Skip to main content
PLOS ONE logoLink to PLOS ONE
. 2021 Mar 26;16(3):e0248775. doi: 10.1371/journal.pone.0248775

The diagnostic accuracy of digital PCR, ARMS and NGS for detecting KRAS mutation in cell-free DNA of patients with colorectal cancer: A systematic review and meta-analysis

Peng Ye 1,*, Peiling Cai 1, Jing Xie 2, Yuanyuan Wei 3,*
Editor: Anthony F Shields4
PMCID: PMC7997033  PMID: 33770081

Abstract

Introduction

Before anti-EGFR therapy is given to patients with colorectal cancer, it is required to determine KRAS mutation status in tumor. When tumor tissue is not available, cell-free DNA (liquid biopsy) is commonly used as an alternative. Due to the low abundance of tumor-derived DNA in cell-free DNA samples, methods with high sensitivity were preferred, including digital polymerase chain reaction, amplification refractory mutation system and next-generation sequencing. The aim of this systemic review and meta-analysis was to investigate the accuracy of those methods in detecting KRAS mutation in cell-free DNA sample from patients with colorectal cancer.

Methods

Literature search was performed in Pubmed, Embase, and Cochrane Library. After removing duplicates from the 170 publications found by literature search, eligible studies were identified using pre-defined criteria. Quality of the publications and relevant data were assessed and extracted thereafter. Meta-DiSc and STATA softwares were used to pool the accuracy parameters from the extracted data.

Results

A total of 33 eligible studies were identified for this systemic review and meta-analysis. After pooling, the overall sensitivity, specificity, and diagnostic odds ratio were 0.77 (95%CI: 0.74–0.79), 0.87 (95%CI: 0.85–0.89), and 23.96 (95%CI: 13.72–41.84), respectively. The overall positive and negative likelihood ratios were 5.55 (95%CI: 3.76–8.19) and 0.29 (95%CI: 0.21–0.38), respectively. Area under curve of the summarized ROC curve was 0.8992.

Conclusion

Digital polymerase chain reaction, amplification refractory mutation system, and next-generation sequencing had overall high accuracy in detecting KRAS mutation in cell-free DNA sample. Large prospective randomized clinical trials are needed to further convince the accuracy and usefulness of KRAS mutation detection using cfDNA/liquid biopsy samples in clinical practice.

Trial registration

PROSPERO CRD42020176682; https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=176682.

Introduction

Colorectal cancer (CRC) is currently a leading cause of cancer-related death worldwide [1]. For resectable CRC, surgery remains the standard of care, while for non-resectable tumors, patients are mostly treated by chemotherapy and targeted therapy, e.g. anti-epithelial growth factor receptor (EGFR) therapy [2,3]. As examples of anti-EGFR therapy, cetuximab and panitumumab were firstly approved for the treatment of chemorefractory metastatic CRC (mCRC) in 2004 and 2006, respectively [2]. In subsequent investigations of the two drugs as second-line treatment of mCRC, several large phase III clinical trials showed benefit in response rate and progression-free survival, but not in overall survival [46]. Retrospective analysis on the results of those trials revealed that mCRC patients with different molecular background differed significantly in treatment response. The first finding was that mutations in KRAS exon 2 were linked to poor response in mCRC patients treated by anti-EGFR therapy [7]. Later, mutations in KRAS exon 3 and 4 (codons 61, 117, and 146), and in NRAS exons 2, 3, 4 were also found to be associated with resistance to anti-EGFR therapy in mCRC patients [8,9]. Retrospective analysis showed that patients with RAS wild-type tumors had significant benefit in all efficacy end points, while no significant benefit was observed in patients with RAS mutated tumors [10]. Those compelling evidences led the administrations to limit the use of cetuximab and panitumumab only in mCRC patients with both KRAS wild-type and NRAS wild-type, and require the test of KRAS and NRAS mutation status before anti-EGFR treatment is given [2,11].

The detection of KRAS and NRAS mutation in CRC is mostly performed on archived surgical tumor tissue samples or tumor biopsy samples using traditional point mutation detection methods (e.g. quantitative polymerase chain reaction, quantitative PCR and amplification refractory mutation system, ARMS) or sequencing technique (e.g. Sanger sequencing) [1214]. However, for refractory CRC or mCRC patients, tumor tissue samples are often not available. A small proportion of cell-free DNA (cfDNA) in liquid biopsy sample (plasma, urine, and etc.) derives from tumor cells (also called circulating tumor DNA, ctDNA), and could serve as an alternative source of tumor-derived DNA to surgical tumor tissue sample or tumor biopsy sample [15,16].

The liquid biopsy-based tumor genotyping has been intensively studied in recent years [17]. Due to the low abundance of ctDNA, several high-sensitivity techniques have been developed and evaluated for the tumor genotyping in liquid biopsy samples, including digital PCR, ARMS, and next generation sequencing (NGS) [1820]. ARMS is already commonly used in clinical laboratories, with acceptable accuracy and low cost [21]. The high-throughput technology, NGS, has the ability to detect hundreds of mutations in a run, but is challenged by its relatively low sensitivity and high cost [22]. Digital PCR is well known by its high sensitivity, but the cost of this technique is still higher than traditional quantitative PCR [23]. For the detection of KRAS mutations, the limit of detection for ARMS, NGS, and digital PCR was reported to be 1%, 2–6%, or as low as 0.01%, respectively [12,22,24,25]. However, although the limit of detection of those techniques was determined, their performance in clinical practice has not been fully validated yet. The aim of this systematic review and meta-analysis was to investigate the accuracy of KRAS mutation detection in cfDNA samples from patients with CRC, compared to paired tissue samples. After searching of eligible studies in databases, and subsequent extraction and analysis of data, those techniques (digital PCR, ARMS, and NGS) showed overall high accuracy in detecting KRAS mutation in cfDNA samples of colorectal cancer patients.

Materials and methods

Registration and publication of study protocol

Study protocol of this systemic review and meta-analysis has been registered on International prospective register of systemic reviews (PROSPERO) and the registration number is CRD42020176682. Detailed study protocol has been published [26].

Literature searching and selection of publication

Literature research was performed independently by PY and PC in June 2020, and no limitation was placed on publication date. Pubmed, Embase, and Cochrane Library were searched using “KRAS”, “digital PCR”, “next-generation sequencing”, “amplification refractory mutation system”, “cell-free DNA”, “circulating tumor DNA”, “liquid biopsy”, and “colorectal cancer”. Alternative spelling or abbreviations were also included in the search (see S1 Table for detailed search strategy). In the searching results, we firstly reviewed the titles and abstracts of the publications. Duplicated publications were removed and irrelevant publications were excluded using the following criteria: 1) not a human study; 2) not describing KRAS mutation; 3) no liquid biopsy samples or tissue samples included; 4) did not use any techniques among digital PCR, ARMS, and NGS; 5) not colorectal cancer; 6) reviews, abstracts, letter to the editor, comments, case reports, or studies with un-interpretable data.

For the remaining publications, full texts were downloaded and examined. Studies were further excluded if 1) data were un-interpretable or mixed (cannot be separated from results of other gene mutations); 2) lacking KRAS mutated or KRAS wild-type tissue samples; 3) no defined criteria for the positively/negativity of KRAS mutation. In the resulting eligible studies, KRAS testing results from paired cfDNA samples and tumor tissue samples (KRAS mutated or wild-type) were extracted from each publication, including sample size, and numbers of true positive, false positive, false negative, and true negative. Sample types of cfDNA (e.g. plasma, serum, etc.) and techniques used for cfDNA samples or tissue samples were also extracted from each of the eligible studies. If multiple techniques were used to determine KRAS mutation status in the same patient cohort, one technique was selected for data extraction using the following criteria: 1) technique used for a larger number of samples; 2) technique having similar detection region with the technique used for KRAS detection in paired tissue samples. Other information was also extracted and recorded, including age of patients, race of patients (Caucasian, Asian, etc.), country of origin (region of the study), type of CRC (metastatic or non-metastatic), and name of the first author of the publication. Disagreement in the literature search results between the two researchers (PY and PC) was solved by a third researcher (YW). Each of the eligible studies included in the data extraction was evaluated using quality assessment of diagnostic accuracy studies 2 (QUADAS-2) [27].

Statistical analysis

Sensitivity, specificity, positive likelihood ratio (PLR), negative likelihood ratio (NLR), and diagnostic odds ratio (DOR) of the eligible studies were pooled using Meta-DiSc statistical software version 1.4 [28], and summary receiver operating characteristic (SROC) and area under curve (AUC) were also generated. Cochran-Q and I2 were used to evaluate inter-study heterogeneity. Random effects model (DerSimonian-Laird) was used for pooling the results if significant heterogeneity was observed (I2 ≥ 50% and P ≤ 0.05), while fixed effects model (Mantel-Haenszel) was used if no significant heterogeneity was identified. Threshold analysis and meta-regression were performed using Meta DiSc to search for potential source of heterogeneity. Publication bias was evaluated using Deek’s funnel plot asymmetry test performed by STATA 12.0 (STATA Corp.). P < 0.05 was considered statistically significant.

Results

Search results

After literature searching, a total of 170 publications were found from Pubmed (73 publications), Embase (77 publications), and Cochrane Library (20 publications), as shown in Fig 1. After removal of duplicated (62 publications) and irrelevant publications (53 publications), full text of the rest 55 publications were reviewed and another 22 studies were excluded due to lack of KRAS mutated/wild-type tissue samples or un-interpretable data. Data were extracted from the rest 33 eligible studies and meta-analysis was performed.

Fig 1. PRISMA 2009 flow diagram.

Fig 1

From: Moher D, Liberati A, Tetzlaff J, Altman DG, The PRISMA Group (2009). Preferred Reporting Items for Systematic Reviews and Meta-Analyses: The PRISMA Statement. PLoS Med 6(7): e1000097. doi:10.1371/journal.pmed1000097 For more information, visit www.prisma-statement.org.

Review of eligible publications

As shown in Table 1, in the 33 eligible studies, KRAS status in cfDNA samples was tested using NGS in 15 studies, digital PCR in 17 studies, or ARMS in 1 study. All the eligible studies used from plasma samples, except for the study by Kitagawa et al [14] which used serum instead. Due to the emergence of the All-RAS sequencing concept, many of the studies tested both KRAS and NRAS (and even HRAS), as well as the expanded isoforms of those genes [19,2947]. Most of those studies reported separate results for KRAS, and the rest 6 studies reported All-RAS status [30,40,41,4446]. For those 6 studies, NRAS results were also included in the subsequent systematic review and meta-analysis. The accuracy of KRAS/All-RAS status detection in cfDNA samples in each study is summarized below.

Table 1. Summary of studies comparing KRAS/All-RAS mutation status in cfDNA and tumor tissue samples from colorectal cancer patients.

Author, year Sample size Technique used for cfDNA samples Sample type for cfDNA Technique used for tissue samples Region of the study Type of colorectal caner
Bidard et al, 2019 [48] 125 digital droplet PCR (Bio-Rad) Plasma standard routine technique Europe metastatic
Sclafani et al, 2018 [49] 90 digital droplet PCR (Bio-Rad) Plasma PCR & Sanger sequencing Europe primary
Kang et al, 2020 [13] 48 NGS (customized panel) Plasma Sanger sequencing Asia metastatic
Kato et al, 2019 [19] 76 NGS (Guardant360, Guardant Health) Plasma NGS (Foundation One, Foundation Medicine) America either primary or metastatic
Chang et al, 2018 [29] 5 NGS (275-gene panel from Qiagen) Plasma NGS (275-gene panel from Qiagen) Asia either primary or metastatic
Garcia et al, 2018 [30] 28 Beads, Emulsion, Amplification and Magnetics (BEAMing) (OncoBEAM-RAS-CRC kita) Plasma NGS (customized Ampliseq library,) Europe metastatic
Wang et al, 2017 [31] 97 NGS (commercial panel from SinoMD) Plasma standard routine technique Asia metastatic
Kidess-Sigal et al, 2016 [50] 3 NGS (SCODAb mutation enrichment and detection technology) Plasma Sanger sequencing America metastatic
Kim et al, 2015 [32] 29 NGS (Guardant360, Guardant Health) Plasma Sanger sequencing Asia metastatic
Vessies et al, 2020 [33] 6 BEAMing (OncoBEAM-RAS-CRC kita) Plasma standard of care Europe metastatic
Cao et al, 2020 [34] 35 NGS (customized 605-gene panel) Plasma NGS (whole exome sequencing) Asia either primary or metastatic
Gupta et al, 2020 [35] 75 NGS (Guardant360, Guardant Health) Plasma NGS (Foundation One, Foundation Medicine) America metastatic
Kitagawa et al, 2019 [14] 40 digital droplet PCR (Bio-Rad) Serum ARMS or Luminex Asia either primary or metastatic
Galbiati et al, 2019 [51] 20 digital droplet PCR (Bio-Rad) Plasma MassARRAY (Sequenom) Europe metastatic
Choi et al, 2019 [36] 61 NGS (Guardant360, Guardant Health) Plasma NGS (Foundation One, Foundation Medicine) America either primary or metastatic
Liebs, et al, 2019 [18] 53 digital droplet PCR (Bio-Rad) Plasma digital droplet PCR (Bio-Rad) Europe either primary or metastatic
Osumi et al, 2019 [37] 101 NGS (Oncomine Colon cfDNA assay) Plasma PCR (RASKET KIT, Luminex) Asia metastatic
Galbiati et al, 2019 [52] 30 digital droplet PCR (Bio-Rad) Plasma MassARRAY (Sequenom) Europe metastatic
Takayama et al, 2018 [53] 85 digital droplet PCR (Bio-Rad) Plasma ARMS/PCR (RASKET kit) Asia metastatic
Yao et al, 2018 [38] 64 NGS (Sureselect, Agilent, targeting KRAS/NRAS/HRAS/BRAF) Plasma ARMS (Human KRAS/NRAS/BRAF mutations detection kitc) Asia metastatic
Sun et al, 2018 [39] 11 NGS (customized 85-gene colorectal cancer gene panel) Plasma NGS (customized 85-gene colorectal cancer gene panel) Asia either primary or metastatic
Normanno et al, 2018 [40] 92 BEAMing (OncoBEAM-RAS-CRC kita) Plasma NGS (AmpliSeq Colon and Lung Cancer Panel, ThermoFisher) Europe metastatic
Garcia-Foncillas et al, 2018 [41] 236 BEAMing (OncoBEAM-RAS-CRC kita) Plasma standard of care (Pyrosequencing/Cobas/Therascreen/Idylla/CLART-CMA kit) Europe metastatic
Beije et al, 2016 [42] 12 NGS (customized 21-gene CRC-specific panel) Plasma NGS (customized 21-gene CRC-specific panel) Europe metastatic
Sefrioui et al, 2017 [54] 29 QuantStudio 3D digital PCR Plasma SNaPshot multiplex assay Europe metastatic
Grasselli et al, 2017 [43] 117 BEAMing (OncoBEAM-RAS-CRC kita) Plasma BEAMing (OncoBEAM-RAS-CRC kita) Europe metastatic
Schmiegel et al, 2017 [44] 98 BEAMing (OncoBEAM-RAS-CRC kita) Plasma standard of care (Pyrosequencing/Sanger sequencing/NGS) Europe metastatic
Vidal et al, 2017 [45] 115 BEAMing (OncoBEAM-RAS-CRC kita) Plasma standard of cared Europe metastatic
Beranek et al, 2016 [47] 32 NGS (Somatic 1e) Plasma Standard routine technique Europe metastatic
Rachiglio et al, 2016 [46] 35 NGS (22-gene Oncomine Solid Tumor DNA kit, Life Technologies) Plasma Pyrosequencing (Therascreen KRAS and NRAS Pyro kit, Qiagen) Europe metastatic
Yamada et al, 2016 [55] 94 QuantStudio 3D digital PCR Plasma MEBGEN-Luminex method Asia metastatic
Spindler et al, 2015 [20] 211 ARMS-based in-house assay Plasma ARMS-based in-house assay Europe metastatic
Taly et al, 2013 [12] 50 picodroplet digital PCR Plasma quantitative PCR Europe metastatic

aFrom Sysmex Inostics, which targets 34 variants in KRAS and NRAS genes.

bSequence-specific synchronous coefficient of drag alteration.

cFrom Beijing ACCB Biotech, which targets KRAS/NRAS codons 12, 13, 59, 61, 117, 146, and BRAF codon 600.

dTherascreen KRAS RGQ PCR kit (Qiagen), COBAS KRAS mutation test (Roche), or pyrosequencing (PyroMark, Qiagen).

eFrom Multiplicom, Belgium, which targets BRAF, KRAS, and NRAS.

NGS

In the 15 studies using NGS to measure KRAS status in cfDNA samples, studies by Kato [19], Kim [32], Gupta [35], or Choi [36] used commercial Guardant360 NGS panel (Guardant Health Inc.), and results showed sensitivity of 59.5%, 83.3%, 75.9%, or 63.6%, and specificity of 87.2%, 86.9%, 97.8%, or 92.9%, respectively. Three of those 4 studies used Foundation One NGS panel (Foundation Medicine) to test KRAS mutation status in paired tissue samples [19,35,36], while the other study by Kim [32] used traditional Sanger sequencing instead. Studies by Osumi [37], or Rachiglio [46] used commercial Oncomine NGS panels (Life Technologies, covering 14 genes or 22 genes, respectively) to detect KRAS or All-RAS status in cfDNA samples of CRC patients, and the sensitivity was 80.6% or 63.2%, and specificity was 81.5% or 100%, respectively. Beranek et al [47] used another commercial NGS panel (Somatic 1, Multiplicom, Belgium) in a 12-patient cohort. The concordance rate was 86% between cfDNA and tumor tissue samples, and the calculated sensitivity and specificity were 80% and 100%, respectively.

The rest 8 studies all used customized NGS panels for cfDNA samples. Kang et al [13] performed a liquid-biopsy-based tumor profiling using a 10-gene NGS panel in 48 mCRC patients, and the calculated sensitivity and specificity were 86.4% and 65.4%. Chang et al [29] analyzed correlation of genomic alterations between paired tumor tissue and plasma samples using a 275-gene NGS panel in 21 patients with different cancer types. In the 5 patients with CRC, the calculated concordance rate of KRAS status was 60% (3/5), with 2 false-negative cases. Wang et al [31] investigated the KRAS mutation status in paired plasma and tumor tissue samples of 97 mCRC patients using NGS, and results showed concordance rate of 65.26%, sensitivity of 70.02%, and specificity of 66.71%. In the study by Kidess-Sigal et al [50], the authors compared KRAS, BRAF, and PIK3CA status between circulating tumor cells, ctDNA, and tissue samples of 15 mCRC patients using a 4-gene NGS panel, and in the 3 patients with both ctDNA and primary tumor samples available, the calculated concordance rate for KRAS was 66.7% (2/3), with 1 false-positive case. In the study by Cao et al [34], a 605-gene NGS panel was used to analyze tumor and plasma samples of CRC patients, and from 35 patients with KRAS status available in both tumor and plasma, the calculated sensitivity was 75.0% and specificity was 82.6%. Using a customized targeted NGS library kit (SureSelect QXT, Agilent Technologies, USA), Yao et al [38] investigated KRAS/NRAF/BRAF mutations in plasma and tumor samples of mCRC patients, and concordance rate of KRAS was 81.25% in 64 patients, with sensitivity of 66.7% and specificity of 90.0%. Sun et al [39] used an 85-gene NGS panel and analyzed paired tumor and plasma samples of 11 CRC patients, and results showed calculated sensitivity of 80% and specificity of 100% in the detection of KRAS status in cfDNA. Beije et al [42] designed a CRC-specific 21-gene NGS panel and plasma samples from 12 mCRC patients analyzed by this panel showed calculated sensitivity of 50% and specificity of 87.5% in KRAS mutation detection.

Digital PCR

Digital PCR was used to detect KRAS status in cfDNA samples in 17 of the eligible studies. Within those studies, 7 publications by Bidard [48], Sclafani [49], Kitagawa [14], Galbiati [51,52], Liebs [18], or Takayama [53] used digital droplet PCR (Bio-Rad) targeting KRAS point mutations in cfDNA samples, and the calculated sensitivity of this technique ranged from 36% to 100% (91.3%, 42.9%, 94.1%, 100%, 66.7%, 36%, or 79.3%, respectively) and specificity ranged from 50% to 100% (92.4%, 64.5%, 100%, 92.3%, 66.7%, 100%, 50%, respectively).

Seven studies by Garcia [30], Vessies [33], Normanno [40], Grasselli [43], Schmiegel [44], Vidal [45], or García-Foncillas [41] used BEAMing digital PCR technology. All those studies used a commercial OncoBEAM RAS CRC Kit (Sysmex Inostics) which could target 34 somatic mutations in KRAS/NRAS exons 2, 3, 4 in one run. Study by Garcia et al [30] compared the accuracy of different platforms, including BEAMing which showed sensitivity of 93.3% and specificity of 69.2% in detecting mutations in KRAS/NRAS in plasma samples, compared to tissue sample results. From a small patient cohort (6 patients), Vessies et al [33] investigated the performance of 4 platforms and BEAMing platform correctly identified KRAS mutations in plasma samples of 4 patients, with 1 false positive and 1 false negative cases. Normanno et al [40] analyzed All-RAS mutations in plasma samples from a sub-cohort of patients from a clinical trial (CAPRI-GOIM study) using BEAMing and digital droplet PCR, and the results revealed a sensitivity of 70.0% and specificity of 83.1% of BEAMing platform. Grasselli et al [43] also investigated the performance of BEAMing in detecting KRAS mutation in plasma and tissue of metastatic colorectal cancer and results showed sensitivity of 85.7%, specificity of 94%, and concordance rate of 89.7%. The rest 3 studies by Schmiegel [44], Vidal [45], or García-Foncillas [41] investigated All-RAS status in plasma samples from mCRC patients using BEAMing and compared the results with paired tissue samples. The results showed concordance rate of 91.8%, 93%, or 89%, sensitivity of 90.4%, 96.4%, or 86.3%, and specificity of 93.5%, 90%, or 92.4%, respectively.

In the rest 3 publications, 2 studies by Sefrioui [54] or Yamada [55] used chip-based digital PCR platform (QuantStudio 3D Digital PCR System, Thermo Fisher Scientific) in the detection of KRAS status in plasma, and obtained sensitivity of 85.7% or 79.5%, and specificity of 100% or 90.9%, respectively. Taly et al [12] used picodroplet digital PCR technique in detecting KRAS mutation in plasma samples from 50 mCRC patients, and results showed calculated sensitivity of 73.7% and specificity of 93.5%.

ARMS

Only 1 study used ARMS to detect KRAS status in cfDNA samples. Spindler et al [20] used ARMS method to detect KRAS mutation in matched tumor tissue and plasma samples from 211 mCRC patients, and the overall concordance rate was 85.0%, with sensitivity of 80.0% and specificity of 95.8%.

In conclusion, the 33 studies comprised 2203 CRC patients with paired cfDNA and tumor tissue samples. Out of the 33 eligible studies, 20 showed high concordance (higher than 80%) in KRAS detection results between cfDNA and tumor tissue samples. High specificity (higher than 80%) was also observed in majority (26 out of 29) of the studies. More than half (17 out of 33) of the studies showed high sensitivity (higher than 80%).

Quality assessment of eligible studies

QUADAS-2 was used to assess the quality of individual studies and the result is shown in Table 2. In the four aspects of risk of bias assessment, percentage of high risk of bias was from 0% (n = 0, patient selection, reference standard) to 15% (n = 5, flow and timing). Percentage of low risk of bias was from 24% (n = 8, index test, flow and timing) to 73% (n = 24, patient selection). Flow and timing showed the highest risk of bias (15% high risk of bias and 24% low risk of bias). Patient selection showed the lowest risk of bias (0% high risk of bias and 73% low risk of bias). For the applicability of studies, all the studies were classified as low concern in the three aspects (patient selection, index test, and reference standard).

Table 2. QUADAS-2 assessment of eligible studies.

Author, year Risk of bias Applicability concerns
Patient selection Index test Reference standard Flow and timing Patient selection Index test Reference standard
Bidard et al, 2019 [48] low low low unclear low low low
Sclafani et al, 2018 [49] low unclear low high low low low
Kang et al, 2020 [13] low unclear unclear high low low low
Kato et al, 2019 [19] low unclear unclear high low low low
Chang et al, 2018 [29] unclear high low unclear low low low
Garcia et al, 2018 [30] low low low low low low low
Wang et al, 2017 [31] unclear unclear low unclear low low low
Kidess-Sigal et al, 2016 [50] low unclear unclear unclear low low low
Kim et al, 2015 [32] low unclear low unclear low low low
Vessies et al, 2020 [33] low unclear low high low low low
Cao et al, 2020 [34] low unclear unclear unclear low low low
Gupta et al, 2020 [35] low low low unclear low low low
Kitagawa et al, 2019 [14] unclear unclear unclear unclear low low low
Galbiati et al, 2019 [51] low unclear low unclear low low low
Choi et al, 2019 [36] low low low unclear low low low
Liebs, et al, 2019 [18] unclear unclear unclear unclear low low low
Osumi et al, 2019 [37] low unclear unclear unclear low low low
Galbiati et al, 2019 [52] unclear unclear low low low low low
Takayama et al, 2018 [53] unclear unclear low unclear low low low
Yao et al, 2018 [38] low unclear low low low low low
Sun et al, 2018 [39] low low low low low low low
Normanno et al, 2018 [40] low unclear unclear unclear low low low
García-Foncillas et al, 2018 [41] unclear unclear low unclear low low low
Beije et al, 2016 [42] low low low low low low low
Sefrioui et al, 2017 [54] low unclear low high low low low
Grasselli et al, 2017 [43] low low low unclear low low low
Schmiegel et al, 2017 [44] low unclear unclear low low low low
Vidal et al, 2017 [45] low unclear low unclear low low low
Beranek et al, 2016 [47] unclear unclear unclear unclear low low low
Rachiglio et al, 2016 [46] low unclear unclear low low low low
Yamada et al, 2016 [55] low low low low low low low
Spindler et al, 2015 [20] low unclear unclear unclear low low low
Taly et al, 2013 [12] unclear unclear unclear unclear low low low

Meta-analysis of the accuracy of KRAS mutation detection using cfDNA samples

The KRAS detection results of the 2203 CRC patients were pooled using statistical software. As shown in Fig 2, results showed a pooled sensitivity of 0.77 [95% confidence interval (CI): 0.74–0.79] and pooled specificity of 0.87 (95%CI: 0.85–0.89). The pooled PLR, NLR and DOR were 5.55 (95%CI: 3.76–8.19), 0.29 (95%CI: 0.21–0.38), and 23.96 (95%CI: 13.72–41.84), respectively. SROC curve was also generated and the AUC was 0.8992.

Fig 2. Pooled sensitivity, specificity, PLR, NLR, DOR and SROC of the eligible studies.

Fig 2

All the forest plots of the meta-analysis (see Fig 2) showed significant inter-study heterogeneity (I2 ≥ 50% and P ≤ .05), indicating significant differences among the studies. Therefore, we focused more on the possible sources of inter-study heterogeneity and subgroup analysis. The Spearman correlation coefficient was -0.053 (P = 0.77), suggesting no significant threshold effect. In the meta-regression analysis, we included 5 covariates (technique used for cfDNA samples, technique used for tissue samples, region, race, type of CRC), and results indicated that inter-study heterogeneity was not associated to technique used for cfDNA samples (P = 0.85), technique used for tissue samples (P = 0.22), region (P = 0.91), race of patients (P = 0.68), and type of CRC (P = 0.20). Age of patients was excluded from the meta-regression because clear age data were not provided in several studies [33,51,52,54,55]. Type of liquid biopsy samples was also not included as a covariate in the meta-regression since almost all the eligible studies used plasma samples (only 1 study used serum samples instead [14]).

Subgroup analysis was conducted according to type of CRC. Several studies involved in this systemic review and meta-analysis attempted to investigate possible roles of liquid biopsy in early detection or monitoring of the disease (e.g. driver mutations, resistance to targeted therapeutics), and therefore included CRC patients of different stages (I-IV) [14,18,19,29,34,36,39]. Since both primary and metastatic CRC patients were involved in those studies [14,18,19,39], we extracted their data separately. Three studies were excluded from the subgroup analysis because we cannot separate their data by groups of primary and metastatic CRC [29,34,36]. As shown in Table 3, compared to primary CRC, mCRC showed higher pooled sensitivity [0.79 (95%CI: 0.76–0.82)] and specificity [0.88 (95%CI: 0.86–0.90)]. The pooled DOR [29.17 (95%CI: 17.00–50.06)] and AUC of SROC curve (0.9045) of mCRC were also higher than that of primary CRC [10.81 (95%CI: 1.00–117.04) and 0.7304]. For the comparison between early- (I, II) and late-stage (III, IV) CRC, we only successfully extracted accuracy data from 3 studies, including 1 study which lacked true positive cases and therefore was excluded by statistical software. Due to the limited number of studies (2 studies only), we did not continue the pooling of accuracy data in early-stage CRC group and the comparison between early- and late-stage CRC.

Table 3. Meta-analysis results.

No. of studies/patient cohorts Sensitivity Specificity PLR NLR DOR AUC of SROC
Overall 33 0.77(0.74–0.79) 0.87(0.85–0.89) 5.55(3.76–8.19) 0.29(0.21–0.38) 23.96(13.72–41.84) 0.8992
Type of CRC
metastatic 24a 0.79(0.76–0.82) 0.88(0.86–0.90) 5.14(3.32–7.98) 0.26(0.19–0.35) 29.17(17.00–50.06) 0.9045
primary 4b 0.57(0.41–0.72) 0.73(0.62–0.82) 2.08(1.25–3.44) 0.46(0.20–1.07) 10.81(1.00–117.04) 0.7304
Technique used for cfDNA samples
NGS 15 0.65(0.59–0.71) 0.88(0.85–0.91) 5.21(3.97–6.83) 0.38(0.28–0.52) 14.61(9.78–21.84) 0.8574
Digital PCR 17 0.81(0.78–0.85) 0.85(0.83–0.88) 5.51(3.02–10.07) 0.23(0.14–0.37) 29.18(11.79–72.25) 0.9067
NGS (Guardant360) 4 0.67(0.57–0.76) 0.92(0.86–0.96) 8.49(4.66–15.46) 0.35(0.26–0.47) 22.47(10.58–47.75) 0.8260
BEAMing 7 0.87(0.83–0.90) 0.90(0.86–0.93) 5.94(2.86–12.34) 0.15(0.09–0.27) 50.96(18.56–139.92) 0.9388
Digital droplet PCR 7 0.71(0.64–0.78) 0.78(0.72–0.83) 4.37(1.66–11.53) 0.33(0.16–0.69) 18.20(3.45–96.00) 0.8703
Region of the study
Europe 18 0.80(0.77–0.83) 0.89(0.87–0.92) 6.42(3.63–11.35) 0.25(0.16–0.39) 32.97(13.63–79.79) 0.9143
Asia 11 0.71(0.65–0.77) 0.81(0.77–0.85) 4.19(2.48–7.07) 0.31(0.19–0.53) 13.87(7–27.48) 0.8539
America 4 0.66(0.56–0.75) 0.92(0.86–0.96) 5.99(1.87–19.15) 0.37(0.28–0.49) 19.98(9.26–43.13) 0.6545
Region of studies using NGS for cfDNA samples
Europe 3 0.64(0.44–0.81) 0.98(0.90–1.00) 15.59(3.57–68.07) 0.37(0.23–0.62) 35.79(6.76–189.46) 0.1279
Asia 8 0.64(0.56–0.72) 0.84(0.79–0.88) 3.95(2.92–5.33) 0.36(0.20–0.65) 11.21(6.81–18.46) 0.8683
America 4 0.66(0.56–0.75) 0.92(0.86–0.96) 5.99(1.87–19.15) 0.37(0.28–0.49) 19.98(9.26–43.13) 0.6545
Region of studies using digital PCR for cfDNA samples
Europe 14 0.81(0.78–0.85) 0.88(0.85–0.90) 5.48(2.98–10.05) 0.23(0.12–0.41) 29.63(10.54–83.32) 0.9119
Asia 3 0.82(0.73–0.90) 0.75(0.67–0.82) 6.59(0.78–55.14) 0.23(0.15–0.37) 28.33(2.87–279.59) 0.8758
Subtypes of digital PCR in studies from Europe
BEAMing 7 0.87(0.83–0.90) 0.90(0.86–0.93) 5.94(2.86–12.34) 0.15(0.09–0.27) 50.96(18.56–139.92) 0.9388
Digital droplet PCR 5 0.66(0.57–0.74) 0.83(0.77–0.88) 4.63(1.31–16.40) 0.38(0.17–0.89) 16.70(1.77–157.26) 0.8750

aOne study/patient cohort [39] was excluded by statistical software due to lack of true positive samples.

bOne study/patient cohort [18] was excluded by statistical software due to lack of true positive samples.

Subgroup analysis was also conducted according to techniques used for cfDNA samples. ARMS was excluded from the subgroup analysis because only 1 study used this technique [20]. After pooling, digital PCR showed higher sensitivity [0.81 (95%CI: 0.78–0.85)] but slightly lower specificity [0.85 (95%CI: 0.83–0.88)], compared to NGS [0.65 (95%CI: 0.59–0.71), 0.88 (95%CI: 0.85–0.91), respectively] (see Table 3). Pooled DOR of digital PCR [29.18 (95%CI: 11.79–72.25)] and AUC of SROC curve (0.9067) were also higher than that of NGS [14.61 (95%CI: 9.78–21.84) and 0.8574]. In subtypes of NGS, a commercial Guardant360 (Guardant Health) NGS panel showed slightly higher sensitivity, specificity, and DOR than the overall accuracy of NGS (Table 3). In subtypes of digital PCR, BEAMing showed higher sensitivity, specificity, and DOR than digital droplet PCR (see Table 3). Other subtypes were excluded from the analysis due to limited number of studies.

In addition, we also conducted subgroup analysis across different regions of the studies. Results showed the highest sensitivity [0.80 (95%CI: 0.77–0.83)] and DOR [32.97 (95%CI: 13.63–79.79)] in studies from Europe, and the lowest sensitivity in studies from America [0.66 (95%CI: 0.56–0.75)] and lowest DOR [13.87 (95%CI: 7–27.48)] in studies from Asia (see Table 3). The specificity was highest in America [0.92 (95%CI: 0.86–0.96)] and lowest in Asia [0.81 (95%CI: 0.77–0.85)]. After looking into the techniques used for cfDNA samples across different regions, we found that majority of the studies from Europe (14 out of 18) used digital PCR, while majority of the studies from Asia (8 of the 11) and all studies from America (4 out of 4) used NGS instead. Therefore, we further grouped the studies by both region and techniques used for cfDNA samples. Interestingly, after taking techniques into consideration, the sensitivity of NGS was similar across different regions, and sensitivity and DOR of digital PCR were also similar between Europe and Asia (Table 3). Those results indicate that the difference in accuracy among different regions of the studies may be partially explained by the significant difference in the techniques used for cfDNA samples across the regions, although we did observe differences in specificity, and PLR (and also DOR in NGS) across the regions. Further investigation on the subtypes of digital PCR revealed that in studies from Europe, BEAMing had higher sensitivity, specificity, and DOR, compared to digital droplet PCR (see Table 3). Due to limited number of studies, we did not further compare the accuracy of BEAMing and digital droplet PCR in Asia and America, or different panels of next-generation sequencing in all the three regions.

Since this study is investigating diagnostic accuracy, we used Deek’s funnel plot asymmetry test to evaluate publication bias (see Deek’s funnel plot in Fig 3). The test results indicated no significant publication bias (P = 0.12).

Fig 3. Deek’s funnel plot.

Fig 3

Discussion

Since anti-EGFR therapies (cetuximab and panitumumab) showed benefit only in RAS wild-type mCRC patients, the precise measurement of RAS mutation status in tumor is very important for the success of the targeted therapy [9,10]. Tumor tissue is commonly used for the detection of RAS mutation, but in some mCRC patients, tumor tissue is not available. Liquid biopsy sample (or cfDNA) has emerged as an alternative for the determination of RAS mutation status [15,16]. However, its accuracy needs to be validated using paired tissue sample as reference (“gold standard”).

Many investigations have validated the accuracy of KRAS/All-RAS mutation detection using liquid biopsy samples. Traditional techniques (e.g. PCR, direct sequencing) were mostly used in the early investigations [56]. In recent years, most of the studies used more sensitive methods, including digital PCR, NGS, and ARMS techniques, and this systemic review and meta-analysis focused on those studies. Thirty-three eligible studies have been involved in our study after database searching and screening. After pooling, the overall sensitivity and specificity of KRAS mutation detection using cfDNA samples were 77% and 87%, respectively. The important indicator of diagnostic test [57], DOR, was 23.96, and AUC of SROC curve was 0.8992. Those results suggest an overall high diagnostic accuracy of the KRAS mutation detection using cfDNA samples. Previous meta-analysis by Xie et al investigated diagnostic accuracy of KRAS mutation detection using ctDNA and the pooled sensitivity, specificity, and DOR were 63.7%, 94.3%, and 37.883, respectively [56]. Our meta-analysis revealed higher sensitivity but lower specificity possibly due to the different diagnostic techniques investigated. Majority of the studies in the study by Xie et al used ARMS or PCR for the detection of KRAS mutation in ctDNA, while our study focused more on digital PCR and NGS which were shown to have higher sensitivity than conventional PCR [58,59].

Since significant inter-study heterogeneity was found during the pooling, we further studied its possible sources. We did not observe significant threshold effect, and meta-regression analysis also indicated no association between inter-study heterogeneity and the 5 covariates in our study (technique used for cfDNA samples, technique used for tissue samples, region, race, and type of CRC). We then performed subgroup analysis. After separating and pooling of the results between primary and metastatic CRC patients, KRAS mutation detection using cfDNA samples showed higher sensitivity (79%), specificity (88%), DOR (29.17), and AUC of SROC curve (0.9045) in mCRC cases, compared to primary CRC patients (57%, 73%, 10.81, and 0.7304, respectively), indicating that this method might be more suitable for mCRC patients. Among the three testing platforms involved in this meta-analysis, ARMS was excluded from the subgroup analysis because of the limited number of study. Comparison between NGS and digital PCR showed higher sensitivity (81%), DOR (29.18), and AUC of SROC curve (0.9067) in digital PCR compared to NGS (65%, 14.61, and 0.8574), which indicates higher accuracy of digital PCR. Further analysis on the subtypes of the techniques showed that Guardant360 NGS panel had only slightly higher sensitivity, specificity, and DOR compared to overall accuracy of NGS (Table 3). In subtypes of digital PCR, BEAMing showed higher sensitivity (87%), specificity (90%), and DOR (50.96) compared to digital droplet PCR. Even after we limited the region of the studies in Europe, BEAMing also showed better accuracy than digital droplet PCR. Those results indicate that BEAMing is a preferable technique for KRAS mutation detection using liquid biopsy samples of CRC patients. Comparison between different regions of the studies showed highest sensitivity (80%) and DOR (32.97) in Europe, compared to Asia and America. Further analysis showed that the accuracy was similar across the three regions when the same type of technique was used, indicating that those differences in accuracy among different regions could be partially due to the different techniques used. Digital PCR showed higher accuracy compared to NGS (Table 3), which may have led to the overall higher accuracy in Europe where digital PCR is more used in the studies. In addition, the difference in diagnostic accuracy between subgroups might partially explain the heterogeneity among the studies. Publication bias was also investigated using Deek’s funnel plot asymmetry test, and results showed no significant publication bias.

Other than the overall or subgroup analyses on the accuracy of KRAS mutation detection using liquid biopsy samples as mentioned above, we did observe a wide range of disparities in accuracy among the studies involved in this systemic review. Even in studies using the same methods for plasma (Guardant360) and tissue (FoundationOne) samples, the sensitivity and specificity varied greatly (59.5% − 75.9% and 87.2% − 97.8%, respectively) [19,35,36]. Similar disparities were also observed in studies using commercial BEAMing kit (OncoBEAM RAS CRC Kit), although different types of methods were used for tissue samples in those studies. Considering that the commercial NGS panels (Guardant360 and FoundationOne) and OncoBEAM RAS CRC Kit should be well standardized and optimized and all the patient populations in the studies were from the same region (America for Guardant360, or Europe for OncoBEAM RAS CRC Kit), the possible sources of those disparities might be from small size of the patient cohort (61–76 patients/study for Guardant360 and 6–236 patients/study for OncoBEAM RAS CRC Kit), or differences in how the experiment and data analysis were performed. Although the exact sources are still unknown, those disparities indicate an urgent need in further standardization and optimization of those techniques. On the other hand, the concordance rate of the studies was also not satisfactory. The concordance rate ranged from 60% [29,53] to 97.5% [14], and nearly 40% (13/33) of the studies showed a concordance rate lower than 80%. Those results indicate risks of misdiagnosis using liquid biopsy to detect KRAS mutation in CRC patients, and KRAS testing results from liquid biopsy samples have to be handled carefully and only be used when tissue samples are not available. Standardization and further optimization of the techniques are needed to hopefully increase the accuracy of the KRAS mutation testing using liquid biopsy samples.

Due to its better availability, quick results turnarounds, and minimal-invasiveness, liquid biopsy has been extensively studied for its use in early detection of cancer, prediction of patient prognosis, and monitoring of disease [15]. Several of the studies involved in this systemic review also performed serial monitoring of ctDNA in colorectal cancer patients. Kim et al [32] collected serial plasma samples of two CRC cancer patients during treatment of cetuximab and observed newly-emerged KRAS mutations in ctDNA results 1.5 months before radiologic progression. Choi et al [36] monitored ctDNA in serial blood samples from CRC patients on anti-EGFR therapy using Guardant360 NGS panel, and observed multiple emerging genetic alterations associated with treatment resistance. Sun et al [39] used a customized 85-gene NGS panel to monitor ctDNA of early-stage CRC patients for 6 months following surgery, and observed decrease in driver mutation in half of the patients after surgery and increase of TP53 and PIK3CA mutations in a patient with liver metastasis. Vidal et al [45] used OncoBEAM RAS CRC Kit to monitor RAS mutations in blood samples from mCRC patients during their anti-EGFR treatment, and found that RAS mutations in ctDNA mirrored the response to treatment. In addition, several on-going prospective clinical trials are also investigating the use of cfDNA in predicting treatment or relapse in earlier stage (I, II or III) or resectable CRC (e.g. NCT04068103, NCT04486378, NCT04264702, NCT04050345). Although many encouraging results were shown in those studies, solid evidence from prospective randomized clinical trials is required before the complete adoption of liquid biopsy in clinical practice [15,60]. From the results of this systemic review and meta-analysis, we also observed wide disparities among the accuracy of KRAS mutation detection using liquid biopsy, even in well standardized and optimized commercial kits (as described above). In addition, pooled accuracy for KRAS mutation detection using liquid biopsy is suboptimal in primary CRC patients (Table 3), indicating significant risk of misdiagnosis. Oncologists should still be very cautious when using liquid biopsy results to guide clinical practices, before evidence from clinical trials proves excellent accuracy of KRAS mutation detection using liquid biopsy, or clear benefit in patient survival in ctDNA/liquid biopsy-guided targeted therapies.

Conclusions

In all, our study showed that NGS, digital PCR, and ARMS techniques had overall high accuracy in detecting KRAS mutation in liquid biopsy samples. The results could be used to guide anti-EGFR therapy in CRC patients with no available tumor tissue samples, but need to be handled carefully considering the potential risk of discordance and misdiagnosis. KRAS mutation detection in liquid biopsy samples had higher accuracy in mCRC patients compared to primary CRC patients, and is therefore more recommended in mCRC patients. Due to its better availability, liquid biopsy could be helpful in early detection and monitoring of CRC, and prediction of patient prognosis, and many studies and clinical trials are investigating its possible roles in those applications. However, oncologists should still be very cautious when using liquid biopsy result in guiding clinical practices, before solid evidence from prospective randomized clinical trials proves its usefulness. Digital PCR also showed higher accuracy than NGS, and among their subtypes, BEAMing showed the highest accuracy, and is recommended for KRAS mutation detection in liquid biopsy samples. Limitation of the study may include that number of studies involved in some subgroups (e.g. primary CRC group) is still quite small, and the results should be handled carefully. In addition, although the accuracy of difference techniques does not differ much when analyzing the highly abundant tumor-derived DNA in tissue samples, different techniques used in the reference group (tumor tissue samples) may still cause potential bias. Other potential variations between the studies (e.g. different patient cohorts, different supplier of experimental reagents, and etc.) may also cause bias to the results. Large prospective randomized clinical trials are needed to further convince the accuracy and usefulness of KRAS mutation detection using cfDNA/liquid biopsy samples in clinical practices.

Supporting information

S1 Table. Search strategy.

(DOCX)

S1 File. PRISMA checklist.

(DOC)

S2 File. Data extracted from eligible studies.

Data were also deposited in Systematic Review Data Repository (SRDR): https://srdr.ahrq.gov/projects/1639.

(XLSX)

Data Availability

All data files are available from the Systematic Review Data Repository (SRDR) database (accession number 1639, URL: https://srdr.ahrq.gov/projects/1639).

Funding Statement

This work was supported by National Natural Science Foundation of China (No.: 81160546; http://www.nsfc.gov.cn/english/site_1/index.html) to YW. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

References

  • 1.Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA: a cancer journal for clinicians. 2015;65(2):87–108. 10.3322/caac.21262 [DOI] [PubMed] [Google Scholar]
  • 2.Troiani T, Napolitano S, Della Corte CM, Martini G, Martinelli E, Morgillo F, et al. Therapeutic value of EGFR inhibition in CRC and NSCLC: 15 years of clinical evidence. ESMO open. 2016;1(5):e000088. 10.1136/esmoopen-2016-000088 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Koulis C, Yap R, Engel R, Jarde T, Wilkins S, Solon G, et al. Personalized Medicine-Current and Emerging Predictive and Prognostic Biomarkers in Colorectal Cancer. Cancers. 2020;12(4). 10.3390/cancers12040812 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Sobrero AF, Maurel J, Fehrenbacher L, Scheithauer W, Abubakr YA, Lutz MP, et al. EPIC: phase III trial of cetuximab plus irinotecan after fluoropyrimidine and oxaliplatin failure in patients with metastatic colorectal cancer. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2008;26(14):2311–9. 10.1200/JCO.2007.13.1193 [DOI] [PubMed] [Google Scholar]
  • 5.Seymour MT, Brown SR, Middleton G, Maughan T, Richman S, Gwyther S, et al. Panitumumab and irinotecan versus irinotecan alone for patients with KRAS wild-type, fluorouracil-resistant advanced colorectal cancer (PICCOLO): a prospectively stratified randomised trial. The Lancet Oncology. 2013;14(8):749–59. 10.1016/S1470-2045(13)70163-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Peeters M, Price TJ, Cervantes A, Sobrero AF, Ducreux M, Hotko Y, et al. Randomized phase III study of panitumumab with fluorouracil, leucovorin, and irinotecan (FOLFIRI) compared with FOLFIRI alone as second-line treatment in patients with metastatic colorectal cancer. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2010;28(31):4706–13. [DOI] [PubMed] [Google Scholar]
  • 7.Van Cutsem E, Kohne CH, Lang I, Folprecht G, Nowacki MP, Cascinu S, et al. Cetuximab plus irinotecan, fluorouracil, and leucovorin as first-line treatment for metastatic colorectal cancer: updated analysis of overall survival according to tumor KRAS and BRAF mutation status. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2011;29(15):2011–9. [DOI] [PubMed] [Google Scholar]
  • 8.Meriggi F, Vermi W, Bertocchi P, Zaniboni A. The Emerging Role of NRAS Mutations in Colorectal Cancer Patients Selected for Anti-EGFR Therapies. Reviews on recent clinical trials. 2014;9(1):8–12. 10.2174/1568026614666140423121525 [DOI] [PubMed] [Google Scholar]
  • 9.Douillard JY, Oliner KS, Siena S, Tabernero J, Burkes R, Barugel M, et al. Panitumumab-FOLFOX4 treatment and RAS mutations in colorectal cancer. The New England journal of medicine. 2013;369(11):1023–34. 10.1056/NEJMoa1305275 [DOI] [PubMed] [Google Scholar]
  • 10.Van Cutsem E, Lenz HJ, Kohne CH, Heinemann V, Tejpar S, Melezinek I, et al. Fluorouracil, leucovorin, and irinotecan plus cetuximab treatment and RAS mutations in colorectal cancer. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2015;33(7):692–700. [DOI] [PubMed] [Google Scholar]
  • 11.Van Cutsem E, Cervantes A, Adam R, Sobrero A, Van Krieken JH, Aderka D, et al. ESMO consensus guidelines for the management of patients with metastatic colorectal cancer. Annals of oncology: official journal of the European Society for Medical Oncology. 2016;27(8):1386–422. 10.1093/annonc/mdw235 [DOI] [PubMed] [Google Scholar]
  • 12.Taly V, Pekin D, Benhaim L, Kotsopoulos SK, Le Corre D, Li X, et al. Multiplex picodroplet digital PCR to detect KRAS mutations in circulating DNA from the plasma of colorectal cancer patients. Clinical chemistry. 2013;59(12):1722–31. 10.1373/clinchem.2013.206359 [DOI] [PubMed] [Google Scholar]
  • 13.Kang JK, Heo S, Kim HP, Song SH, Yun H, Han SW, et al. Liquid biopsy-based tumor profiling for metastatic colorectal cancer patients with ultra-deep targeted sequencing. PloS one. 2020;15(5):e0232754. 10.1371/journal.pone.0232754 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Kitagawa Y, Okumura K, Watanabe T, Tsukamoto K, Kitano S, Nankinzan R, et al. Enrichment technique to allow early detection and monitor emergence of KRAS mutation in response to treatment. Scientific reports. 2019;9(1):11346. 10.1038/s41598-019-47700-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Harle A. Cell-Free DNA in the Management of Colorectal Cancer. Recent results in cancer research Fortschritte der Krebsforschung Progres dans les recherches sur le cancer. 2020;215:253–61. 10.1007/978-3-030-26439-0_13 [DOI] [PubMed] [Google Scholar]
  • 16.Mader S, Pantel K. Liquid Biopsy: Current Status and Future Perspectives. Oncology research and treatment. 2017;40(7–8):404–8. 10.1159/000478018 [DOI] [PubMed] [Google Scholar]
  • 17.Normanno N, Cervantes A, Ciardiello F, De Luca A, Pinto C. The liquid biopsy in the management of colorectal cancer patients: Current applications and future scenarios. Cancer treatment reviews. 2018;70:1–8. 10.1016/j.ctrv.2018.07.007 [DOI] [PubMed] [Google Scholar]
  • 18.Liebs S, Keilholz U, Kehler I, Schweiger C, Hayback J, Nonnenmacher A. Detection of mutations in circulating cell-free DNA in relation to disease stage in colorectal cancer. Cancer medicine. 2019;8(8):3761–9. 10.1002/cam4.2219 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Kato S, Schwaederle MC, Fanta PT, Okamura R, Leichman L, Lippman SM, et al. Genomic Assessment of Blood-Derived Circulating Tumor DNA in Patients With Colorectal Cancers: Correlation With Tissue Sequencing, Therapeutic Response, and Survival. JCO precision oncology. 2019;3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Spindler KL, Pallisgaard N, Andersen RF, Brandslund I, Jakobsen A. Circulating free DNA as biomarker and source for mutation detection in metastatic colorectal cancer. PloS one. 2015;10(4):e0108247. 10.1371/journal.pone.0108247 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Gu J, Zang W, Liu B, Li L, Huang L, Li S, et al. Evaluation of digital PCR for detecting low-level EGFR mutations in advanced lung adenocarcinoma patients: a cross-platform comparison study. Oncotarget. 2017;8(40):67810–20. 10.18632/oncotarget.18866 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Lin MT, Mosier SL, Thiess M, Beierl KF, Debeljak M, Tseng LH, et al. Clinical validation of KRAS, BRAF, and EGFR mutation detection using next-generation sequencing. American journal of clinical pathology. 2014;141(6):856–66. 10.1309/AJCPMWGWGO34EGOD [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Basu AS. Digital Assays Part I: Partitioning Statistics and Digital PCR. SLAS technology. 2017;22(4):369–86. 10.1177/2472630317705680 [DOI] [PubMed] [Google Scholar]
  • 24.Dong L, Wang S, Fu B, Wang J. Evaluation of droplet digital PCR and next generation sequencing for characterizing DNA reference material for KRAS mutation detection. Scientific reports. 2018;8(1):9650. 10.1038/s41598-018-27368-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Linardou H, Briasoulis E, Dahabreh IJ, Mountzios G, Papadimitriou C, Papadopoulos S, et al. All about KRAS for clinical oncology practice: gene profile, clinical implications and laboratory recommendations for somatic mutational testing in colorectal cancer. Cancer treatment reviews. 2011;37(3):221–33. 10.1016/j.ctrv.2010.07.008 [DOI] [PubMed] [Google Scholar]
  • 26.Ye P, Cai P, Xie J, Wei Y. The diagnostic accuracy of digital PCR, ARMS and NGS for detecting KRAS mutation in cell-free DNA of patients with colorectal cancer: A protocol for systematic review and meta-analysis. Medicine. 2020;99(26):e20708. 10.1097/MD.0000000000020708 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.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. Annals of internal medicine. 2011;155(8):529–36. 10.7326/0003-4819-155-8-201110180-00009 [DOI] [PubMed] [Google Scholar]
  • 28.Zamora J, Abraira V, Muriel A, Khan K, Coomarasamy A. Meta-DiSc: a software for meta-analysis of test accuracy data. BMC medical research methodology. 2006;6:31. 10.1186/1471-2288-6-31 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Chang YS, Fang HY, Hung YC, Ke TW, Chang CM, Liu TY, et al. Correlation of genomic alterations between tumor tissue and circulating tumor DNA by next-generation sequencing. Journal of cancer research and clinical oncology. 2018;144(11):2167–75. 10.1007/s00432-018-2747-9 [DOI] [PubMed] [Google Scholar]
  • 30.Garcia J, Forestier J, Dusserre E, Wozny AS, Geiguer F, Merle P, et al. Cross-platform comparison for the detection of RAS mutations in cfDNA (ddPCR Biorad detection assay, BEAMing assay, and NGS strategy). Oncotarget. 2018;9(30):21122–31. 10.18632/oncotarget.24950 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Wang L, Wang M, Zhao C, Wang W, Li Y. Detection of RAS/BRAF mutations in the ctDNA of mCRC patients by NGS technique. Anti-tumor Pharmacy. 2017;7(3):296–308. [Google Scholar]
  • 32.Kim ST, Lee WS, Lanman RB, Mortimer S, Zill OA, Kim KM, et al. Prospective blinded study of somatic mutation detection in cell-free DNA utilizing a targeted 54-gene next generation sequencing panel in metastatic solid tumor patients. Oncotarget. 2015;6(37):40360–9. 10.18632/oncotarget.5465 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Vessies DCL, Greuter MJE, van Rooijen KL, Linders TC, Lanfermeijer M, Ramkisoensing KL, et al. Performance of four platforms for KRAS mutation detection in plasma cell-free DNA: ddPCR, Idylla, COBAS z480 and BEAMing. Scientific reports. 2020;10(1):8122. 10.1038/s41598-020-64822-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Cao H, Liu X, Chen Y, Yang P, Huang T, Song L, et al. Circulating Tumor DNA Is Capable of Monitoring the Therapeutic Response and Resistance in Advanced Colorectal Cancer Patients Undergoing Combined Target and Chemotherapy. Frontiers in oncology. 2020;10:466. 10.3389/fonc.2020.00466 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Gupta R, Othman T, Chen C, Sandhu J, Ouyang C, Fakih M. Guardant360 Circulating Tumor DNA Assay Is Concordant with FoundationOne Next-Generation Sequencing in Detecting Actionable Driver Mutations in Anti-EGFR Naive Metastatic Colorectal Cancer. The oncologist. 2020;25(3):235–43. 10.1634/theoncologist.2019-0441 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Choi IS, Kato S, Fanta PT, Leichman L, Okamura R, Raymond VM, et al. Genomic Profiling of Blood-Derived Circulating Tumor DNA from Patients with Colorectal Cancer: Implications for Response and Resistance to Targeted Therapeutics. Molecular cancer therapeutics. 2019;18(10):1852–62. 10.1158/1535-7163.MCT-18-0965 [DOI] [PubMed] [Google Scholar]
  • 37.Osumi H, Shinozaki E, Takeda Y, Wakatsuki T, Ichimura T, Saiura A, et al. Clinical relevance of circulating tumor DNA assessed through deep sequencing in patients with metastatic colorectal cancer. Cancer medicine. 2019;8(1):408–17. 10.1002/cam4.1913 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Yao J, Zang W, Ge Y, Weygant N, Yu P, Li L, et al. RAS/BRAF Circulating Tumor DNA Mutations as a Predictor of Response to First-Line Chemotherapy in Metastatic Colorectal Cancer Patients. Canadian journal of gastroenterology & hepatology. 2018;2018:4248971. 10.1155/2018/4248971 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Sun X, Huang T, Cheng F, Huang K, Liu M, He W, et al. Monitoring colorectal cancer following surgery using plasma circulating tumor DNA. Oncology letters. 2018;15(4):4365–75. 10.3892/ol.2018.7837 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Normanno N, Esposito Abate R, Lambiase M, Forgione L, Cardone C, Iannaccone A, et al. RAS testing of liquid biopsy correlates with the outcome of metastatic colorectal cancer patients treated with first-line FOLFIRI plus cetuximab in the CAPRI-GOIM trial. Annals of oncology: official journal of the European Society for Medical Oncology. 2018;29(1):112–8. 10.1093/annonc/mdx417 [DOI] [PubMed] [Google Scholar]
  • 41.Garcia-Foncillas J, Tabernero J, Elez E, Aranda E, Benavides M, Camps C, et al. Prospective multicenter real-world RAS mutation comparison between OncoBEAM-based liquid biopsy and tissue analysis in metastatic colorectal cancer. British journal of cancer. 2018;119(12):1464–70. 10.1038/s41416-018-0293-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Beije N, Helmijr JC, Weerts MJA, Beaufort CM, Wiggin M, Marziali A, et al. Somatic mutation detection using various targeted detection assays in paired samples of circulating tumor DNA, primary tumor and metastases from patients undergoing resection of colorectal liver metastases. Molecular oncology. 2016;10(10):1575–84. 10.1016/j.molonc.2016.10.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Grasselli J, Elez E, Caratu G, Matito J, Santos C, Macarulla T, et al. Concordance of blood- and tumor-based detection of RAS mutations to guide anti-EGFR therapy in metastatic colorectal cancer. Annals of oncology: official journal of the European Society for Medical Oncology. 2017;28(6):1294–301. 10.1093/annonc/mdx112 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Schmiegel W, Scott RJ, Dooley S, Lewis W, Meldrum CJ, Pockney P, et al. Blood-based detection of RAS mutations to guide anti-EGFR therapy in colorectal cancer patients: concordance of results from circulating tumor DNA and tissue-based RAS testing. Molecular oncology. 2017;11(2):208–19. 10.1002/1878-0261.12023 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Vidal J, Muinelo L, Dalmases A, Jones F, Edelstein D, Iglesias M, et al. Plasma ctDNA RAS mutation analysis for the diagnosis and treatment monitoring of metastatic colorectal cancer patients. Annals of oncology: official journal of the European Society for Medical Oncology. 2017;28(6):1325–32. 10.1093/annonc/mdx125 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Rachiglio AM, Esposito Abate R, Sacco A, Pasquale R, Fenizia F, Lambiase M, et al. Limits and potential of targeted sequencing analysis of liquid biopsy in patients with lung and colon carcinoma. Oncotarget. 2016;7(41):66595–605. 10.18632/oncotarget.10704 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Beranek M, Sirak I, Vosmik M, Petera J, Drastikova M, Palicka V. Carrier molecules and extraction of circulating tumor DNA for next generation sequencing in colorectal cancer. Acta Medica (Hradec Kralove). 2016;59(2):54–8. 10.14712/18059694.2016.54 [DOI] [PubMed] [Google Scholar]
  • 48.Bidard FC, Kiavue N, Ychou M, Cabel L, Stern MH, Madic J, et al. Circulating Tumor Cells and Circulating Tumor DNA Detection in Potentially Resectable Metastatic Colorectal Cancer: A Prospective Ancillary Study to the Unicancer Prodige-14 Trial. Cells. 2019;8(6). 10.3390/cells8060516 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Sclafani F, Chau I, Cunningham D, Hahne JC, Vlachogiannis G, Eltahir Z, et al. KRAS and BRAF mutations in circulating tumour DNA from locally advanced rectal cancer. Scientific reports. 2018;8(1):1445. 10.1038/s41598-018-19212-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Kidess-Sigal E, Liu HE, Triboulet MM, Che J, Ramani VC, Visser BC, et al. Enumeration and targeted analysis of KRAS, BRAF and PIK3CA mutations in CTCs captured by a label-free platform: Comparison to ctDNA and tissue in metastatic colorectal cancer. Oncotarget. 2016;7(51):85349–64. 10.18632/oncotarget.13350 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Galbiati S, Damin F, Ferraro L, Soriani N, Burgio V, Ronzoni M, et al. Microarray Approach Combined with ddPCR: An Useful Pipeline for the Detection and Quantification of Circulating Tumour dna Mutations. Cells. 2019;8(8). 10.3390/cells8080769 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Galbiati S, Damin F, Burgio V, Brisci A, Soriani N, Belcastro B, et al. Evaluation of three advanced methodologies, COLD-PCR, microarray and ddPCR, for identifying the mutational status by liquid biopsies in metastatic colorectal cancer patients. Clinica chimica acta; international journal of clinical chemistry. 2019;489:136–43. 10.1016/j.cca.2018.12.004 [DOI] [PubMed] [Google Scholar]
  • 53.Takayama Y, Suzuki K, Muto Y, Ichida K, Fukui T, Kakizawa N, et al. Monitoring circulating tumor DNA revealed dynamic changes in KRAS status in patients with metastatic colorectal cancer. Oncotarget. 2018;9(36):24398–413. 10.18632/oncotarget.25309 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Sefrioui D, Mauger F, Leclere L, Beaussire L, Di Fiore F, Deleuze JF, et al. Comparison of the quantification of KRAS mutations by digital PCR and E-ice-COLD-PCR in circulating-cell-free DNA from metastatic colorectal cancer patients. Clinica chimica acta; international journal of clinical chemistry. 2017;465:1–4. 10.1016/j.cca.2016.12.004 [DOI] [PubMed] [Google Scholar]
  • 55.Yamada T, Iwai T, Takahashi G, Kan H, Koizumi M, Matsuda A, et al. Utility of KRAS mutation detection using circulating cell-free DNA from patients with colorectal cancer. Cancer science. 2016;107(7):936–43. 10.1111/cas.12959 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Xie W, Xie L, Song X. The diagnostic accuracy of circulating free DNA for the detection of KRAS mutation status in colorectal cancer: A meta-analysis. Cancer medicine. 2019;8(3):1218–31. 10.1002/cam4.1989 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Glas AS, Lijmer JG, Prins MH, Bonsel GJ, Bossuyt PM. The diagnostic odds ratio: a single indicator of test performance. Journal of clinical epidemiology. 2003;56(11):1129–35. 10.1016/s0895-4356(03)00177-x [DOI] [PubMed] [Google Scholar]
  • 58.Zhang BO, Xu CW, Shao Y, Wang HT, Wu YF, Song YY, et al. Comparison of droplet digital PCR and conventional quantitative PCR for measuring EGFR gene mutation. Experimental and therapeutic medicine. 2015;9(4):1383–8. 10.3892/etm.2015.2221 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Gao J, Wu H, Wang L, Zhang H, Duan H, Lu J, et al. Validation of targeted next-generation sequencing for RAS mutation detection in FFPE colorectal cancer tissues: comparison with Sanger sequencing and ARMS-Scorpion real-time PCR. BMJ open. 2016;6(1):e009532. 10.1136/bmjopen-2015-009532 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Poulet G, Massias J, Taly V. Liquid Biopsy: General Concepts. Acta cytologica. 2019;63(6):449–55. 10.1159/000499337 [DOI] [PubMed] [Google Scholar]

Decision Letter 0

Anthony F Shields

8 Jan 2021

PONE-D-20-31082

The diagnostic accuracy of digital PCR, ARMS and NGS for detecting KRAS mutation in cell-free DNA of patients with colorectal cancer: a systematic review and meta-analysis

PLOS ONE

Dear Dr. Ye,

Thank you for submitting your manuscript to PLOS ONE. After careful consideration, we feel that it has merit but does not fully meet PLOS ONE’s publication criteria as it currently stands. Therefore, we invite you to submit a revised version of the manuscript that addresses the points raised during the review process.

As noted by the reviewer, the authors should better describe the assay systems used and the differences in technical approaches.  They should also include the full names, not just the abbreviations such as for ARMS (Amplification Refractory Mutation System) and what assay versions were used, such as the new SuperARMS.  While some of the studies they discussed may have used KRAS alone, others included NRAS and this should be noted in the review. Finally, as suggested the article is written in English, but this is not the authors’ first language and a through review of the grammar is needed.

Please submit your revised manuscript by Feb 13 2021 11:59PM. If you will need more time than this to complete your revisions, please reply to this message or contact the journal office at plosone@plos.org. When you're ready to submit your revision, log on to https://www.editorialmanager.com/pone/ and select the 'Submissions Needing Revision' folder to locate your manuscript file.

Please include the following items when submitting your revised manuscript:

  • A rebuttal letter that responds to each point raised by the academic editor and reviewer(s). You should upload this letter as a separate file labeled 'Response to Reviewers'.

  • A marked-up copy of your manuscript that highlights changes made to the original version. You should upload this as a separate file labeled 'Revised Manuscript with Track Changes'.

  • An unmarked version of your revised paper without tracked changes. You should upload this as a separate file labeled 'Manuscript'.

If you would like to make changes to your financial disclosure, please include your updated statement in your cover letter. Guidelines for resubmitting your figure files are available below the reviewer comments at the end of this letter.

If applicable, we recommend that you deposit your laboratory protocols in protocols.io to enhance the reproducibility of your results. Protocols.io assigns your protocol its own identifier (DOI) so that it can be cited independently in the future. For instructions see: http://journals.plos.org/plosone/s/submission-guidelines#loc-laboratory-protocols

We look forward to receiving your revised manuscript.

Kind regards,

Anthony F. Shields, M.D., Ph.D.

Academic Editor

PLOS ONE

Journal Requirements:

When submitting your revision, we need you to address these additional requirements.

1.) Please ensure that your manuscript meets PLOS ONE's style requirements, including those for file naming. The PLOS ONE style templates can be found at

https://journals.plos.org/plosone/s/file?id=wjVg/PLOSOne_formatting_sample_main_body.pdf and

https://journals.plos.org/plosone/s/file?id=ba62/PLOSOne_formatting_sample_title_authors_affiliations.pdf

2.) Thank you for including the statement that 'Literature research was performed independently by PY and PC in June 2020'. Please revise this statement to clarify whether all databases were searched from inception, or if there were any limits placed on the publication dates in your search.

3.) At this time, we ask that you please provide the full search strategy and search terms for at least one database used as Supplementary Information.

4.) In the Methods section, please provide the methodology used for data extraction, including the specific reporting items that were extracted from the included studies.

[Note: HTML markup is below. Please do not edit.]

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #1: Partly

Reviewer #2: Yes

**********

2. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #1: I Don't Know

Reviewer #2: Yes

**********

3. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #1: Yes

Reviewer #2: Yes

**********

4. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #1: Yes

Reviewer #2: No

**********

5. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #1: The investigators performed literature review to identify studies that have examined KRAS status using cell free DNA. They performed meta-analysis of these data and present the results here.

The cited references appear to be during or after 2016, shortly after a time when expansion of standard tissue-based platforms adapted to the concept of All-RAS sequencing (more isoforms and testing of NRAS as well). Please clarify confirmation that the papers using meta-analysis adhered to expanded testing, and also clarify whether NRAS (while much less prevalent, 5-10% in CRC) was also assessed in any or all the studies. Also, it would be helpful to clarify whether there were notable differences in testing approach in relation to accuracy by region/country of testing, and whether any potential differences could be accounted for by significant differences in testing approaches.

One of the proposed advantages of cell free DNA is more accurate identification of tumor cell heterogeneity, including potential mixed populations of KRAS mutant and KRAS wild type cells. Not all the studies divided examined here, reported what percentage of samples yielded positive results for both within the same specimens.

The authors pool results across many different studies, using many different approaches, and the final pulled result does not mean much considering the wide range. For presentation of statistical analysis, it would be preferable to emphasize more the wide range and median values, with a subset analyses to report common themes and approaches that appear to be reproducible and precise (and also hopefully providing accuracy ). There are many disparate results reported by a number of these papers, including a wide range of sensitivity of assays, even when using the same method (eg NGS), and differences between cell free DNA-based assessment versus tissue-based assessment. The latter point is well documented, and more in-depth comments about the wide range of disparities should be included in the discussion section. Concordance rates are poor, yet many oncologists worldwide depend heavily on liquid-based biopsy at assessment for determination of using EGFR inhibitor treatments. What is the authors’ perspective on how this practice should change, and how, based on the results of this meta-analysis? I encourage the investigators to leverage this manuscript as a platform to propose change in the field for better and more accurate use of these tests.

The use of serial assessment of KRAS and other markers over time in colorectal cancer has been promoted without strong evidence that this changes management on a large scale, nor results in improvement of overall survival. Can the authors comment on perspective on this angle, noting also that there are multiple prospective trials examining the use of cell free DNA in general in the postoperative setting for earlier stage/resectable colorectal cancers (in the U.S.).

On page 18, it is mentioned that some studies assessing KRAS were performed in patients with primary colorectal cancer; please clarify why this was done in the studies, as this type of assessment is not routinely indicated in nonmetastatic patients. As the data are there, it would also be helpful to discuss any notable differences in sensitivity, and/or concordance, between primary and metastatic tumors that were assessed in relation to cell free DNA, and early-stage versus later stage patients.

Reviewer #2: In this review/meta-analysis, Peng Ye tried to compare different methods used in detecting KRAS mutations in plasma of CRC patients. Although the authors put a lot of efforts into, there remain some issues/changes to be done:

1.) Improve English! There are several grammer errors!

2.) In this study, the authors are comparing PCR, NGS and ARMS. However, they are describing these methods only superficial. Thus, in the introduction section the authors should describe these techniques (what is better in one to another method? costs issues? ecc...).

3.) The authors are describing that only KRAS is important in the selection for anti-EGFR treatment. This is not correct! The authors should focus, or at least discuss, that next to KRAS mutations also NRAS mutations are very important to select the best treatment for our CRC patients.

4.) The authors are concluding that liquid biopsy may be only important in the metastatic stage, however, also in limited stages liquid biopys may be helpful in monitoring disease or detecting early relapse. Please, discuss this fact.

**********

6. PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy.

Reviewer #1: No

Reviewer #2: No

[NOTE: If reviewer comments were submitted as an attachment file, they will be attached to this email and accessible via the submission site. Please log into your account, locate the manuscript record, and check for the action link "View Attachments". If this link does not appear, there are no attachment files.]

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool, https://pacev2.apexcovantage.com/. PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Registration is free. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email PLOS at figures@plos.org. Please note that Supporting Information files do not need this step.

PLoS One. 2021 Mar 26;16(3):e0248775. doi: 10.1371/journal.pone.0248775.r002

Author response to Decision Letter 0


1 Feb 2021

Comments from Editor:

1) As noted by the reviewer, the authors should better describe the assay systems used and the differences in technical approaches.

Response: Many thanks for the suggestions. We have revised the manuscript and included more descriptions on the three assay systems and their advantages and disadvantages. Please find the revision in the 3rd paragraph of the Introduction section (page 5 of the Revised Manuscript with Track Changes).

2) They should also include the full names, not just the abbreviations such as for ARMS (Amplification Refractory Mutation System) and what assay versions were used, such as the new SuperARMS.

Response: Many thanks for the suggestions. We have revised the manuscript and included the detailed type/version of techniques used in the studies. Please find the revision in Table 1 on page 10-17 in the Revised Manuscript with Track Changes. In addition, we also revised the names of techniques and avoided usage of abbreviations. Please find the revisions in the Revised Manuscript with Track Changes.

3) While some of the studies they discussed may have used KRAS alone, others included NRAS and this should be noted in the review.

Response: Thanks very much for the suggestion. In the 33 eligible studies, there were in all 20 studies which targeted All-RAS, or expanded isoforms of KRAS. Most of those studies reported KRAS sequencing results separately from NRAS/HRAS, while 6 studies reported only All-RAS results. In this case, the NRAS testing results were included in the systematic review and meta-analysis. We have revised the manuscript accordingly. Please find the revisions in the 1st paragraph of “Review of eligible publications” section (page 9 of the Revised Manuscript with Track Changes).

4) Finally, as suggested the article is written in English, but this is not the authors’ first language and a through review of the grammar is needed.

Response: Very sorry for the grammar errors. We have carefully proofread the manuscript and tried our best to remove the grammar errors. Please find the revisions in the Revised Manuscript with Track Changes.

Additional requirements:

1.) Please ensure that your manuscript meets PLOS ONE's style requirements, including those for file naming. The PLOS ONE style templates can be found at

https://journals.plos.org/plosone/s/file?id=wjVg/PLOSOne_formatting_sample_main_body.pdf and

https://journals.plos.org/plosone/s/file?id=ba62/PLOSOne_formatting_sample_title_authors_affiliations.pdf

Response: Many thanks for your comments. We have carefully checked and ensured that our manuscript meets PLOS ONE’s style requirement.

2.) Thank you for including the statement that 'Literature research was performed independently by PY and PC in June 2020'. Please revise this statement to clarify whether all databases were searched from inception, or if there were any limits placed on the publication dates in your search.

Response: Thanks very much for your suggestion. We did not place any limitation on the publication dates in the literature search. Please find the revised statement in the 1st paragraph in “Literature searching and selection of publication” section (page 6 of the Revised Manuscript with Track Changes).

3.) At this time, we ask that you please provide the full search strategy and search terms for at least one database used as Supplementary Information.

Response: Thanks very much for your suggestion. We have included the detailed search strategy as S1 Table. Please find it in the supporting information.

4.) In the Methods section, please provide the methodology used for data extraction, including the specific reporting items that were extracted from the included studies.

Response: Thanks very much for your comments. We have revised the manuscript and included all the reporting items used in our data extraction. Please find the revisions in the 2nd paragraph of “Literature searching and selection of publication” section (page 6-8 of the Revised Manuscript with Track Changes).

Comments from Review #1:

The investigators performed literature review to identify studies that have examined KRAS status using cell free DNA. They performed meta-analysis of these data and present the results here.

1) The cited references appear to be during or after 2016, shortly after a time when expansion of standard tissue-based platforms adapted to the concept of All-RAS sequencing (more isoforms and testing of NRAS as well). Please clarify confirmation that the papers using meta-analysis adhered to expanded testing, and also clarify whether NRAS (while much less prevalent, 5-10% in CRC) was also assessed in any or all the studies.

Response: Thanks very much for your suggestions. In our review, 20 eligible studies performed All-RAS sequencing with expanded isoforms of the genes. Most of the studies reported KRAS testing results separately, or ignored NRAS/HRAS testing results. The rest 6 studies reported All-RAS sequencing results only, and in this case, the NRAS testing results were included in the subsequent systematic review and meta-analysis. We also revised the manuscript to describe those details. Please find them in the 1st paragraph of “Review of eligible publications” section (page 9 of Revised Manuscript with Track Changes).

2) Also, it would be helpful to clarify whether there were notable differences in testing approach in relation to accuracy by region/country of testing, and whether any potential differences could be accounted for by significant differences in testing approaches.

Response: Thanks very much for the helpful suggestions. We performed meta-analysis across different regions in studies using next-generation sequencing or digital polymerase chain reaction, and the sensitivity was similar across different regions for next-generation sequencing, and sensitivity and diagnostic odds ratio were similar between Europe and Asia, indicating that the differences in accuracy among different regions could be partially explained by significant difference in techniques used in those regions. We also inserted those results and discussions in the 5th paragraph of “Meta-analysis of the accuracy of KRAS mutation detection using cfDNA samples” section (page 29-30 of the Revised Manuscript with Track Changes).

3) One of the proposed advantages of cell free DNA is more accurate identification of tumor cell heterogeneity, including potential mixed populations of KRAS mutant and KRAS wild type cells. Not all the studies divided examined here, reported what percentage of samples yielded positive results for both within the same specimens.

Response: Thanks very much for the comments. We agree that the tumor heterogeneity is not a major focus of this meta-analysis or the references of this manuscript. The expression was removed accordingly. Please find the revision in the 2nd paragraph of Introduction section (page 5 of the Revised Manuscript with Track Changes).

4) The authors pool results across many different studies, using many different approaches, and the final pulled result does not mean much considering the wide range. For presentation of statistical analysis, it would be preferable to emphasize more the wide range and median values, with a subset analyses to report common themes and approaches that appear to be reproducible and precise (and also hopefully providing accuracy ).

Response: Thanks very much for your suggestions. We have put more effort on the subgroup analysis. In addition to the comparison among different regions in studies using next-generation sequencing or digital polymerase chain reaction, we also investigated the accuracy of subtypes of next-generation sequencing (Guardant360 panel) and digital polymerase chain reaction (Beads, Emulsion, Amplification and Magnetics, and digital droplet polymerase chain reaction). In addition, we emphasized more on the heterogeneity among the studies and also addressed that we focus more on the possible sources of heterogeneity and subgroup analysis. Please find the revisions in the 2nd and 4th paragraph of “Meta-analysis of the accuracy of KRAS mutation detection using cfDNA samples” section (page 25, 29 of the Revised Manuscript with Track Changes), and Table 3 (page 26-28 of the Revised Manuscript with Track Changes).

5) There are many disparate results reported by a number of these papers, including a wide range of sensitivity of assays, even when using the same method (eg NGS), and differences between cell free DNA-based assessment versus tissue-based assessment. The latter point is well documented, and more in-depth comments about the wide range of disparities should be included in the discussion section.

Response: Thanks very much for your helpful suggestions. We have looked into the wide range of disparities among the studies, and inserted comments in the discussion section. Please find the revision in the 4th paragraph of Discussion section (page 34 of the Revised Manuscript with Track Changes).

6) Concordance rates are poor, yet many oncologists worldwide depend heavily on liquid-based biopsy at assessment for determination of using EGFR inhibitor treatments. What is the authors’ perspective on how this practice should change, and how, based on the results of this meta-analysis? I encourage the investigators to leverage this manuscript as a platform to propose change in the field for better and more accurate use of these tests.

Response: Thanks very much for your helpful suggestions. Accordingly, we also commented on the poor concordance rate and proposed possible ways to limit the risk of misdiagnosis. Please find the comments in the 4th paragraph of Discussion section, after the comments for wide range of disparities among the studies (page 34-35 of the Revised Manuscript with Track Changes).

7) The use of serial assessment of KRAS and other markers over time in colorectal cancer has been promoted without strong evidence that this changes management on a large scale, nor results in improvement of overall survival. Can the authors comment on perspective on this angle, noting also that there are multiple prospective trials examining the use of cell free DNA in general in the postoperative setting for earlier stage/resectable colorectal cancers (in the U.S.).

Response: Thanks very much for your helpful suggestions. We have looked into the serial assessment of KRAS using liquid biopsy samples. Please find our comments in the 5th paragraph of Discussion section in the revised manuscript (page 35-36 of the Revised Manuscript with Track Changes).

8) On page 18, it is mentioned that some studies assessing KRAS were performed in patients with primary colorectal cancer; please clarify why this was done in the studies, as this type of assessment is not routinely indicated in nonmetastatic patients.

Response: Thanks very much for your suggestions. We have looked into the reasons for the involvement of nonmetastatic patients in those studies, and inserted them in the start of the 3rd paragraph in “Meta-analysis of the accuracy of KRAS mutation detection using cfDNA samples” in Results section (page 26 of the Revised Manuscript with Track Changes).

9) As the data are there, it would also be helpful to discuss any notable differences in sensitivity, and/or concordance, between primary and metastatic tumors that were assessed in relation to cell free DNA, and early-stage versus later stage patients.

Response: Thanks very much for your suggestions. We compared the accuracy data between primary and metastatic tumors in relation to cell free DNA, and the results could be found in the 3rd paragraph in “Meta-analysis of the accuracy of KRAS mutation detection using cfDNA samples” in Results section (page 26 of the Revised Manuscript with Track Changes), and the 3rd paragraph (page 32 of the Revised Manuscript with Track Changes) and 5th paragraph of Discussion section (line 3-5 on page 36 of the Revised Manuscript with Track Changes). In addition, following your advices, we tried to extract the accuracy data from early-stage CRC patients. But unfortunately, data could only be extracted from 3 studies, including 1 study which lacked true positive cases and was therefore excluded by the statistical software. The number of the rest studies (2 studies) was too small for meta-analysis. Therefore, we did not continue the comparison between early-stage and late-stage patients. Those results were also described at the end of 3rd paragraph in “Meta-analysis of the accuracy of KRAS mutation detection using cfDNA samples” in Results section (page 26 of the Revised Manuscript with Track Changes).

Comments from Review #2:

In this review/meta-analysis, Peng Ye tried to compare different methods used in detecting KRAS mutations in plasma of CRC patients. Although the authors put a lot of efforts into, there remain some issues/changes to be done:

1.) Improve English! There are several grammer errors!

Response: Very sorry for the grammar errors. We have carefully proofread the manuscript and tried our best to remove the grammar errors. Please find the revisions in the Revised Manuscript with Track Changes.

2.) In this study, the authors are comparing PCR, NGS and ARMS. However, they are describing these methods only superficial. Thus, in the introduction section the authors should describe these techniques (what is better in one to another method? costs issues? ecc...).

Response: Thanks very much for the suggestions. We have revised the introduction section and described more advantages and disadvantages of those techniques. Please find the revision in the 3rd paragraph of Introduction section (page 5-6 of the Revised Manuscript with Track Changes).

3.) The authors are describing that only KRAS is important in the selection for anti-EGFR treatment. This is not correct! The authors should focus, or at least discuss, that next to KRAS mutations also NRAS mutations are very important to select the best treatment for our CRC patients.

Response: Thanks very much for your comments. We have revised the introduction section and emphasized the importance of NRAS in the anti-EGFR treatment for CRC patients. Please find the revision in the 1st paragraph of Introduction section (page 4 of the Revised Manuscript with Track Changes).

4.) The authors are concluding that liquid biopsy may be only important in the metastatic stage, however, also in limited stages liquid biopys may be helpful in monitoring disease or detecting early relapse. Please, discuss this fact.

Response: Many thanks for your comments. We discussed more about the use of liquid biopsy in early detection and monitoring of the disease and prediction of patients prognosis. Please find the revision in the 5th paragraph of Discussion section (page 35-36 of the Revised Manuscript with Track Changes), and in Conclusion section (at end of page 36 of the Revised Manuscript with Track Changes).

Attachment

Submitted filename: Response to Reviewers.docx

Decision Letter 1

Anthony F Shields

18 Feb 2021

PONE-D-20-31082R1

The diagnostic accuracy of digital PCR, ARMS and NGS for detecting KRAS mutation in cell-free DNA of patients with colorectal cancer: a systematic review and meta-analysis

PLOS ONE

Dear Dr. Ye,

Thank you for submitting your manuscript to PLOS ONE. After careful consideration, we feel that it has merit but does not fully meet PLOS ONE’s publication criteria as it currently stands. Therefore, we invite you to submit a revised version of the manuscript that addresses the points raised during the review process.

==============================

The authors misinterpreted the editor's suggestion to "include the full names, not just the abbreviations such as for ARMS" etc. Terms such as ARMS, PCR, and NGS need to be defined at the start, but abbreviations can be used after that. Please correct that. Otherwise, the manuscript is much improved and acceptable for publication.

==============================

Please submit your revised manuscript by Apr 04 2021 11:59PM. If you will need more time than this to complete your revisions, please reply to this message or contact the journal office at plosone@plos.org. When you're ready to submit your revision, log on to https://www.editorialmanager.com/pone/ and select the 'Submissions Needing Revision' folder to locate your manuscript file.

Please include the following items when submitting your revised manuscript:

  • A marked-up copy of your manuscript that highlights changes made to the original version. You should upload this as a separate file labeled 'Revised Manuscript with Track Changes'.

  • An unmarked version of your revised paper without tracked changes. You should upload this as a separate file labeled 'Manuscript'.

If you would like to make changes to your financial disclosure, please include your updated statement in your cover letter. Guidelines for resubmitting your figure files are available below the reviewer comments at the end of this letter.

If applicable, we recommend that you deposit your laboratory protocols in protocols.io to enhance the reproducibility of your results. Protocols.io assigns your protocol its own identifier (DOI) so that it can be cited independently in the future. For instructions see: http://journals.plos.org/plosone/s/submission-guidelines#loc-laboratory-protocols

We look forward to receiving your revised manuscript.

Kind regards,

Anthony F. Shields, M.D., Ph.D.

Academic Editor

PLOS ONE

Additional Editor Comments (if provided):

The authors misinterpreted the editor's suggestion to "include the full names, not just the abbreviations such as for ARMS" etc. Terms such as ARMS, PCR, and NGS need to be defined at the start, but abbreviations can be used after that. Please correct that. Otherwise, the manuscript is much improved and acceptable for publication.

[Note: HTML markup is below. Please do not edit.]

[NOTE: If reviewer comments were submitted as an attachment file, they will be attached to this email and accessible via the submission site. Please log into your account, locate the manuscript record, and check for the action link "View Attachments". If this link does not appear, there are no attachment files.]

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool, https://pacev2.apexcovantage.com/. PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Registration is free. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email PLOS at figures@plos.org. Please note that Supporting Information files do not need this step.

PLoS One. 2021 Mar 26;16(3):e0248775. doi: 10.1371/journal.pone.0248775.r004

Author response to Decision Letter 1


18 Feb 2021

Dear Editor,

Thank you very much for your decision letter and advice on our manuscript entitled “The diagnostic accuracy of digital PCR, ARMS and NGS for detecting KRAS mutation in cell-free DNA of patients with colorectal cancer: a systematic review and meta-analysis” (PONE-D-20-31082R1) to be considered for publication in PLOS ONE.

Please see our response to your comments below.

1. The authors misinterpreted the editor's suggestion to "include the full names, not just the abbreviations such as for ARMS" etc. Terms such as ARMS, PCR, and NGS need to be defined at the start, but abbreviations can be used after that. Please correct that. Otherwise, the manuscript is much improved and acceptable for publication.

Response: Very sorry for the misinterpretation and troubles made. Following your suggestions, we have changed the full names of ARMS, PCR, NGS, and BEAMing into abbreviations after defining them at first use. Please find the corrections in the Revised Manuscript with Track Changes.

Look forward to hearing from you soon.

Sincerely yours,

Peng Ye

Chengdu University, Chengdu 610106, P.R.China

Tel: (86)18602885572

E-mail: yepeng@cdu.edu.cn

Decision Letter 2

Anthony F Shields

5 Mar 2021

The diagnostic accuracy of digital PCR, ARMS and NGS for detecting KRAS mutation in cell-free DNA of patients with colorectal cancer: a systematic review and meta-analysis

PONE-D-20-31082R2

Dear Dr. Ye,

We’re pleased to inform you that your manuscript has been judged scientifically suitable for publication and will be formally accepted for publication once it meets all outstanding technical requirements.

Within one week, you’ll receive an e-mail detailing the required amendments. When these have been addressed, you’ll receive a formal acceptance letter and your manuscript will be scheduled for publication.

An invoice for payment will follow shortly after the formal acceptance. To ensure an efficient process, please log into Editorial Manager at http://www.editorialmanager.com/pone/, click the 'Update My Information' link at the top of the page, and double check that your user information is up-to-date. If you have any billing related questions, please contact our Author Billing department directly at authorbilling@plos.org.

If your institution or institutions have a press office, please notify them about your upcoming paper to help maximize its impact. If they’ll be preparing press materials, please inform our press team as soon as possible -- no later than 48 hours after receiving the formal acceptance. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information, please contact onepress@plos.org.

Kind regards,

Anthony F. Shields, M.D., Ph.D.

Academic Editor

PLOS ONE

Acceptance letter

Anthony F Shields

17 Mar 2021

PONE-D-20-31082R2

The diagnostic accuracy of digital PCR, ARMS and NGS for detecting KRAS mutation in cell-free DNA of patients with colorectal cancer: a systematic review and meta-analysis

Dear Dr. Ye:

I'm pleased to inform you that your manuscript has been deemed suitable for publication in PLOS ONE. Congratulations! Your manuscript is now with our production department.

If your institution or institutions have a press office, please let them know about your upcoming paper now to help maximize its impact. If they'll be preparing press materials, please inform our press team within the next 48 hours. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information please contact onepress@plos.org.

If we can help with anything else, please email us at plosone@plos.org.

Thank you for submitting your work to PLOS ONE and supporting open access.

Kind regards,

PLOS ONE Editorial Office Staff

on behalf of

Dr. Anthony F. Shields

Academic Editor

PLOS ONE

Associated Data

    This section collects any data citations, data availability statements, or supplementary materials included in this article.

    Supplementary Materials

    S1 Table. Search strategy.

    (DOCX)

    S1 File. PRISMA checklist.

    (DOC)

    S2 File. Data extracted from eligible studies.

    Data were also deposited in Systematic Review Data Repository (SRDR): https://srdr.ahrq.gov/projects/1639.

    (XLSX)

    Attachment

    Submitted filename: Response to Reviewers.docx

    Data Availability Statement

    All data files are available from the Systematic Review Data Repository (SRDR) database (accession number 1639, URL: https://srdr.ahrq.gov/projects/1639).


    Articles from PLoS ONE are provided here courtesy of PLOS

    RESOURCES