, Yamaguchi K, Tanaka T, Matsunaga T, Yamamoto N. Size-based isolation of circulating tumor cells in lung cancer patients using a microcavity array system. PLoS One. 2013;8:e67466. doi: 10.1371/journal.pone.0067466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Hofman V, Long E, Ilie M, Bonnetaud C, Vignaud JM, Fléjou JF, Lantuejoul S, Piaton E, Mourad N, Butori C, Selva E, Marquette CH, Poudenx M, Sibon S, Kelhef S, Vénissac N, Jais JP, Mouroux J, Molina TJ, Vielh P, Hofman P. 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;23:30–38. doi: 10.1111/j.1365-2303.2010.00835.x. [DOI] [PubMed] [Google Scholar]
- 22.Sawada T, Watanabe M, Fujimura Y, Yagishita S, Shimoyama T, Maeda Y, Kanda S, Yunokawa M, Tamura K, Tamura T, Minami H, Koh Y, Koizumi F. Sensitive cytometry based system for enumeration, capture and analysis of gene mutations of circulating tumor cells. Cancer Sci. 2016;107:307–314. doi: 10.1111/cas.12868. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Watanabe M, Serizawa M, Sawada T, Takeda K, Takahashi T, Yamamoto N, Koizumi F, Koh Y. A novel flow cytometry-based cell capture platform for the detection, capture and molecular characterization of rare tumor cells in blood. J Transl Med. 2014;12:143. doi: 10.1186/1479-5876-12-143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Sun D, Chen Z, Wu M, Zhang Y. Nanomaterial-based microfluidic chips for the capture and detection of circulating tumor cells. Nanotheranostics. 2017;1:389–402. doi: 10.7150/ntno.21268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Schwarzenbach H, Hoon DS, Pantel K. Cell-free nucleic acids as biomarkers in cancer patients. Nat Rev Cancer. 2011;11:426–437. doi: 10.1038/nrc3066. [DOI] [PubMed] [Google Scholar]
- 26.Bronkhorst AJ, Wentzel JF, Aucamp J, van Dyk E, du Plessis L, Pretorius PJ. Characterization of the cell-free DNA released by cultured cancer cells. Biochim Biophys Acta. 2016;1863:157–165. doi: 10.1016/j.bbamcr.2015.10.022. [DOI] [PubMed] [Google Scholar]
- 27.Stroun M, Lyautey J, Lederrey C, Olson-Sand A, Anker P. About the possible origin and mechanism of circulating DNA apoptosis and active DNA release. Clin Chim Acta. 2001;313:139–142. doi: 10.1016/s0009-8981(01)00665-9. [DOI] [PubMed] [Google Scholar]
- 28.Ma M, Zhu H, Zhang C, Sun X, Gao X, Chen G. “Liquid biopsy”-ctDNA detection with great potential and challenges. Ann Transl Med. 2015;3:235. doi: 10.3978/j.issn.2305-5839.2015.09.29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Liu L, Liu H, Shao D, Liu Z, Wang J, Deng Q, Tang H, Yang H, Zhang Y, Qiu Y, Cui F, Tan M, Zhang P, Li Z, Liu J, Liang W, Wang Y, Peng Z, Wang J, Yang H, Mao M, Kristiansen K, Ye M, He J. Development and clinical validation of a circulating tumor DNA test for the identification of clinically actionable mutations in nonsmall cell lung cancer. Genes Chromosomes Cancer. 2018;57:211–220. doi: 10.1002/gcc.22522. [DOI] [PubMed] [Google Scholar]
- 30.Tran B, Brown AM, Bedard PL, Winquist E, Goss GD, Hotte SJ, Welch SA, Hirte HW, Zhang T, Stein LD, Ferretti V, Watt S, Jiao W, Ng K, Ghai S, Shaw P, Petrocelli T, Hudson TJ, Neel BG, Onetto N, Siu LL, McPherson JD, Kamel-Reid S, Dancey JE. Feasibility of real time next generation sequencing of cancer genes linked to drug response: results from a clinical trial. Int J Cancer. 2013;132:1547–1555. doi: 10.1002/ijc.27817. [DOI] [PubMed] [Google Scholar]
- 31.Toor OM, Ahmed Z, Bahaj W, Boda U, Cummings LS, McNally ME, Kennedy KF, Pluard TJ, Hussain A, Subramanian J, Masood A. Correlation of somatic genomic alterations between tissue genomics and ctDNA employing next-generation sequencing: analysis of lung and gastrointestinal cancers. Mol Cancer Ther. 2018;17:1123–1132. doi: 10.1158/1535-7163.MCT-17-1015. [DOI] [PubMed] [Google Scholar]
- 32.Yung TK, Chan KC, Mok TS, Tong J, To KF, Lo YM. Single-molecule detection of epidermal growth factor receptor mutations in plasma by microfluidics digital PCR in non-small cell lung cancer patients. Clin Cancer Res. 2009;15:2076–2084. doi: 10.1158/1078-0432.CCR-08-2622. [DOI] [PubMed] [Google Scholar]
- 33.Hindson BJ, Ness KD, Masquelier DA, Belgrader P, Heredia NJ, Makarewicz AJ, Bright IJ, Lucero MY, Hiddessen AL, Legler TC, Kitano TK, Hodel MR, Petersen JF, Wyatt PW, Steenblock ER, Shah PH, Bousse LJ, Troup CB, Mellen JC, Wittmann DK, Erndt NG, Cauley TH, Koehler RT, So AP, Dube S, Rose KA, Montesclaros L, Wang S, Stumbo DP, Hodges SP, Romine S, Milanovich FP, White HE, Regan JF, Karlin-Neumann GA, Hindson CM, Saxonov S, Colston BW. High-throughput droplet digital PCR system for absolute quantitation of DNA copy number. Anal Chem. 2011;83:8604–8610. doi: 10.1021/ac202028g. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D, Bray F. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136:E359–386. doi: 10.1002/ijc.29210. [DOI] [PubMed] [Google Scholar]
- 35.Hirsch FR, Scagliotti GV, Mulshine JL, Kwon R, Curran WJ Jr, Wu YL, Paz-Ares L. Lung cancer: current therapies and new targeted treatments. Lancet. 2017;389:299–311. doi: 10.1016/S0140-6736(16)30958-8. [DOI] [PubMed] [Google Scholar]
- 36.Heuvers ME, Hegmans JP, Stricker BH, Aerts JG. Improving lung cancer survival; time to move on. BMC Pulm Med. 2012;12:77. doi: 10.1186/1471-2466-12-77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Tang JH, Chia D. Liquid biopsies in the screening of oncogenic mutations in NSCLC and its application in targeted therapy. Crit Rev Oncog. 2015;20:357–371. doi: 10.1615/CritRevOncog.v20.i5-6.90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Tanaka F, Yoneda K, Kondo N, Hashimoto M, Takuwa T, Matsumoto S, Okumura Y, Rahman S, Tsubota N, Tsujimura T, Kuribayashi K, Fukuoka K, Nakano T, Hasegawa S. Circulating tumor cell as a diagnostic marker in primary lung cancer. Clin Cancer Res. 2009;15:6980–6986. doi: 10.1158/1078-0432.CCR-09-1095. [DOI] [PubMed] [Google Scholar]
- 39.Ilie M, Hofman V, Long-Mira E, Selva E, Vignaud JM, Padovani B, Mouroux J, Marquette CH, Hofman P. “Sentinel” circulating tumor cells allow early diagnosis of lung cancer in patients with chronic obstructive pulmonary disease. PLoS One. 2014;9:e111597. doi: 10.1371/journal.pone.0111597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Yu Y, Chen Z, Dong J, Wei P, Hu R, Zhou C, Sun N, Luo M, Yang W, Yao R, Gao Y, Li J, Yang G, He W, He J. Folate receptor-positive circulating tumor cells as a novel diagnostic biomarker in non-small cell lung cancer. Transl Oncol. 2013;6:697–702. doi: 10.1593/tlo.13535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Wang L, Wu C, Qiao L, Yu W, Guo Q, Zhao M, Yang G, Zhao H, Lou J. Clinical significance of folate receptor-positive circulating tumor cells detected by ligand-targeted polymerase chain reaction in lung cancer. J Cancer. 2017;8:104–110. doi: 10.7150/jca.16856. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Ulivi P, Mercatali L, Casoni GL, Scarpi E, Bucchi L, Silvestrini R, Sanna S, Monteverde M, Amadori D, Poletti V, Zoli W. Multiple marker detection in peripheral blood for NSCLC diagnosis. PLoS One. 2013;8:e57401. doi: 10.1371/journal.pone.0057401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Jahr S, Hentze H, Englisch S, Hardt D, Fackelmayer FO, Hesch RD, Knippers R. DNA fragments in the blood plasma of cancer patients: quantitations and evidence for their origin from apoptotic and necrotic cells. Cancer Res. 2001;61:1659–1665. [PubMed] [Google Scholar]
- 44.Catarino R, Coelho A, Araújo A, Gomes M, Nogueira A, Lopes C, Medeiros R. Circulating DNA: diagnostic tool and predictive marker for overall survival of NSCLC patients. PLoS One. 2012;7:e38559. doi: 10.1371/journal.pone.0038559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Ulivi P, Silvestrini R. Role of quantitative and qualitative characteristics of free circulating DNA in the management of patients with non-small cell lung cancer. Cell Oncol (Dordr) 2013;36:439–448. doi: 10.1007/s13402-013-0155-3. [DOI] [PubMed] [Google Scholar]
- 46.Luo J, Shen L, Zheng D. Diagnostic value of circulating free DNA for the detection of EGFR mutation status in NSCLC: a systematic review and meta-analysis. Sci Rep. 2014;4:6269. doi: 10.1038/srep06269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Normanno N, Denis MG, Thress KS, Ratcliffe M, Reck M. Guide to detecting epidermal growth factor receptor (EGFR) mutations in ctDNA of patients with advanced non-small-cell lung cancer. Oncotarget. 2017;8:12501–12516. doi: 10.18632/oncotarget.13915. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Xu-Welliver M, Carbone DP. Blood-based biomarkers in lung cancer: prognosis and treatment decisions. Transl Lung Cancer Res. 2017;6:708–712. doi: 10.21037/tlcr.2017.09.08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Syrigos K, Fiste O, Charpidou A, Grapsa D. Circulating tumor cells count as a predictor of survival in lung cancer. Crit Rev Oncol Hematol. 2018;125:60–68. doi: 10.1016/j.critrevonc.2018.03.004. [DOI] [PubMed] [Google Scholar]
- 50.Jia J, Huang B, Zhuang Z, Chen S. Circulating tumor DNA as prognostic markers for late stage NSCLC with bone metastasis. Int J Biol Markers. 2018;33:222–230. doi: 10.1177/1724600817753576. [DOI] [PubMed] [Google Scholar]
- 51.Lianidou ES, Markou A, Strati A. The role of CTCs as tumor biomarkers. Adv Exp Med Biol. 2015;867:341–367. doi: 10.1007/978-94-017-7215-0_21. [DOI] [PubMed] [Google Scholar]
- 52.Krebs MG, Sloane R, Priest L, Lancashire L, Hou JM, Greystoke A, Ward TH, Ferraldeschi R, Hughes A, Clack G, Ranson M, Dive C, Blackhall FH. Evaluation and prognostic significance of circulating tumor cells in patients with non-small-cell lung cancer. J. Clin. Oncol. 2011;29:1556–1563. doi: 10.1200/JCO.2010.28.7045. [DOI] [PubMed] [Google Scholar]
- 53.Spiliotaki M, Mavroudis D, Kapranou K, Markomanolaki H, Kallergi G, Koinis F, Kalbakis K, Georgoulias V, Agelaki S. Evaluation of proliferation and apoptosis markers in circulating tumor cells of women with early breast cancer who are candidates for tumor dormancy. Breast Cancer Res. 2014;16:485. doi: 10.1186/s13058-014-0485-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Gallo M, De Luca A, Maiello MR, D’Alessio A, Esposito C, Chicchinelli N, Forgione L, Piccirillo MC, Rocco G, Morabito A, Botti G, Normanno N. Clinical utility of circulating tumor cells in patients with non-small-cell lung cancer. Transl Lung Cancer Res. 2017;6:486–498. doi: 10.21037/tlcr.2017.05.07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Truini A, Alama A, Dal Bello MG, Coco S, Vanni I, Rijavec E, Genova C, Barletta G, Biello F, Grossi F. Clinical applications of circulating tumor cells in lung cancer patients by cell search system. Front Oncol. 2014;4:242. doi: 10.3389/fonc.2014.00242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Cheung KJ, Ewald AJ. A collective route to metastasis: seeding by tumor cell clusters. Science. 2016;352:167–169. doi: 10.1126/science.aaf6546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Murlidhar V, Reddy RM, Fouladdel S, Zhao L, Ishikawa MK, Grabauskiene S, Zhang Z, Lin J, Chang AC, Carrott P, Lynch WR, Orringer MB, Kumar-Sinha C, Palanisamy N, Beer DG, Wicha MS, Ramnath N, Azizi E, Nagrath S. Poor prognosis indicated by venous circulating tumor cell clusters in early-stage lung cancers. Cancer Res. 2017;77:5194–5206. doi: 10.1158/0008-5472.CAN-16-2072. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Veldore VH, Choughule A, Routhu T, Mandloi N, Noronha V, Joshi A, Dutt A, Gupta R, Vedam R, Prabhash K. Validation of liquid biopsy: plasma cell-free DNA testing in clinical management of advanced non-small cell lung cancer. Lung Cancer (Auckl) 2018;9:1–11. doi: 10.2147/LCTT.S147841. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Liang Z, Cheng Y, Chen Y, Hu Y, Liu WP, Lu Y, Wang J, Wang Y, Wu G, Ying JM, Zhang HL, Zhang XC, Wu YL. EGFR T790M ctDNA testing platforms and their role as companion diagnostics: correlation with clinical outcomes to EGFR-TKIs. Cancer Lett. 2017;403:186–194. doi: 10.1016/j.canlet.2017.06.008. [DOI] [PubMed] [Google Scholar]
- 60.Nygaard AD, Holdgaard PC, Spindler KL, Pallisgaard N, Jakobsen A. The correlation between cell-free DNA and tumour burden was estimated by PET/CT in patients with advanced NSCLC. Br J Cancer. 2014;110:363–368. doi: 10.1038/bjc.2013.705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Zhang C, Wei B, Li P, Yang K, Wang Z, Ma J, Guo Y. Prognostic value of plasma EGFR ctDNA in NSCLC patients treated with EGFR-TKIs. PLoS One. 2017;12:e0173524. doi: 10.1371/journal.pone.0173524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Sirera R, Bremnes RM, Cabrera A, Jantus-Lewintre E, Sanmartín E, Blasco A, Del Pozo N, Rosell R, Guijarro R, Galbis J, Sánchez JJ, Camps C. Circulating DNA is a useful prognostic factor in patients with advanced non-small cell lung cancer. J Thorac Oncol. 2011;6:286–290. doi: 10.1097/JTO.0b013e31820189a5. [DOI] [PubMed] [Google Scholar]
- 63.Byron E, Pinder-Schenck M. Systemic and targeted therapies for early-stage lung cancer. Cancer Control. 2014;21:21–31. doi: 10.1177/107327481402100104. [DOI] [PubMed] [Google Scholar]
- 64.Drizou M, Kotteas EA, Syrigos N. Treating patients with ALK-rearranged non-small-cell lung cancer: mechanisms of resistance and strategies to overcome it. Clin Transl Oncol. 2017;19:658–666. doi: 10.1007/s12094-016-1605-y. [DOI] [PubMed] [Google Scholar]
- 65.Mamdani H, Ahmed S, Armstrong S, Mok T, Jalal SI. Blood-based tumor biomarkers in lung cancer for detection and treatment. Transl Lung Cancer Res. 2017;6:648–660. doi: 10.21037/tlcr.2017.09.03. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Punnoose EA, Atwal SK, Spoerke JM, Savage H, Pandita A, Yeh RF, Pirzkall A, Fine BM, Amler LC, Chen DS, Lackner MR. Molecular biomarker analyses using circulating tumor cells. PLoS One. 2010;5:e12517. doi: 10.1371/journal.pone.0012517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Tsui DW, Berger MF. Profiling non-small cell lung cancer: from tumor to blood. Clin Cancer Res. 2016;22:790–792. doi: 10.1158/1078-0432.CCR-15-2514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Villaflor V, Won B, Nagy R, Banks K, Lanman RB, Talasaz A, Salgia R. Biopsy-free circulating tumor DNA assay identifies actionable mutations in lung cancer. Oncotarget. 2016;7:66880–66891. doi: 10.18632/oncotarget.11801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Punnoose EA, Atwal S, Liu W, Raja R, Fine BM, Hughes BG, Hicks RJ, Hampton GM, Amler LC, Pirzkall A, Lackner MR. Evaluation of circulating tumor cells and circulating tumor DNA in non-small cell lung cancer: association with clinical endpoints in a phase II clinical trial of pertuzumab and erlotinib. Clin Cancer Res. 2012;18:2391–2401. doi: 10.1158/1078-0432.CCR-11-3148. [DOI] [PubMed] [Google Scholar]
- 70.Santarpia M, Liguori A, D’Aveni A, Karachaliou N, Gonzalez-Cao M, Daffinà MG, Lazzari C, Altavilla G, Rosell R. Liquid biopsy for lung cancer early detection. J Thorac Dis. 2018;10(Suppl 7):S882–S897. doi: 10.21037/jtd.2018.03.81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.McCoach CE, Blakely CM, Banks KC, Levy B, Chue BM, Raymond VM, Le AT, Lee CE, Diaz J, Waqar SN, Purcell WT, Aisner DL, Davies KD, Lanman RB, Shaw AT, Doebele RC. Clinical utility of cell-free DNA for the detection of ALK fusions and genomic mechanisms of ALK inhibitor resistance in non-small cell lung cancer. Clin Cancer Res. 2018;24:2758–2770. doi: 10.1158/1078-0432.CCR-17-2588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Guibert N, Pradines A, Farella M, Casanova A, Gouin S, Keller L, Favre G, Mazieres J. Monitoring KRAS mutations in circulating DNA and tumor cells using digital droplet PCR during treatment of KRAS-mutated lung adenocarcinoma. Lung Cancer. 2016;100:1–4. doi: 10.1016/j.lungcan.2016.07.021. [DOI] [PubMed] [Google Scholar]
- 73.Tong B, Xu Y, Zhao J, Chen M, Zhong W, Xing J, Wang M. Prognostic role of circulating tumor cells in patients with EGFR-mutated or ALK-rearranged non-small cell lung cancer. Thorac Cancer. 2018;9:640–645. doi: 10.1111/1759-7714.12631. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Chang CL, Huang W, Jalal SI, Chan BD, Mahmood A, Shahda S, O’Neil BH, Matei DE, Savran CA. Circulating tumor cell detection using a parallel flow micro-aperture chip system. Lab Chip. 2015;15:1677–1688. doi: 10.1039/c5lc00100e. [DOI] [PubMed] [Google Scholar]
- 75.Xu YH, Zhou J, Pan XF. Detecting circulating tumor cells in patients with advanced non-small cell lung cancer. Genet Mol Res. 2015;14:10352–10358. doi: 10.4238/2015.September.1.1. [DOI] [PubMed] [Google Scholar]
- 76.Horton CE, Kamal M, Leslie M, Zhang R, Tanaka T, Razaq M. Circulating tumor cells accurately predicting progressive disease after treatment in a patient with non-small cell lung cancer showing response on scans. Anticancer Res. 2018;38:1073–1076. doi: 10.21873/anticanres.12325. [DOI] [PubMed] [Google Scholar]
- 77.Yeo T, Tan SJ, Lim CL, Lau DP, Chua YW, Krisna SS, Iyer G, Tan GS, Lim TK, Tan DS, Lim WT, Lim CT. Microfluidic enrichment for the single cell analysis of circulating tumor cells. Sci Rep. 2016;6:22076. doi: 10.1038/srep22076. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Xu R, Zhong G, Huang T, He W, Kong C, Zhang X, Wang Y, Liu M, Xu M, Chen S. Sequencing of circulating tumor DNA for dynamic monitoring of gene mutations in advanced non-small cell lung cancer. Oncol Lett. 2018;15:3726–3734. doi: 10.3892/ol.2018.7808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Moding EJ, Diehn M, Wakelee HA. Circulating tumor DNA testing in advanced non-small cell lung cancer. Lung Cancer. 2018;119:42–47. doi: 10.1016/j.lungcan.2018.02.019. [DOI] [PubMed] [Google Scholar]
- 80.Thompson JC, Yee SS, Troxel AB, Savitch SL, Fan R, Balli D, Lieberman DB, Morrissette JD, Evans TL, Bauml J, Aggarwal C, Kosteva JA, Alley E, Ciunci C, Cohen RB, Bagley S, Stonehouse-Lee S, Sherry VE, Gilbert E, Langer C, Vachani A, Carpenter EL. 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;22:5772–5782. doi: 10.1158/1078-0432.CCR-16-1231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Yang Y, Shen X, Li R, Shen J, Zhang H, Yu L, Liu B, Wang L. The detection and significance of EGFR and BRAF in cell-free DNA of peripheral blood in NSCLC. Oncotarget. 2017;8:49773–49782. doi: 10.18632/oncotarget.17937. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Hou H, Yang X, Zhang J, Zhang Z, Xu X, Zhang X, Zhang C, Liu D, Yan W, Zhou N, Zhu H, Qian Z, Li Z, Zhang X. Discovery of targetable genetic alterations in advanced non-small cell lung cancer using a next-generation sequencing-based circulating tumor DNA assay. Sci Rep. 2017;7:14605. doi: 10.1038/s41598-017-14962-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Taus Á, Camacho L, Rocha P, Hardy-Werbin M, Pijuan L, Piquer G, López E, Dalmases A, Longarón R, Clavé S, Salido M, Albanell J, Bellosillo B, Arriola E. Dynamics of EGFR mutation load in plasma for prediction of treatment response and disease progression in patients with EGFR-mutant lung adenocarcinoma. Clin Lung Cancer. 2018;19:387–394. e2. doi: 10.1016/j.cllc.2018.03.015. [DOI] [PubMed] [Google Scholar]
- 84.Wang Y, Tian PW, Wang WY, Wang K, Zhang Z, Chen BJ, He YQ, Li L, Liu H, Chuai S, Li WM. Noninvasive genotyping and monitoring of anaplastic lymphoma kinase (ALK) rearranged non-small cell lung cancer by capture-based next-generation sequencing. Oncotarget. 2016;7:65208–65217. doi: 10.18632/oncotarget.11569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Riediger AL, Dietz S, Schirmer U, Meister M, Heinzmann-Groth I, Schneider M, Muley T, Thomas M, Sültmann H. Mutation analysis of circulating plasma DNA to determine response to EGFR tyrosine kinase inhibitor therapy of lung adenocarcinoma patients. Sci Rep. 2016;6:33505. doi: 10.1038/srep33505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Diehl F, Schmidt K, Choti MA, Romans K, Goodman S, Li M, Thornton K, Agrawal N, Sokoll L, Szabo SA, Kinzler KW, Vogelstein B, Diaz LA Jr. Circulating mutant DNA to assess tumor dynamics. Nat Med. 2008;14:985–990. doi: 10.1038/nm.1789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Deig CR, Thowe RT, Frye ED, Chin-Sinex HJ, Mendonca MS, Lautenschlaeger T. In vitro cell-free DNA quantification: a novel method to accurately quantify cell survival after irradiation. Radiat Res. 2018;190:22–27. doi: 10.1667/RR15016.1. [DOI] [PubMed] [Google Scholar]
