Background: Circulating tumor DNA (ctDNA) has offered a minimally invasive and feasible approach for detection of EGFR mutation for non–small cell lung cancer (NSCLC). This meta-analysis was designed to investigate the diagnostic value of ctDNA, compared with current “gold standard,” tumor tissues.

Methods: We searched PubMed, EMBASE, Cochrane Library, and Web of Science to identify eligible studies that reported the sensitivity and specificity of ctDNA for detection of EGFR mutation status in NSCLC. Eligible studies were pooled to calculate the pooled sensitivity, specificity, and diagnostic odds ratio (DOR). The summary ROC curve (SROC) and area under SROC (AUSROC) were used to evaluate the overall diagnostic performance.

Results: Twenty-seven eligible studies involving 3,110 participants were included and analyzed in our meta-analysis, and most studies were conducted among Asian population. The pooled sensitivity, specificity, and DOR were 0.620 [95% confidence intervals (CI), 0.513–0.716), 0.959 (95% CI, 0.929–0.977), and 38.270 (95% CI, 21.090–69.444), respectively. The AUSROC was 0.91 (95% CI, 0.89–0.94), indicating the high diagnostic performance of ctDNA.

Conclusion: ctDNA is a highly specific and effective biomarker for the detection of EGFR mutation status.

Impact: ctDNA analysis will be a key part of personalized cancer therapy of NSCLC. Cancer Epidemiol Biomarkers Prev; 24(1); 206–12. ©2014 AACR.

One of the most exciting breakthroughs in cancer treatment is the application of personalized chemotherapy tailored according to the individual's genetic background. For non–small cell lung cancer (NSCLC), EGFR-TKIs, such as gefitinib and erlotinib, have been used for years (1, 2). It has been documented that EGFR mutation status is a sensitive and reliable biomarker for EGFR-TKIs therapy (3, 4). Patients carrying the point mutation in exon 21 (L858R) or deletion in exon 19 show good response to EGFR-TKIs (4); on the other hand, the point mutation (T790M) in exon20 indicates resistance to EGFR-TKIs and poor prognosis (5). It has also been reported that EGFR mutation status might change after chemotherapy. Bai and colleagues observed better response in patients whose EGFR mutation status switched from positive to negative after chemotherapy (6). Therefore, the examination of EGFR mutations is essential to determine an appropriate treatment strategy, especially for the administration of EGFR-TKIs and it is also necessary to monitor the dynamic change of EGFR mutation to identify acquired resistance at early time.

Currently, tumor tissue is the gold standard for detection of EGFR mutation, which is usually obtained by biopsy or surgery (7). Biopsy and surgery are invasive procedures, which cannot be performed repeatedly and cannot reflect the heterogeneity of tumor. Furthermore, biopsy is not without complications (7, 8). What is more important is that biopsy is only a snapshot, which is subjected to selection bias resulting from tumor heterogeneity (9).

In patients with cancer, dead tumor cells shed DNA into bloodstream and these DNA fragments carry tumor-specific sequence alterations (circulating tumor DNA, ctDNA; refs. 10, 11). Compared with tumor tissues, ctDNA is a potential source of tumor DNA for the identification of tumor-associated genetic and molecular alterations (12). Compared with biopsy, ctDNA is much more feasible, suitable for a general screening test for patients with cancer to characterize the genetic profile, which will greatly promote personalized cancer therapy. In addition, due to its nature of minimal invasiveness, ctDNA is suitable for real-time tumor monitoring (10, 13). Many studies have shown the feasibility and predictive value of using ctDNA to monitor tumor dynamics in various solid tumors (14–17), in which ctDNA even showed better test performance than circulating tumor cells and conventional serum biomarkers (18). As for NSCLC, many clinical centers have investigated the diagnostic accuracy of ctDNA for detection of EGFR mutation (19–21). The concordance rate of EGFR mutation between ctDNA and tumor tissues is largely dependent on detection techniques, and varies from 66% to 100% (22). In addition, these studies also differ in many aspects except for detection techniques, such as storage of tumor tissue, collection time of blood sample, and tumor–node–metastasis (TNM) stage; while no conclusion could be drawn for these factors.

Therefore, we conducted this meta-analysis to investigate the diagnostic accuracy of ctDNA for EGFR mutation detection compared with the “gold standard”-tumor tissues and address the effect of individual covariates.

Searching strategy

The present meta-analysis was performed and reported according to the guideline about diagnostic studies. MEDLINE (via PubMed), EMBASE (via OvidSP), the Cochrane library, and ISI Web of Knowledge were searched for potentially relevant studies. The searching strategy included the combination of following key words and medical subheadings: “lung neoplasms” or “lung cancer,” “EGFR” or “erbB1,” “serum” or “plasma” or “circulating,” and “mutations.” No limitation was performed. We searched the database between inception and September 28, 2014. Reference lists of included studies and relevant reviews were also manually screened.

Inclusion and exclusion criteria

Records retrieved from databases and reference lists were first screened by titles and abstracts and then full-text articles were further reviewed for eligibility. Eligible studies were selected according to the following inclusion criteria: (i) patients with NSCLC should be diagnosed histopathologically or cytologically; (ii) EGFR mutation status should be detected by circulating free DNA and tumor tissues; and (iii) providing sufficient information to construct the diagnostic 2 × 2 table, that is, false and true positives and negatives were provided.

The exclusion criteria were as follows: (i) tumor tissues and blood samples were not paired; (ii) EGFR mutation status was not compared with tumor tissues; and (iii) duplicate reports from the same center (refs. 23–25; the one with most patients with NSCLC were included; ref. 23). All records were reviewed by the authors independently and they reached consensus on each eligible study.

Data extraction

Name of first author, year of publication, country where the study was performed, histologic type of NSCLC, TNM stage, techniques used for EGFR mutation detection in ctDNA, collection time of blood sample (before or after chemotherapy), serum or plasma, format of tumor tissues, true positive (TP), false positive (FP), false negative (FN), and true negative (TN) were collected from eligible studies. When EGFR mutation was detected by multiple methods, the one with best sensitivity or specificity was extracted. Two authors (M. Qiu and X. Ding) extracted these data independently and discrepancy between two authors was resolved by discussion with the third author (R. Yin).

Quality assessment

QUADAS-2 (quality assessment of diagnostic accuracy studies 2; QUADAS-2) is a tool (26) designed to evaluate the quality of primary diagnostic accuracy studies, which consists of four key domains (patient selection, index test, reference standard, and flow and timing). Methodologic quality of eligible studies was evaluated by QUADAS-2 by two investigators.

Statistical analysis

EGFR mutation status detected in tumor tissues was treated as the “gold standard.” We tabulated true positives, false positives, false negatives, and true negatives stratified by study. These diagnostic numbers were used to calculate the pooled sensitivity, specificity, positive likelihood ratio (PLR), negative likelihood ratio (NLR), diagnostic odds ratio (DOR), and corresponding 95% confidence intervals (95% CI). The PLR is calculated as: sensitivity/(1−specificity) and the NLR is calculated as (1 − sensitivity)/specificity. A clinically useful test was defined with a PLR>5.0 and a NLR<0.2 (27, 28). DOR is a measure that combined sensitivity and specificity, which is calculated as: PLR/NLR19. The summary ROC curve (SROC) was generated and the area under the SROC (AUSROC) was calculated.

The heterogeneity caused by non-threshold effect was measured by the Q test and the inconsistency index (I2), and a P value≤0.05 and a I2 value ≥50% indicated significant heterogeneity caused by non-threshold effect. Subgroup analyses were performed for detection techniques, collection time of blood sample, format of tissues, and TNM stages. Publication bias was detected by the Deek's funnel plot 21 and a P vale < 0.05 indicated the presence of publication bias (29).

All statistical analyses were performed using the STATA software (version 11.2, STATA Corp.) with the MIDAS module (30).

Our database search retrieved 976 records. After reviewing the title and abstracts, 927 records were excluded. By reviewing full-text articles, we excluded further 23 records, leaving 26 eligible articles (refs. 19–23, 31–51; Fig. 1). In the study reported by Li and colleagues (49), EGFR mutation was detected in both plasma and serum, and the data of plasma and serum were analyzed as two independently studies. Thus, 27 eligible studies were included in meta-analysis. A manual search of reference lists of eligible studies and related reviews did not identify more relevant articles.

Figure 1.

Flow diagram of study selection. *, In the study reported by Li et al. (49), EGFR mutation was detected both in plasma and serum, and the data of plasma and serum were analyzed as two independent studies. Thus, 27 eligible studies were included in the meta-analysis.

Figure 1.

Flow diagram of study selection. *, In the study reported by Li et al. (49), EGFR mutation was detected both in plasma and serum, and the data of plasma and serum were analyzed as two independent studies. Thus, 27 eligible studies were included in the meta-analysis.

Close modal

Characteristics of 27 eligible studies are shown in Table 1. A total of 3,110 patients with NSCLC were included in analysis. Most studies were conducted in Asia and only recruited patients with advanced disease. Formalin-fixed paraffin-embedded (FFPE) tumor tissues were used for detecting EGFR mutation status in 14 studies. Only six studies reported the exact collection time point of both samples (tumor tissues and blood samples). Various detection methods were reported and the ARMS was the most frequently used method.

Table 1.

Characteristics of eligible studies

AuthorYearCountryTNMTreatmentCollection timeSampleDetection methodsTPFPFNTN
Sriram KB 2011 Australia NA Frozen NA Serum ME-PCR 58 
He C 2009 China NA FFPE NA Plasma ME-PCR 
Yung TK 2009 China NA FFPE NA Plasma Digital PCR 11 17 
Jiang B 2011 China Advanced NA NA Serum Mutant-enriched sequencing 14 40 
Hu C 2012 China I-IV Frozen Yes Serum HRM 22 
Huang Z 2012 China NA FFPE NA Plasma DHPLC 188 81 108 445 
Xu F 2012 China Advanced FFPE NA Serum ARMS 26 
Yam I 2012 China I-IV NA NA Plasma AS-APEX 30 
Jing CW 2013 China I-IV FFPE NA Plasma HRM 29 16 73 
Liu X 2013 China Advanced FFPE NA Plasma ARMS 27 13 46 
Lv C 2013 China Advanced FFPE Yes Plasma DHPLC 
Zhang H 2013 China Advanced FFPE Yes Plasma MEL 15 64 
Zhao X 2013 China I-IV FFPE Yes Plasma ME-PCR 16 29 63 
Wang S 2014 China Advanced FFPE NA Plasma ARMS 15 53 64 
Kimura H 2006 Japan Advanced FFPE NA Serum ARMS 
Kimura H 2007 Japan Advanced FFPE NA Serum ARMS 33 
Taniguchi K 2011 Japan Advanced NA NA Plasma BEAMing 32 12 
Goto K 2012 Japan NA NA NA Serum AS-APEX 22 29 35 
Nakamura T 2012 Japan I-IV NA No Plasma Inhibiting PCR-quenching probe method 21 26 23 
Kim HR 2013 Korea Advanced NA No Plasma PNAClamp 29 
Kim ST 2013 Korea Advanced FFPE NA Serum PNA–LNA PCR clamp 42 
Kuang Y 2009 USA NA NA NA Plasma ARMS 21 11 
Brevet M 2011 USA Advanced NA NA Plasma Sequenom 10 11 
Li X (plasma) 2014 China I-IV Frozen NA Plasma ARMS 27 29 62 
Li X (serum) 2014 China I-IV Frozen NA Serum ARMS 19 29 42 
Douillard JY 2014 Europe NA NA YES Plasma ARMS 69 36 546 
Weber B 2014 Denmark I-IV FFPE YES Plasma Cobas EGFR blood test 17 11 162 
AuthorYearCountryTNMTreatmentCollection timeSampleDetection methodsTPFPFNTN
Sriram KB 2011 Australia NA Frozen NA Serum ME-PCR 58 
He C 2009 China NA FFPE NA Plasma ME-PCR 
Yung TK 2009 China NA FFPE NA Plasma Digital PCR 11 17 
Jiang B 2011 China Advanced NA NA Serum Mutant-enriched sequencing 14 40 
Hu C 2012 China I-IV Frozen Yes Serum HRM 22 
Huang Z 2012 China NA FFPE NA Plasma DHPLC 188 81 108 445 
Xu F 2012 China Advanced FFPE NA Serum ARMS 26 
Yam I 2012 China I-IV NA NA Plasma AS-APEX 30 
Jing CW 2013 China I-IV FFPE NA Plasma HRM 29 16 73 
Liu X 2013 China Advanced FFPE NA Plasma ARMS 27 13 46 
Lv C 2013 China Advanced FFPE Yes Plasma DHPLC 
Zhang H 2013 China Advanced FFPE Yes Plasma MEL 15 64 
Zhao X 2013 China I-IV FFPE Yes Plasma ME-PCR 16 29 63 
Wang S 2014 China Advanced FFPE NA Plasma ARMS 15 53 64 
Kimura H 2006 Japan Advanced FFPE NA Serum ARMS 
Kimura H 2007 Japan Advanced FFPE NA Serum ARMS 33 
Taniguchi K 2011 Japan Advanced NA NA Plasma BEAMing 32 12 
Goto K 2012 Japan NA NA NA Serum AS-APEX 22 29 35 
Nakamura T 2012 Japan I-IV NA No Plasma Inhibiting PCR-quenching probe method 21 26 23 
Kim HR 2013 Korea Advanced NA No Plasma PNAClamp 29 
Kim ST 2013 Korea Advanced FFPE NA Serum PNA–LNA PCR clamp 42 
Kuang Y 2009 USA NA NA NA Plasma ARMS 21 11 
Brevet M 2011 USA Advanced NA NA Plasma Sequenom 10 11 
Li X (plasma) 2014 China I-IV Frozen NA Plasma ARMS 27 29 62 
Li X (serum) 2014 China I-IV Frozen NA Serum ARMS 19 29 42 
Douillard JY 2014 Europe NA NA YES Plasma ARMS 69 36 546 
Weber B 2014 Denmark I-IV FFPE YES Plasma Cobas EGFR blood test 17 11 162 

Abbreviations: HRM, high-resolution melting; PNA-LNA, peptide nucleic acid-locked nucleic acid; AS-APEX, allele-specific arrayed primer extension; ME-PCR, mutant-enriched-PCR; DHPLC, denaturing high-performance liquid chromatography; BEAMing, beads, emulsion, amplification, and magnetics; ARMS, amplification refractory mutation system; MEL, mutant-enriched liquid chip.

Methodologic quality of eligible studies was assessed by QUADAS-2. The overall quality of included studies was moderate (Supplementary Fig. S1). The Deek regression test was performed to detect potential publication bias and no significant publication bias was detected (P = 0.896, Supplementary Fig. S2).

The pooled specificity was 0.959 (95% CI, 0.929–0.977) and the pooled sensitivity was 0.620 (95% CI, 0.513–0.716). The AUSROC and the pooled DOR was 0.91 (95% CI, 0.89–0.94, Fig. 2) and 38.270 (95% CI, 21.090–69.444), respectively. Between-studies heterogeneity was detected among eligible studies (bivariate model 98.54, 95% CI, 97.88–99.21), while we did not find any evidence of threshold effect.

Figure 2.

SROC curve. The figure also shows 95% confidence contour and 95% prediction contour.

Figure 2.

SROC curve. The figure also shows 95% confidence contour and 95% prediction contour.

Close modal

To investigate the effect of potential confounding factors, we conducted stratified analysis according to detection methods, TNM stages, collection time and format of blood sample, and treatment of tumor tissues. As measured by AUC, ME-PCR (0.97, 95% CI, 0.95–0.98) had higher diagnostic accuracy than other methods (significance not tested). For TNM stage, the diagnostic accuracy of ctDNA was higher in patients with advanced stage of NSCLC (0.96, 95% CI, 0.94–0.97; significance not tested). The diagnostic accuracy of ctDNA would be higher if the ctDNA was extracted from plasma and before chemotherapy (significance not tested). Unexpectedly, we found that the diagnostic performance of ctDNA would be better when compared with FFPE tissues than frozen tissues.

Detection of EGFR mutation status in NSCLC has become a routine clinical test providing important information for patient prognosis and selection of EGFR-TKI therapy. In this meta-analysis, we found that compared with tumor tissues, detection of EGFR mutation status by ctDNA had high diagnostic accuracy. Detection EGFR mutation status by ctDNA will be widely applied in clinical practice and improve personalized cancer therapy and make real-time monitoring possible during chemotherapy (7, 11, 17).

The area under ROC serves as a global measure of diagnostic performance. According to the suggested guideline for interpretation of area under ROC (52), ctDNA had high diagnostic accuracy (0.9<AUC<1) for detection of EGFR mutation status in NSCLC. The value of DOR ranges from 0 to infinity, with higher values indicating better discriminatory test performance (53). Meta-analysis results showed that ctDNA had high diagnostic performance with a DOR of 38.270.

Likelihood ratios and posttest probabilities are parameters used for evaluating clinical or patient-relevant utility of the diagnostic test (27). Likelihood ratios and posttest probabilities are also important for a biomarker. They provided information about the likelihood that a patient with positive or negative result has EGFR mutation or not. In our study, the PLR (PLR>10) and negative posttest probability (<0.1) were high (Fig. 3). Given the PLR and NLR, ctDNA is located in the right upper quadrant (Fig. 4), indicating that ctDNA could serve as a test to confirm EGFR mutation.

Figure 3.

Fagan nomogram of ctDNA for the detection of EGFR mutation.

Figure 3.

Fagan nomogram of ctDNA for the detection of EGFR mutation.

Close modal
Figure 4.

The likelihood ratio matrix of ctDNA for the detection of EGFR mutation.

Figure 4.

The likelihood ratio matrix of ctDNA for the detection of EGFR mutation.

Close modal

In overall analysis, ctDNA showed high diagnostic accuracy; however, no conclusion could be drawn for other important covariates, like detection methods, source of ctDNA, collection time of blood sample, TNM stage, and treatment of tumor tissues. Therefore, stratified analyses were performed to investigate whether these factors could influence diagnostic accuracy of ctDNA. ARMS was the most frequently used method and several commercial detection kits based on ARMS have been developed. Therefore, the diagnostic performance of ARMS was most useful for clinical practice. Our meta-analysis results showed that the specificity of ARMS was highest among all the methods assessed and the overall diagnostic performance was high (AUSROC and DOR, Table 2). In addition, the ME-PCR showed highest AUSROC, while the sample size of ME-PCR was relatively small and further studies are warranted. Although several highly sensitive methods such as digital PCR were also reported, subgroup analysis was not allowed because of too few studies.

Table 2.

Meta-analysis results

StudiesAUSROCSensitivitySpecificityPLRNLRDOR
Overall 27 0.91 (0.89–0.94) 0.620 (0.513–0.716) 0.959 (0.929–0.977) 15.176 (8.924–25.807) 0.397 (0.305–0.515) 38.270 (21.090–69.444) 
Format of blood sample 
 Plasma 18 0.92 (0.89–0.94) 0.599 (0.468–0.717) 0.960 (0.925–0.979) 14.952 (7.876–28.386) 0.418 (0.305–0.572) 35.798 (16.375–78.259) 
 Serum 0.90 (0.87–0.92) 0.658 (0.463–0.811) 0.954 (0.864–0.986) 14.428 (5.440–38.268) 0.359 (0.219–0.587) 40.223 (16.538–97.826) 
TNM stage 
 I-IV 0.94 (0.91–0.95) 0.786 (0.420–0.949) 0.921 (0.751–0.978) 9.935 (3.771–26.175) 0.233 (0.071–0.761) 42.703 (17.732–102.837) 
 Advanced 14 0.96 (0.94–0.97) 0.521 (0.399–0.641) 0.962 (0.940–0.977) 13.865 (7.861–24.454) 0.497 (0.382–0.647) 27.877 (13.047–59.565) 
Storage method of tumor tissues 
 FFPE 14 0.93 (0.90–0.95) 0.607 (0.484–0.718) 0.957 (0.925–0.975) 14.011 (7.942–24.720) 0.411 (0.304–0.555) 34.104 (16.564–70.217) 
 Frozen 0.84 (0.81–0.87) 0.627 (0.253–0.893) 0.908 (0.479–0.991) 6.785 (1.292–35.636) 0.411 (0.181–0.935) 16.507 (4.647–58.635) 
Detection methods 
 ARMS 0.88 (0.85–0.91) 0.549 (0.419–0.672) 0.975 (0.937–0.991) 22.283 (8.244–60.230) 0.463 (0.347–0.617) 48.168 (15.479–149.887) 
 AS-APEX 0.96 (0.94–0.98) 0.859 (0.189–0.994) 0.935 (0.527–0.995) 13.313 (1.635–108.404) 0.151 (0.010–2.212) 88.339 (8.851–881.676) 
 DHPLC 0.82 (0.78–0.85) 0.628 (0.572–0.681) 0.846 (0.813–0.874) 4.086 (3.286–5.081) 0.439 (0.377–0.511) 9.303 (6.672–12.973) 
 HRM 0.91 (0.88–0.93) 0.887 (0.402–0.989) 0.736 (0.042–0.994) 3.360 (0.197–57.314) 0.153 (0.035–0.673) 21.974 (1.522–317.223) 
 ME-PCR 0.97 (0.95–0.98) 0.556 (0.290–0.794) 0.975 (0.906–0.994) 22.469 (4.628–109.078) 0.455 (0.241–0.858) 49.369 (6.522–373.727) 
Collection time of blood sample 
 BC 0.89 (0.86–0.91) 0.647 (0.375–0.848) 0.967 (0.773–0.996) 19.572 (2.931–130.691) 0.365 (0.184–0.725) 53.568 (8.694–330.045) 
 AC 0.81 (0.78–0.85) 0.307 (0.149–0.528) 0.961 (0.732–0.995) 7.784 (0.711–85.209) 0.721 (0.522–0.997) 10.790 (0.767–151.869) 
StudiesAUSROCSensitivitySpecificityPLRNLRDOR
Overall 27 0.91 (0.89–0.94) 0.620 (0.513–0.716) 0.959 (0.929–0.977) 15.176 (8.924–25.807) 0.397 (0.305–0.515) 38.270 (21.090–69.444) 
Format of blood sample 
 Plasma 18 0.92 (0.89–0.94) 0.599 (0.468–0.717) 0.960 (0.925–0.979) 14.952 (7.876–28.386) 0.418 (0.305–0.572) 35.798 (16.375–78.259) 
 Serum 0.90 (0.87–0.92) 0.658 (0.463–0.811) 0.954 (0.864–0.986) 14.428 (5.440–38.268) 0.359 (0.219–0.587) 40.223 (16.538–97.826) 
TNM stage 
 I-IV 0.94 (0.91–0.95) 0.786 (0.420–0.949) 0.921 (0.751–0.978) 9.935 (3.771–26.175) 0.233 (0.071–0.761) 42.703 (17.732–102.837) 
 Advanced 14 0.96 (0.94–0.97) 0.521 (0.399–0.641) 0.962 (0.940–0.977) 13.865 (7.861–24.454) 0.497 (0.382–0.647) 27.877 (13.047–59.565) 
Storage method of tumor tissues 
 FFPE 14 0.93 (0.90–0.95) 0.607 (0.484–0.718) 0.957 (0.925–0.975) 14.011 (7.942–24.720) 0.411 (0.304–0.555) 34.104 (16.564–70.217) 
 Frozen 0.84 (0.81–0.87) 0.627 (0.253–0.893) 0.908 (0.479–0.991) 6.785 (1.292–35.636) 0.411 (0.181–0.935) 16.507 (4.647–58.635) 
Detection methods 
 ARMS 0.88 (0.85–0.91) 0.549 (0.419–0.672) 0.975 (0.937–0.991) 22.283 (8.244–60.230) 0.463 (0.347–0.617) 48.168 (15.479–149.887) 
 AS-APEX 0.96 (0.94–0.98) 0.859 (0.189–0.994) 0.935 (0.527–0.995) 13.313 (1.635–108.404) 0.151 (0.010–2.212) 88.339 (8.851–881.676) 
 DHPLC 0.82 (0.78–0.85) 0.628 (0.572–0.681) 0.846 (0.813–0.874) 4.086 (3.286–5.081) 0.439 (0.377–0.511) 9.303 (6.672–12.973) 
 HRM 0.91 (0.88–0.93) 0.887 (0.402–0.989) 0.736 (0.042–0.994) 3.360 (0.197–57.314) 0.153 (0.035–0.673) 21.974 (1.522–317.223) 
 ME-PCR 0.97 (0.95–0.98) 0.556 (0.290–0.794) 0.975 (0.906–0.994) 22.469 (4.628–109.078) 0.455 (0.241–0.858) 49.369 (6.522–373.727) 
Collection time of blood sample 
 BC 0.89 (0.86–0.91) 0.647 (0.375–0.848) 0.967 (0.773–0.996) 19.572 (2.931–130.691) 0.365 (0.184–0.725) 53.568 (8.694–330.045) 
 AC 0.81 (0.78–0.85) 0.307 (0.149–0.528) 0.961 (0.732–0.995) 7.784 (0.711–85.209) 0.721 (0.522–0.997) 10.790 (0.767–151.869) 

Abbreviations: BC, before chemotherapy; AC, after chemotherapy.

Usually, ctDNAs were extracted from serum or plasma. Stratified analysis showed that ctDNA extracted from plasma had higher diagnostic accuracy than ctDNA extracted from serum. As measured by AUSROC, ctDNA had higher diagnostic accuracy when blood sample was collected before chemotherapy. Many investigators have reported that EGFR mutation status would change after chemotherapy (6) and this would lead to the inconsistence between tissues and ctDNA when blood sample was collected after chemotherapy. Compared with patients at early stage, those at advanced stage have high level of circulating-free DNA. It has been suggested that if the fraction of ctDNA in a sample is lower than 0.01%, it is considered negative for ctDNA (54, 55). These results suggested that the detection performance of ctDNA would be higher when ctDNA was in large amount. Currently, the exact mechanism that determines the release of ctDNA is unclear, but current hypotheses indicate that the amount of ctDNA is associated with tumor volume and status of metastasis. TNM stage is suitable marker that combines tumor volume and metastasis. By subgroup analysis, we found that in patients with advanced stage of NSCLC, ctDNA had higher AUSROC.

FFPE tissue is the most common method used for tissue storage, but this will lead to cross-link between nucleic acids and proteins, which then lead to false-positive or false-negative results. On the other hand, liquid nitrogen frozen tissues do not have the problem. However, stratified analysis found that concordance rate was higher in FFPE; this might be explained by the fact that too few studies were available for frozen tissues.

In our meta-analysis, we showed that ctDNA had high diagnostic accuracy, especially the high degree of specificity. As Diaz and Bardelli pointed, the key advantage of ctDNA is the high degree of specificity (7), since the mutations are defined by their presence in the tumor DNA and absence in matched normal DNA. As the likelihood ration scattergram showed, ctDNA is suitable for a screening test to identify those with sensitive EGFR mutation. Because of its noninvasive nature, ctDNA is a perfect marker for real-time monitoring during management of NSCLC and acquired resistance timely (11, 18).

We would like to acknowledge and discuss the potential limitations of present study to prevent misinterpretation of our findings. First, several point mutations and deletion of EGFR were reported, while we did not perform stratified analysis for individual mutations specifically. Second, substantial heterogeneity was detected but none of the analyzed characteristics could account for the majority of heterogeneity. Except for the factors analyzed, the included studies differ in many aspects, like ethnicity, percentage of lung adenocarcinoma, and methodologic quality. These unrecorded differences might be the potential sources of heterogeneity. Third, for several stratified analyses, the number of included studies was relatively small and the results were easily biased. And results of these stratified analyses should be interpreted with caution. In addition, some important information was not available in all studies, such as collection time of blood sample, the relationship between treatment and blood sample collection, and the detailed chemotherapy regimens. Further studies are warranted to investigate these issues.

In conclusion, ctDNA is an effective method to detect EGFR mutation status in NSCLC. Given the high diagnostic accuracy and specificity, ctDNA could be a primary screening test for NSCLC and development of standardized methodologies for ctDNA analyses and validation is necessary.

No potential conflicts of interest were disclosed.

Conception and design: M. Qiu, L. Xu, R. Yin

Development of methodology: M. Qiu, L. Xu

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): M. Qiu, J. Wang, Y. Xu, X. Ding, M. Li, F. Jiang, L. Xu

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): M. Qiu, J. Wang, Y. Xu, X. Ding, L. Xu

Writing, review, and/or revision of the manuscript: M. Qiu, J. Wang, Y. Xu, X. Ding, L. Xu, R. Yin

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): M. Qiu, M. Li, F. Jiang, L. Xu

Study supervision: M. Qiu, L. Xu, R. Yin

This work was funded by the Natural Science Foundation of China (81372321 to L. Xu; 81201830 and 81472200 to R. Yin), Natural Science Foundation for High Education of Jiangsu Province (13KJB320010 to R. Yin), Research and Innovation Program for Graduates of Jiangsu Province (CXLX13_571 to M. Qiu), Human Resource Summit Grant of Jiangsu Province (10-D-078 to R. Yin and WS-116 to M. Li), and Jiangsu Provincial Special Program of Medical Science (BL2012030 to L. Xu).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Mok
TS
,
Wu
YL
,
Thongprasert
S
,
Yang
CH
,
Chu
DT
,
Saijo
N
, et al
Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma
.
N Engl J Med
2009
;
361
:
947
57
.
2.
Zhou
C
,
Wu
YL
,
Chen
G
,
Feng
J
,
Liu
XQ
,
Wang
C
, et al
Erlotinib versus chemotherapy as first-line treatment for patients with advanced EGFR mutation-positive non-small-cell lung cancer (OPTIMAL, CTONG-0802): a multicentre, open-label, randomised, phase 3 study
.
Lancet Oncol
2011
;
12
:
735
42
.
3.
Hirsch
FR
,
Varella-Garcia
M
,
Bunn
PA
 Jr
,
Franklin
WA
,
Dziadziuszko
R
,
Thatcher
N
, et al
Molecular predictors of outcome with gefitinib in a phase III placebo-controlled study in advanced non-small-cell lung cancer
.
J Clin Oncol
2006
;
24
:
5034
42
.
4.
Sharma
SV
,
Bell
DW
,
Settleman
J
,
Haber
DA
. 
Epidermal growth factor receptor mutations in lung cancer
.
Nat Rev Cancer
2007
;
7
:
169
81
.
5.
Engelman
JA
,
Mukohara
T
,
Zejnullahu
K
,
Lifshits
E
,
Borras
AM
,
Gale
CM
, et al
Allelic dilution obscures detection of a biologically significant resistance mutation in EGFR-amplified lung cancer
.
J Clin Invest
2006
;
116
:
2695
706
.
6.
Bai
H
,
Wang
Z
,
Chen
K
,
Zhao
J
,
Lee
JJ
,
Wang
S
, et al
Influence of chemotherapy on EGFR mutation status among patients with non-small-cell lung cancer
.
J Clin Oncol
2012
;
30
:
3077
83
.
7.
Diaz
LA
 Jr
,
Bardelli
A
. 
Liquid biopsies: genotyping circulating tumor DNA
.
J Clin Oncol
2014
;
32
:
579
86
.
8.
Overman
MJ
,
Modak
J
,
Kopetz
S
,
Murthy
R
,
Yao
JC
,
Hicks
ME
, et al
Use of research biopsies in clinical trials: are risks and benefits adequately discussed?
J Clin Oncol
2013
;
31
:
17
22
.
9.
Gerlinger
M
,
Rowan
AJ
,
Horswell
S
,
Larkin
J
,
Endesfelder
D
,
Gronroos
E
, et al
Intratumor heterogeneity and branched evolution revealed by multiregion sequencing
.
N Engl J Med
2012
;
366
:
883
92
.
10.
Spellman
PT
,
Gray
JW
. 
Detecting cancer by monitoring circulating tumor DNA
.
Nat Med
2014
;
20
:
474
5
.
11.
De Mattos-Arruda
L
,
Cortes
J
,
Santarpia
L
,
Vivancos
A
,
Tabernero
J
,
Reis-Filho
JS
, et al
Circulating tumour cells and cell-free DNA as tools for managing breast cancer
.
Nat Rev Clin Oncol
2013
;
10
:
377
89
.
12.
Schwarzenbach
H
,
Hoon
DS
,
Pantel
K
. 
Cell-free nucleic acids as biomarkers in cancer patients
.
Nat Rev Cancer
2011
;
11
:
426
37
.
13.
ctDNA is a specific and sensitive biomarker in multiple human cancers
.
Cancer Discov
2014
;
4
:
OF8
.
14.
Bettegowda
C
,
Sausen
M
,
Leary
RJ
,
Kinde
I
,
Wang
Y
,
Agrawal
N
, et al
Detection of circulating tumor DNA in early- and late-stage human malignancies
.
Sci Transl Med
2014
;
6
:
224ra24
.
15.
Zhou
J
,
Shi
YH
,
Fan
J
. 
Circulating cell-free nucleic acids: promising biomarkers of hepatocellular carcinoma
.
Semin Oncol
2012
;
39
:
440
8
.
16.
Newman
AM
,
Bratman
SV
,
To
J
,
Wynne
JF
,
Eclov
NC
,
Modlin
LA
, et al
An ultrasensitive method for quantitating circulating tumor DNA with broad patient coverage
.
Nat Med
2014
;
20
:
548
54
.
17.
Esposito
A
,
Bardelli
A
,
Criscitiello
C
,
Colombo
N
,
Gelao
L
,
Fumagalli
L
, et al
Monitoring tumor-derived cell-free DNA in patients with solid tumors: clinical perspectives and research opportunities
.
Cancer Treat Rev
2014
;
40
:
648
55
.
18.
Dawson
SJ
,
Tsui
DW
,
Murtaza
M
,
Biggs
H
,
Rueda
OM
,
Chin
SF
, et al
Analysis of circulating tumor DNA to monitor metastatic breast cancer
.
N Engl J Med
2013
;
368
:
1199
209
.
19.
Wang
S
,
Han
X
,
Hu
X
,
Wang
X
,
Zhao
L
,
Tang
L
, et al
Clinical significance of pretreatment plasma biomarkers in advanced non-small
.
Clin Chim Acta
2014
;
430C
:
63
70
.
20.
Jing
CW
,
Wang
Z
,
Cao
HX
,
Ma
R
,
Wu
JZ
. 
High resolution melting analysis for epidermal growth factor receptor mutations
.
Asian Pac J Cancer Prev
2013
;
14
:
6619
23
.
21.
Zhang
H
,
Liu
D
,
Li
S
,
Zheng
Y
,
Yang
X
,
Li
X
, et al
Comparison of EGFR signaling pathway somatic DNA mutations derived from
.
J Mol Diagn
2013
;
15
:
819
26
.
22.
Brevet
M
,
Johnson
ML
,
Azzoli
CG
,
Ladanyi
M
. 
Detection of EGFR mutations in plasma DNA from lung cancer patients by mass
.
Lung Cancer
2011
;
73
:
96
102
.
23.
Huang
Z
,
Wang
Z
,
Bai
H
,
Wu
M
,
An
T
,
Zhao
J
, et al
The detection of EGFR mutation status in plasma is reproducible and can dynamically predict the efficacy of EGFR‐TKI
.
Thoracic Cancer
2012
;
3
:
334
40
.
24.
Bai
H
,
Mao
L
,
Wang
HS
,
Zhao
J
,
Yang
L
,
An
TT
, et al
Epidermal growth factor receptor mutations in plasma DNA samples predict tumor response in Chinese patients with stages IIIB to IV non-small-cell lung cancer
.
J Clin Oncol
2009
;
27
:
2653
9
.
25.
Bai
H
,
Zhao
J
,
Wang
SH
,
An
TT
,
Wang
X
,
Wu
MN
, et al
The detection by denaturing high performance liquid chromatography of epidermal growth factor receptor mutation in tissue and peripheral blood from patients with advanced non-small cell lung cancer
.
Zhonghua Jie He He Hu Xi Za Zhi
2008
;
31
:
891
6
.
26.
Whiting
PF
,
Rutjes
AW
,
Westwood
ME
,
Mallett
S
,
Deeks
JJ
,
Reitsma
JB
, et al
QUADAS-2: a revised tool for the quality assessment of diagnostic accuracy studies
.
Ann Intern Med
2011
;
155
:
529
36
.
27.
Jaeschke
R
,
Guyatt
GH
,
Sackett
DL
. 
Users' guides to the medical literature. III. How to use an article about a diagnostic test. B. What are the results and will they help me in caring for my patients? The evidence-based medicine working group
.
JAMA
1994
;
271
:
703
7
.
28.
Fischer
JE
,
Bachmann
LM
,
Jaeschke
R
. 
A readers' guide to the interpretation of diagnostic test properties: clinical example of sepsis
.
Intensive Care Med
2003
;
29
:
1043
51
.
29.
Deeks
JJ
,
Macaskill
P
,
Irwig
L
. 
The performance of tests of publication bias and other sample size effects in systematic reviews of diagnostic test accuracy was assessed
.
J Clin Epidemiol
2005
;
58
:
882
93
.
30.
Dwamena
BA
. 
midas: computational and graphical routines for meta-analytical integration of diagnostic accuracy studies in Stata
.
Division of Nuclear Medicine, Department of Radiology, University of Michigan Medical School
,
Ann Arbor, Michigan
; 
2007
.
31.
Kim
HR
,
Lee
SY
,
Hyun
DS
,
Lee
MK
,
Lee
HK
,
Choi
CM
, et al
Detection of EGFR mutations in circulating free DNA by PNA-mediated PCR clamping
.
J Exp Clin Cancer Res
2013
;
32
:
50
.
32.
Liu
X
,
Lu
Y
,
Zhu
G
,
Lei
Y
,
Zheng
L
,
Qin
H
, et al
The diagnostic accuracy of pleural effusion and plasma samples versus tumour
.
J Clin Pathol
2013
;
66
:
1065
9
.
33.
Lv
C
,
Ma
Y
,
Feng
Q
,
Fang
F
,
Bai
H
,
Zhao
B
, et al
A pilot study: sequential gemcitabine/cisplatin and icotinib as induction therapy for stage IIB to IIIA non-small-cell lung adenocarcinoma
.
World J Surg Oncol
2013
;
11
:
96
.
34.
Kim
ST
,
Sung
JS
,
Jo
UH
,
Park
KH
,
Shin
SW
,
Kim
YH
. 
Can mutations of EGFR and KRAS in serum be predictive and prognostic markers in patients with advanced non-small cell lung cancer (NSCLC)
?
Med Oncol
2013
;
30
:
328
.
35.
Xu
F
,
Wu
J
,
Xue
C
,
Zhao
Y
,
Jiang
W
,
Lin
L
, et al
Comparison of different methods for detecting epidermal growth factor receptor mutations in peripheral blood and tumor tissue of non-small cell lung cancer as a predictor of response to gefitinib
.
Onco Targets Ther
2012
;
5
:
439
47
.
36.
Hu
C
,
Liu
X
,
Chen
Y
,
Sun
X
,
Gong
Y
,
Geng
M
, et al
Direct serum and tissue assay for EGFR mutation in non-small cell lung cancer by high-resolution melting analysis
.
Oncol Rep
2012
;
28
:
1815
21
.
37.
Nakamura
T
,
Sueoka-Aragane
N
,
Iwanaga
K
,
Sato
A
,
Komiya
K
,
Kobayashi
N
, et al
Application of a highly sensitive detection system for epidermal growth factor
.
J Thorac Oncol
2012
;
7
:
1369
81
.
38.
Zhao
X
,
Han
RB
,
Zhao
J
,
Wang
J
,
Yang
F
,
Zhong
W
, et al
Comparison of epidermal growth factor receptor mutation statuses in tissue and plasma in stage I-IV non-small cell lung cancer patients
.
Respiration
2013
;
85
:
119
25
.
39.
Yam
I
,
Lam
DC
,
Chan
K
,
Chung-Man Ho
J
,
Ip
M
,
Lam
WK
, et al
EGFR array: uses in the detection of plasma EGFR mutations in non-small cell lung
.
J Thorac Oncol
2012
;
7
:
1131
40
.
40.
Jiang
B
,
Liu
F
,
Yang
L
,
Zhang
W
,
Yuan
H
,
Wang
J
, et al
Serum detection of epidermal growth factor receptor gene mutations using
.
J Int Med Res
2011
;
39
:
1392
401
.
41.
Taniguchi
K
,
Uchida
J
,
Nishino
K
,
Kumagai
T
,
Okuyama
T
,
Okami
J
, et al
Quantitative detection of EGFR mutations in circulating tumor DNA derived from
.
Clin Cancer Res
2011
;
17
:
7808
15
.
42.
Goto
K
,
Ichinose
Y
,
Ohe
Y
,
Yamamoto
N
,
Negoro
S
,
Nishio
K
, et al
Epidermal growth factor receptor mutation status in circulating free DNA in serum: from IPASS, a phase III study of gefitinib or carboplatin/paclitaxel in non-small cell lung cancer
.
J Thorac Oncol
2012
;
7
:
115
21
.
43.
Sriram
KB
,
Tan
ME
,
Savarimuthu
SM
,
Wright
CM
,
Relan
V
,
Stockwell
RE
, et al
Screening for activating EGFR mutations in surgically resected nonsmall cell lung
.
Eur Respir J
2011
;
38
:
903
10
.
44.
Kuang
Y
,
Rogers
A
,
Yeap
BY
,
Wang
L
,
Makrigiorgos
M
,
Vetrand
K
, et al
Noninvasive detection of EGFR T790M in gefitinib or erlotinib resistant non-small
.
Clin Cancer Res
2009
;
15
:
2630
6
.
45.
Yung
TK
,
Chan
KC
,
Mok
TS
,
Tong
J
,
To
KF
,
Lo
YM
. 
Single-molecule detection of epidermal growth factor receptor mutations in plasma
.
Clin Cancer Res
2009
;
15
:
2076
84
.
46.
He
C
,
Liu
M
,
Zhou
C
,
Zhang
J
,
Ouyang
M
,
Zhong
N
, et al
Detection of epidermal growth factor receptor mutations in plasma by mutant-enriched PCR assay for prediction of the response to gefitinib in patients with non-small-cell lung cancer
.
Int J Cancer
2009
;
125
:
2393
9
.
47.
Kimura
H
,
Kasahara
K
,
Kawaishi
M
,
Kunitoh
H
,
Tamura
T
,
Holloway
B
, et al
Detection of epidermal growth factor receptor mutations in serum as a predictor
.
Clin Cancer Res
2006
;
12
:
3915
21
.
48.
Kimura
H
,
Suminoe
M
,
Kasahara
K
,
Sone
T
,
Araya
T
,
Tamori
S
, et al
Evaluation of epidermal growth factor receptor mutation status in serum DNA as a predictor of response to gefitinib (IRESSA)
.
Br J Cancer
2007
;
97
:
778
84
.
49.
Li
X
,
Ren
R
,
Ren
S
,
Chen
X
,
Cai
W
,
Zhou
F
, et al
Peripheral blood for epidermal growth factor receptor mutation detection in non-small cell lung cancer patients
.
Transl Oncol
2014
;
7
:
341
8
.
50.
Douillard
JY
,
Ostoros
G
,
Cobo
M
,
Ciuleanu
T
,
Cole
R
,
McWalter
G
, et al
Gefitinib treatment in EGFR mutated caucasian NSCLC: circulating-free tumor DNA as a surrogate for determination of EGFR status
.
J Thorac Oncol
2014
;
9
:
1345
53
.
51.
Weber
B
,
Meldgaard
P
,
Hager
H
,
Wu
L
,
Wei
W
,
Tsai
J
, 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
.
52.
Swets
JA
. 
Measuring the accuracy of diagnostic systems
.
Science
1988
;
240
:
1285
93
.
53.
Glas
AS
,
Lijmer
JG
,
Prins
MH
,
Bonsel
GJ
,
Bossuyt
PM
. 
The diagnostic odds ratio: a single indicator of test performance
.
J Clin Epidemiol
2003
;
56
:
1129
35
.
54.
Diehl
F
,
Schmidt
K
,
Choti
MA
,
Romans
K
,
Goodman
S
,
Li
M
, et al
Circulating mutant DNA to assess tumor dynamics
.
Nat Med
2008
;
14
:
985
90
.
55.
Li
M
,
Diehl
F
,
Dressman
D
,
Vogelstein
B
,
Kinzler
KW
. 
BEAMing up for detection and quantification of rare sequence variants
.
Nat Methods
2006
;
3
:
95
7
.