Supplemental Digital Content is available in the text
Keywords: BRAF, diagnostic accuracy, genotyping, liquid biopsy, plasma
Abstract
Background:
Testing of B-Raf proto-oncogene (BRAF) mutation in tumor is necessary before targeted therapies are given. When tumor samples are not available, plasma samples are commonly used for the testing of BRAF mutation. The aim of this study was to investigate the diagnostic accuracy of BRAF mutation testing using plasma sample of cancer patients.
Methods:
Databases of Pubmed, Embase, and Cochrane Library were searched for eligible studies investigating BRAF mutation in paired tissue and plasma samples of cancer patients. A total of 798 publications were identified after database searching. After removing 229 duplicated publications, 569 studies were screened using the following exclusion criteria: (1) BRAF mutation not measured in plasma or in tumor sample; (2) lacking BRAF-wildtype or BRAF-mutated samples; (3) tissue and plasma samples not paired; (4) lacking tumor or plasma samples; (5) not plasma sample; (6) not cancer; (7) un-interpretable data. Accuracy data and relevant information were extracted from each eligible study by 2 independent researchers and analyzed using statistical software.
Results:
After pooling the accuracy data from 3943 patients of the 53 eligible studies, the pooled sensitivity, specificity, and diagnostic odds ratio of BRAF mutation testing using plasma sample were 69%, 98%, and 55.78, respectively. Area under curve of summary receiver operating characteristic curve was 0.9435. Subgroup analysis indicated that BRAF mutation testing using plasma had overall higher accuracy (diagnostic odds ratio of 89.17) in colorectal cancer, compared to melanoma and thyroid carcinoma. In addition, next-generation sequencing had an overall higher accuracy in detecting BRAF mutation using plasma sample (diagnostic odds ratio of 63.90), compared to digital polymerase chain reaction (PCR) and conventional PCR, while digital PCR showed the highest sensitivity (74%) among the 3 techniques.
Conclusion:
BRAF testing using plasma sample showed an overall high accuracy compared to paired tumor tissue sample, which could be used for cancer genotyping when tissue sample is not available. Large prospective studies are needed to further investigate the accuracy of BRAF mutation testing in plasma sample.
1. Introduction
During the development of cancer, tumor cells accumulate hundreds of mutations, a subset of which was found to play key roles in cancer development and progression.[1,2] As one of those so-called “driver mutations,” B-Raf proto-oncogene (BRAF) mutation was observed in many types of cancer, which is most prevalent in thyroid carcinoma, melanoma, colorectal cancer (CRC), and non-small cell lung cancer (NSCLC).[3] On the basis of those findings, targeted therapies on BRAF-mutant cancer have been developed. Two specific inhibitors for BRAF, Vemurafenib, and Dabrafenib, have been approved for treatment of advanced-stage melanoma patients with BRAF V600E mutation in 2011 and 2013, respectively.[4,5] In combination with mitogen-activated protein kinase inhibitor, dual inhibition on BRAF and mitogen-activated protein kinase kinase showed significant improvement of patient prognosis and was approved by Food and Drug Administration for treatment of BRAF-mutant advanced melanoma and BRAF-mutant advanced NSCLC.[6–8]
Before those targeted therapies are given, it is required to determine the BRAF mutation status of tumor.[9] When available, tumor tissue is a more reliable sample type for the testing of BRAF mutation status due to its high abundance of tumor DNA.[10,11] However, tissue sample is sometimes not available (e.g., in metastatic or recurrent cancer patients), and liquid biopsy sample (e.g., plasma, urine, etc) could serve as an alternative.[12,13] Liquid biopsy sample contains circulating tumor DNA (ctDNA) which derives from tumor cells and carries tumor-specific mutations,[13] making it possible to determine the gene mutation status in tumor using liquid biopsy samples.
Due to the low abundance of ctDNA,[14] measurement of tumor-specific mutations using liquid biopsy samples requires highly-sensitive techniques (e.g., digital polymerase chain reaction [PCR]), and their reliability is still under debate. Many studies have investigated the accuracy of BRAF mutation testing using liquid biopsy samples.[15–17] In this systemic review and meta-analysis, we aimed to investigate the diagnostic accuracy of BRAF mutation testing using ctDNA in plasma samples, with BRAF mutation status in paired tissue sample as reference.
2. Methods
2.1. Literature searching and selection of publication
Literature search was performed independently by PY and PC in April 2020. Databases including Pubmed, Embase, and Cochrane Library were searched using keywords “BRAF,” “cell-free DNA,” “circulating tumor DNA,” “plasma,” and “cancer,” and alternative spelling or abbreviations were also searched. After obtaining the searching results, duplicates were firstly removed and irrelevant studies were excluded after carefully reviewing the title and abstract of publications using the following criteria. Inclusion criteria: all original studies describing accuracy of BRAF mutation testing using plasma samples from patients with cancer, with tissue sample as reference. Exclusion criteria:
-
(1)
not a human study;
-
(2)
not describing BRAF mutation;
-
(3)
no plasma or tissue samples included;
-
(4)
not from patients with cancer;
-
(5)
reviews, abstracts, letter to the editor, comments, case reports, or studies with un-interpretable data.
Full text of the rest publications were then downloaded and examined carefully by 2 investigators. Publications were further excluded due to:
-
(1)
BRAF mutation was not measured in plasma or in tumor sample;
-
(2)
lacking BRAF-wildtype or BRAF-mutated samples;
-
(3)
tissue and plasma samples were not paired;
-
(4)
lacking tumor or plasma samples;
-
(5)
not plasma sample;
-
(6)
not cancer;
-
(7)
un-interpretable data (data were mixed with other genes, or difficult to extract accuracy data from the results).
For the rest eligible studies, accuracy data were extracted from BRAF mutation results from paired plasma and tissue samples, which included true positive, false positive, false negative, true negative, and sample size. Other relevant information was also extracted, including cancer type, technique used to detect BRAF mutation in plasma and tissue samples, region of the study. When several techniques were used to detect BRAF mutation in plasma sample from the same cohort of patients, only 1 of those techniques was used for data extraction and the selection criteria was:
-
(1)
technique used for a larger number of samples;
-
(2)
technique with similar detection region with the one used for paired tissue sample.
When a series of plasma samples were collected at multiple time points, results of plasma sample collected at the time point which was closest to the collection time point of tissue samples (usually at baseline) were used. Quality assessment of diagnostic accuracy studies 2 was also used to evaluate every eligible studies.[18] When there was disagreement between the 2 investigators (PY and PC), it was solved by a third investigator (JZ). Ethical approval was not necessary for this study because all the data obtained and analyzed were extracted from previously-published literature and not on individual patients.
2.2. Statistical analysis
The accuracy parameters of the eligible studies were pooled or calculated using Meta-DiSc 1.4, including sensitivity, specificity, positive likelihood ratio (PLR), negative likelihood ratio (NLR), diagnostic odds ratio (DOR), and area under curve (AUC) of summary receiver operating characteristic (SROC) curve. When significant heterogeneity was observed (I2 ≥ 50% and P ≤ .05) during the pooling, random effects model (DerSimonian-Laird model) was used; otherwise, fixed effects model (Mantel-Haenszel model) was used. When significant inter-study heterogeneity was observed after evaluating Cochran-Q and I2, threshold analysis and meta-regression were used to investigate potential source of heterogeneity using Meta-DiSc 1.4. Deek funnel plot asymmetry test was used to evaluate potential publication bias using STATA 12.0 (STATA Corp.). Results were considered statistical significant if P < .05.
3. Results
3.1. Search results
As shown in Figure 1, a total of 798 publications were identified after searching Pubmed (n = 395), Embase (n = 354), and Cochrane Library (n = 49). After removing duplicates, titles and abstracts of 569 publications were screened and another 445 irrelevant publications were excluded. Full text of the rest 124 studies were downloaded and evaluated, and another 71 studies were further excluded due to lacking of BRAF-wildtype or -mutated samples, or due to un-interpretable data. Data from the rest 53 eligible studies were extracted (see Table S1, Supplemental Digital Content, http://links.lww.com/MD2/A778 which summarizes the extracted data from eligible studies), and meta-analysis was performed.
Figure 1.
Preferred Reporting Items for Systematic Reviews and Meta-Analyses 2009 flow diagram.
3.2. Review of eligible publications
In the 53 eligible studies, 21 studies used next-generation sequencing (NGS) to test BRAF mutation in plasma sample, while this number was 13 for digital PCR, 16 for conventional PCR, and 3 for MassARRAY (Table 1). For the testing of BRAF mutation in paired tissue sample, more than half (30 out of 53) of the eligible studies used the same technique as plasma sample (15/21 for NGS, 3/13 for digital PCR, 10/16 for conventional PCR, and 2/3 for MassARRAY). In the rest 23 studies, 16 studies used standard of care instead (4/21 for NGS, 6/13 for digital PCR, 5/16 for conventional PCR, and 1/3 for MassARRAY), 4 studies used Sanger sequencing (1/21 for NGS, and 3/13 for digital PCR), 2 studies used conventional PCR (1/21 for NGS, and 1/13 for digital PCR), and 1 study used pyrosequencing (1/16 for conventional PCR). Overall, for the testing of BRAF mutation in tissue sample, 15 studies used NGS, 3 used digital PCR, 12 used conventional PCR, 2 used MassARRAY, 16 used standard of care, 4 used Sanger sequencing, and 1 used pyrosequencing.
Table 1.
Summary of studies comparing BRAF mutation status in plasma and tumor tissue samples from cancer patients.
| Author, year | Sample size | Type of cancer | Detection method (plasma) | Detection method (tissue) | Region |
| Gupta et al, 2020[15] | 75 | Colorectal cancer | NGS | NGS | America |
| Tzanikou et al, 2020[16] | 34 | Melanoma | Digital PCR | Sanger sequencing | Europe |
| Nguyen et al, 2020[26] | 50 | Colorectal cancer | NGS | NGS | Asia |
| García-Romero et al, 2019[46] | 13 | Central nervous system tumors | Digital PCR | Sanger sequencing | Europe |
| Maurel et al, 2019[51] | 178 | Colorectal cancer | PCR | PCR | Europe |
| Wong et al, 2017[32] | 52 | Melanoma | NGS | NGS | Australia |
| Iyer et al, 2018[19] | 44 | Thyroid carcinoma | NGS | NGS | America |
| Diefenbach et al, 2019[27] | 10 | Melanoma | NGS | NGS | Australia |
| Li et al, 2019[47] | 59 | Thyroid carcinoma | Digital PCR | Digital PCR | Asia |
| Choi et al, 2019[20] | 61 | Colorectal cancer | NGS | NGS | America |
| Sakai et al, 2015[33] | 15 | Colorectal cancer | NGS | NGS | Asia |
| Lin et al, 2014[17] | 191 | Colorectal cancer | MassARRAY | MassARRAY | Asia |
| Spindler et al, 2013[55] | 94 | Colorectal cancer | PCR | PCR | Europe |
| Leighl et al, 2019[21] | 92 | Lung cancer | NGS | Standard of care | America |
| Mas et al, 2019[28] | 405 | Colorectal cancer | NGS | Standard of care | Europe |
| Haselmann et al, 2018[49] | 187 | Melanoma | Digital PCR | Sanger sequencing | Europe |
| Liebs et al, 2019[39] | 53 | Colorectal cancer | Digital PCR | Digital PCR | Europe |
| Tang et al, 2018[48] | 57 | Melanoma | Digital PCR | Standard of care | Asia |
| Mohrmann et al, 2018 [40] | 41 | Mixed type | Digital PCR | Standard of care | America |
| Gangadhar et al, 2018[34] | 25 | Melanoma | NGS | NGS | America |
| Long-Mira et al, 2018[52] | 19 | Melanoma | PCR | Pyrosequencing | Europe |
| Sclafani et al, 2018[41] | 97 | Colorectal cancer | Digital PCR | PCR | Europe |
| Thierry et al, 2017[63] | 97 | Colorectal cancer | PCR | Standard of care | Europe |
| Mithraprabhu et al, 2017[30] | 48 | Multiple myeloma | NGS | NGS | Australia |
| Sandulache et al, 2017[22] | 23 | Thyroid carcinoma | NGS | NGS | America |
| Wang et al, 2017[35] | 103 | Lung cancer | NGS | PCR | Asia |
| Yang et al, 2017[65] | 107 | Lung cancer | PCR | PCR | Asia |
| Kidess-Sigal et al, 2016[36] | 3 | Colorectal cancer | NGS | Sanger sequencing | America |
| Jovelet et al, 2016[29] | 283 | Mixed type | NGS | NGS | Europe |
| Janku et al, 2016[53] | 160 | Mixed type | PCR | Standard of care | America |
| Andersen et al, 2016[42] | 11 | Cholangiocarcinoma | Digital PCR | Standard of care | Europe |
| Beranek et al, 2016[31] | 32 | Colorectal cancer | NGS | staNdard of care | Europe |
| Janku et al, 2015[50] | 137 | Mixed type | digital PCR | Standard of care | America |
| Gonzalez-Cao et al, 2015[62] | 92 | Mixed type | PCR | PCR | Europe |
| Kim et al, 2015[23] | 27 | Mixed type | NGS | Standard of care | Asia |
| Thierry et al, 2014[64] | 95 | Colorectal cancer | PCR | Standard of care | Europe |
| Oxnard et al, 2014[43] | 13 | Melanoma | digital PCR | Standard of care | America |
| Perkins et al, 2012[67] | 85 | Mixed type | MassARRAY | MassARRAY | Europe |
| Solit et al, 2008 [58] | 13 | Melanoma | PCR | PCR | America |
| Yancovitz et al, 2007[59] | 17 | Melanoma | PCR | PCR | America |
| Arnold et al, 2020[66] | 28 | Mixed type | PCR | Standard of care | America |
| Khatami et al, 2020[56] | 57 | Thyroid carcinoma | PCR | PCR | Asia |
| Liu et al, 2019[57] | 175 | Colorectal cancer | PCR | PCR | Asia |
| Kato et al, 2019[24] | 76 | Colorectal cancer | NGS | NGS | America |
| Janku et al, 2019[25] | 22 | Histiocytosis | NGS | NGS | America |
| Gray et al, 2019[68] | 51 | Melanoma | MassARRAY | Standard of care | Australia |
| Burjanivova et al, 2019[44] | 87 | Melanoma | Digital PCR | digital PCR | Europe |
| Jin et al, 2018[37] | 14 | Colorectal cancer | NGS | NGS | Asia |
| Kidess et al, 2015 [38] | 38 | Colorectal cancer | NGS | NGS | America |
| Hyman et al, 2015[45] | 13 | Histiocytosis | Digital PCR | Standard of care | America |
| Aung et al, 2014[54] | 108 | melanoma | PCR | Standard of care | Europe |
| Cradic et al, 2009 [60] | 56 | Thyroid carcinoma | PCR | PCR | America |
| Lilleberg et al, 2004[61] | 20 | Colorectal cancer | PCR | PCR | America |
NGS = next generation sequencing, PCR = polymerase chain reaction.
Detailed accuracy results of those studies are summarized below.
3.2.1. NGS
In the 21 studies using NGS for plasma sample, 8 studies by Gupta,[15] Iyer,[19] Choi,[20] Leighl,[21] Sandulache,[22] Kim,[23] Kato,[24] and Janku[25] used commercial Guardant NGS panel (Guardant Health) and the sensitivity ranged from 50.0%[20] to 100%,[21,23] and specificity were all high (from 89.5%[20] to 100%[19,21–23,25]). The concordance rate ranged from 72.7%[25] to 100%.[21,23] In the study by Leighl et al,[21]BRAF V600E mutation was tested in 92 paired plasma and tissue samples of patients with metastatic NSCLC, and results showed complete agreement between plasma and tissue. Similarly, study by Kim et al[23] also showed 100% agreement in BRAF V600E mutation statuses between 22 paired plasma and tissue samples of patients with CRC or melanoma.
Another 6 studies also used commercial NGS panel for BRAF mutation testing in plasma sample. Nguyen et al[26] used commercial xGen predesigned gene capture pools (Integrated DNA Technologies) and obtained complete agreement of BRAF mutation results between plasma and tumor tissue sample from 50 CRC patients. Diefenbach et al[27] used whole exome sequencing panel (SureSelect, Agilent) in 10 melanoma patients and the calculated sensitivity and specificity were 66.7% and 100%, respectively, with concordance rate at 80%. Mas et al[28] used AmpliSeq Colon and Lung Cancer Panel V2 (Life Technology) and tested BRAF mutation in plasma samples from 405 CRC patients, and the sensitivity, specificity, and overall concordance rate were 76.7%, 98.9%, and 97.3%, respectively. Jovelet et al[29] also used commercial panel from Life Technology (Cancer Hotspot Panel V2) in plasma samples from 283 patients with various types of cancer, and results showed sensitivity of only 25%, but high specificity (100%) and overall concordance rate (98.9%). Mithraprabhu et al[30] used OnTarget Mutation Detection platform (Boreal Genomics, Canada) for plasma samples from 48 patients with multiple myeloma, and the sensitivity was 50%, and specificity and concordance rate were 97.6% and 91.7%, respectively. Beranek et al[31] used Somatic 1 Master Kit (Multiplicom, Belgium) for BRAF mutation testing in plasma samples from 32 CRC patients, and results showed a complete agreement between plasma and paired tissue sample results.
The rest 7 studies used customized targeted NGS panels instead. Wong et al[32] sequenced 15 genes using Access ArrayTM system (Fluidigm) in plasma samples from 52 melanoma patients and results showed sensitivity of 75.7%, specificity of 100%, and concordance rate of 82.7%. Sakai et al[33] used a customized NGS panel targeting Kirsten rat sarcoma viral oncogene homolog, neuroblastoma ras oncogene, and BRAF in plasma samples of 15 CRC patients, and achieved 100% agreement between plasma and tissue results. Gangadhar et al[34] used a customized 61-gene panel to test BRAF mutation in plasma samples from 25 melanoma patients, and the sensitivity was 20% only, with high specificity of 93.3% and concordance rate of 64%. Wang et al[35] used a highly sensitive NGS-based technique, cSMART, and obtained complete agreement between plasma and tissue samples of 103 patients with advance stage lung adenocarcinoma. The rest 3 studies by Kidess-Sigal et al,[36] Jin et al,[37] and Kidess et al[38] all used a multiplexed synchronous coefficient of drag alteration mutation enrichment and detection platform, and all achieved 100% agreement between plasma and tissue samples from CRC patients.
3.2.2. Digital PCR
Eight of the 13 studies using digital PCR used droplet digital PCR (Bio-Rad) for BRAF mutation testing in plasma samples of cancer patients.[16,39–45] Results showed a highly variable sensitivity from 20% to 100%. The specificity of the 8 studies was all high, ranging from 89.3% to 100%, with concordance rate from 72.7% to 100%.
In the rest 5 studies, 3 studies by García-Romero et al,[46] Li et al,[47] and Tang et al[48] used QuantStudioTM 3D digital PCR system (ThermoFisher Scientific), and the calculated sensitivity was 25.0%, 61.5%, and 76.0%, respectively. The specificity was 77.8%, 90.9%, and 28.6%, with concordance rate at 61.5%, 78.0%, and 70.2%, respectively. The rest 2 studies used BEAMing instead. Haselmann et al[49] tested BRAF mutation in plasma samples of 187 melanoma patients using BEAMing, and the sensitivity and specificity were 86.2% and 93.4%, with concordance rate at 90.9%. Study by Janku et al[50] also used BEAMing in 137 cancer patients and results showed calculated sensitivity, specificity, and concordance rate of 76.3%, 96.0%, and 90.5%, respectively.
3.2.3. Conventional PCR
The conventional PCR discussed in this section included real-time PCR, amplification refractory mutation system, mutation/allele-specific PCR, and quantitative PCR. In those 16 studies using conventional PCR for BRAF mutation testing in plasma sample, 3 of them[51–53] used real-time PCR performed on IdyllaTM platform (Biocartis, Belgium), and the calculated sensitivity ranged from 64.3%[51] to 98.0%[53], with specificity ranging from 88.1%[53] to 99.4%,[51] and concordance rate from 84.2%[52] to 96.6%.[51]
Four studies used amplification refractory mutation system for BRAF mutation testing in plasma.[54–57] The sensitivity was from 94.1% to 100%, specificity was from 64.8% to 100%, and concordance rate was from 64.8% to 100%. Spindler et al[55] tested BRAF mutation in plasma samples from 94 CRC patients, and obtained 100% agreement between plasma and tissue results.
Five studies used mutation/allele-specific PCR to detect BRAF mutation in plasma samples.[58–61] Solit et al[58] detected BRAF mutation in plasma samples from 13 melanoma patients and results showed sensitivity, specificity, and concordance rate of 66.7%, 76.9%, and 76.9%, respectively. Yancovitz et al[59] tested BRAF mutation in 17 melanoma patients and the calculated sensitivity, specificity, and concordance rate were 60%, 58.8%, and 58.8%, respectively. Gonzalez-Cao et al[62] measured BRAF mutation in plasma of 92 patients and got a 100% sensitivity, 73.9% specificity, and 73.9% concordance rate. Plasma samples from 56 thyroid carcinoma patients were tested for BRAF mutation using allele-specific real-time PCR, and results showed 92.9% sensitivity, 37.5% specificity, and 37.5% overall concordance rate.[60] Lilleberg et al[61] used allele-specific PCR combined with denaturing high-performance liquid chromatography, and achieved complete agreement in BRAF mutation results between plasma and tissue samples of 20 CRC patients.
In the rest 4 studies, Thierry et al used an optimized quantitative PCR method to detect BRAF mutation in plasma samples from 97 CRC patients, and obtained sensitivity, specificity, and concordance rate of 88.9%, 86.6%, and 86.6%.[63] Another study by Thierry et al used the same method in 95 CRC patients and achieved 100% agreement between plasma and tissue results.[64] Yang et al used CastPCR and the calculated sensitivity and specificity were 93.0% and 88.8%, with overall concordance rate of 88.8%.[65] Arnold et al used a real-time PCR-based Target Selector ctDNA platform and results showed calculated sensitivity of 100%, specificity of 92.9%, and concordance of 92.9%.[66]
3.2.4. MassARRAY
Only 3 studies used MassARRAY to test BRAF mutation in plasma sample of cancer patients.[17,67,68] Specificity of the 3 studies were all 100%, with sensitivity ranging from 37.5%,[17] 75%,[67] to 92.5%,[68] and concordance rate from 94.1%[68] to 97.6%.[67]
In summary, the 53 studies comprised 3943 cancer patients with paired plasma and tumor tissue samples. High concordance rate (≥ 80%) was observed in majority (42/53) of the studies, while 46 studies (86.8%) showed high specificity (≥ 80%). High sensitivity was observed in more than half of the studies (31/53).
3.3. Quality assessment of eligible studies
Quality of each eligible study was assessed using quality assessment of diagnostic accuracy studies 2, as shown in Table 2. In the assessment of risk of bias, the percentage of high risk ranged from 0% (n = 0, patient selection, reference standard) to 6% (n = 3, flow and timing), while percentage of low risk ranged from 19% (n = 10, flow and timing) to 36% (n = 19, patient selection). Flow and timing showed the highest risk of bias (6% high risk and 19% low risk) among the 4 aspects in risk of bias assessment. In applicability concerns, index test showed the highest risk (2% high risk and 55% low risk), while reference standard showed the lowest risk (100% low risk).
Table 2.
QUADAS-2 assessment of eligible studies.
| Risk of bias | Applicability concerns | ||||||
| Author, year | Patient selection | Index test | Reference standard | Flow and timing | Patient selection | Index test | Reference standard |
| Gupta et al, 2020[15] | Unclear | Unclear | Unclear | Unclear | Low | Low | Low |
| Tzanikou et al, 2020[16] | Unclear | Unclear | Unclear | Unclear | Unclear | Low | Low |
| Nguyen et al, 2020[26] | Unclear | Unclear | Unclear | Unclear | Low | Unclear | Low |
| García-Romero et al, 2019[46] | Unclear | Unclear | Unclear | Unclear | Unclear | Low | Low |
| Maurel et al, 2019[51] | Low | Unclear | Unclear | Unclear | Low | Unclear | Low |
| Wong et al, 2017[32] | Unclear | Unclear | Unclear | Unclear | Low | Unclear | Low |
| Iyer et al, 2018[19] | Unclear | Unclear | Unclear | Unclear | Low | Low | Low |
| Diefenbach et al, 2019[27] | Unclear | Unclear | Unclear | Unclear | Low | Unclear | Low |
| Li et al, 2019[47] | Unclear | Unclear | Unclear | Unclear | Low | Unclear | Low |
| Choi et al, 2019[20] | Low | Unclear | Unclear | Unclear | Low | Unclear | Low |
| Sakai et al, 2015[33] | Unclear | Unclear | Unclear | Unclear | Low | Low | Low |
| Lin et al, 2014[17] | Unclear | Unclear | Unclear | Low | Low | Unclear | Low |
| Spindler et al, 2013[55] | Unclear | Unclear | Unclear | Unclear | Low | Unclear | Low |
| Leighl et al, 2019[21] | Unclear | Unclear | Unclear | Low | Low | Unclear | Low |
| Mas et al, 2019[28] | Low | Low | Low | Unclear | Low | Low | Low |
| Haselmann et al, 2018[49] | Low | Low | Low | Unclear | Low | Low | Low |
| Liebs et al, 2019[39] | Unclear | Unclear | Unclear | Low | Low | Low | Low |
| Tang et al, 2018[48] | Unclear | Unclear | Unclear | Unclear | Low | Unclear | Low |
| Mohrmann et al, 2018[40] | Low | Unclear | Unclear | Unclear | Low | Unclear | Low |
| Gangadhar et al, 2018[34] | Unclear | Low | Unclear | High | Low | Low | Low |
| Long-Mira et al, 2018[52] | Low | Unclear | Low | Unclear | Low | Low | Low |
| Sclafani et al, 2018[41] | Low | Low | Low | Unclear | Low | Low | Low |
| Thierry et al, 2017[63] | Low | Low | Low | Unclear | Low | Low | Low |
| Mithraprabhu et al, 2017[30] | Low | Unclear | Unclear | Unclear | Low | Unclear | Low |
| Sandulache et al, 2017[22] | Low | Unclear | Unclear | Low | Low | Low | Low |
| Wang et al, 2017[35] | Unclear | Unclear | Unclear | Unclear | Low | Unclear | Low |
| Yang et al, 2017[65] | Unclear | Unclear | Unclear | High | Low | Low | Low |
| Kidess-Sigal et al, 2016[36] | Unclear | Unclear | Unclear | Unclear | Low | Unclear | Low |
| Jovelet et al, 2016[29] | Low | Low | Low | Unclear | Low | Low | Low |
| Janku et al, 2016[53] | Unclear | Unclear | Unclear | Unclear | Low | Low | Low |
| Andersen et al, 2016[42] | Unclear | High | Low | Unclear | Unclear | High | Low |
| Beranek et al, 2016[31] | Unclear | Unclear | Unclear | Unclear | Low | Unclear | Low |
| Janku et al, 2015[50] | Unclear | Low | Low | High | Low | Low | Low |
| Gonzalez-Cao et al, 2015[62] | Unclear | Unclear | Unclear | Unclear | Low | Unclear | Low |
| Kim et al, 2015[23] | Low | Low | Low | Low | Low | Low | Low |
| Thierry et al, 2014[64] | Unclear | Low | Low | Unclear | Low | Low | Low |
| Oxnard et al, 2014[43] | Unclear | High | Unclear | Low | Low | Unclear | Low |
| Perkins et al, 2012[67] | Unclear | Low | Low | Low | Low | Low | Low |
| Solit et al, 2008[58] | Unclear | Unclear | Unclear | Unclear | Low | Unclear | Low |
| Yancovitz et al, 2007[59] | Unclear | Unclear | Unclear | Unclear | Low | Unclear | Low |
| Arnold et al, 2020[66] | Unclear | Low | Low | Unclear | Unclear | Low | Low |
| Khatami et al, 2020[56] | Low | Unclear | Unclear | Low | Low | Unclear | Low |
| Liu et al, 2019[57] | Unclear | Unclear | Unclear | Low | Low | Low | Low |
| Kato et al, 2019[24] | Low | Unclear | Unclear | Unclear | Low | Low | Low |
| Janku et al, 2019[25] | Unclear | Unclear | Unclear | Unclear | Low | Low | Low |
| Gray et al, 2019[68] | Unclear | Low | Low | Unclear | Low | Low | Low |
| Burjanivova et al, 2019[44] | Low | Unclear | Unclear | Unclear | Low | Unclear | Low |
| Jin et al, 2018[37] | Low | Unclear | Unclear | Unclear | Low | Unclear | Low |
| Kidess et al, 2015[38] | Unclear | Unclear | Unclear | Low | Low | Low | Low |
| Hyman et al, 2015[45] | Low | Low | Low | Unclear | Low | Low | Low |
| Aung et al, 2014[54] | Low | Low | Low | Unclear | Low | Low | Low |
| Cradic et al, 2009[60] | Low | Low | Low | Unclear | Low | Low | Low |
| Lilleberg et al, 2004[61] | Unclear | Unclear | Unclear | Unclear | Low | Unclear | Low |
high = high risk, low = low risk, unclear = unclear risk.
3.4. Meta-analysis of the accuracy of BRAF mutation testing using plasma samples
The BRAF mutation results in paired tissue and plasma samples from 3943 cancer patients were pooled using Meta-DiSc v1.4 statistical software. As shown in Figure 2, results showed pooled sensitivity of 0.69 (95% confidence interval [CI]: 0.66–0.72) and pooled specificity of 0.98 (95% CI: 0.97–0.98). Pooled PLR, NLR, and DOR were 16.84 (95% CI: 10.59–26.78), 0.35 (95% CI: 0.28–0.44), and 55.78 (95% CI: 33.62–92.54), respectively. AUC of the SROC curve was 0.9435 (also see Figure S2, Supplemental Digital Content, http://links.lww.com/MD2/A779 which illustrates the detailed and pooled PLR, NLR, and SROC curve).
Figure 2.
Detailed and pooled sensitivity, specificity, and DOR of the eligible studies. DOR = diagnostic odds ratio.
Since the forest plots indicated significant inter-study heterogeneity (I2 ≥ 50% and P ≤ .05), we further looked for possible sources of heterogeneity. Analysis of diagnostic threshold showed a Spearman correlation coefficient of –0.093 (P = .51), indicating no significant threshold effect. We then performed meta-regression analysis, and results indicated that inter-study heterogeneity was not associated with cancer type (P = .84), technique used for plasma sample (P = .86), technique used for tissue sample (P = .84), or region of the study (P = .76).
Subgroup analysis was performed on different cancer types. Eight of the 53 eligible studies were performed on patient cohorts of mixed types of cancer.[23,29,40,50,53,62,66,67] For those studies, we successfully separated the data by cancer types from 2 studies,[23,67] and the rest 6 studies[29,40,50,53,62,66] were excluded from subgroup analysis since we cannot separate their data by cancer type. After data separation, cancer types other than melanoma, CRC, and thyroid carcinoma were further excluded from subgroup analysis due to limited number of studies. As shown in Table 3, among the 3 cancer types, melanoma showed the highest pooled sensitivity (0.74 [95% CI: 0.69–0.79]), while CRC showed the highest specificity (0.99 [95% CI: 0.98–0.99]), PLR (32.79 [95% CI: 17.16–62.68]), and DOR (89.17 [95% CI: 50.65–156.97]), and thyroid carcinoma showed the highest AUC of SROC curve (0.9896).
Table 3.
Meta-analysis results.
| No. of studies/ patient cohorts | Sensitivity | Specificity | PLR | NLR | DOR | AUC of SROC curve | |
| Overall | 53 | 0.69 (0.66–0.72) | 0.98 (0.97–0.98) | 16.84 (10.59–26.78) | 0.35 (0.28–0.44) | 55.78 (33.62–92.54) | 0.9435 |
| Type of cancer | |||||||
| Melanoma | 15 | 0.74 (0.69–0.79) | 0.91 (0.88–0.94) | 6.06 (2.74–13.39) | 0.32 (0.19–0.52) | 23.29 (9.13–59.39) | 0.8962 |
| Colorectal cancer | 21 | 0.71 (0.62–0.78) | 0.99 (0.98–0.99) | 32.79 (17.16–62.68) | 0.34 (0.24–0.50) | 89.17 (50.65–156.97) | 0.9195 |
| Thyroid carcinoma | 5 | 0.58 (0.50–0.67) | 0.96 (0.90–0.99) | 12.21 (5.26–28.33) | 0.35 (0.13–0.92) | 25.85 (9.95–67.15) | 0.9896 |
| Techniques used for plasma sample | |||||||
| NGS | 21 | 0.71 (0.63–0.77) | 0.99 (0.98–0.99) | 23.61 (14.29–39.02) | 0.36 (0.25–0.51) | 63.90 (33.24–122.83) | 0.9336 |
| Digital PCR | 13 | 0.78 (0.72–0.82) | 0.94 (0.92–0.96) | 9.28 (3.66–23.54) | 0.32 (0.18–0.57) | 35.38 (12.81–97.71) | 0.9128 |
| Conventional PCR | 16 | 0.60 (0.55–0.65) | 0.97 (0.96–0.98) | 14.39 (6.39–32.42) | 0.38 (0.26–0.56) | 45.18 (16.82–121.31) | 0.8537 |
| Techniques used for plasma sample (for studies using standard of care for tissue sample) | |||||||
| NGS | 4 | 0.82 (0.66–0.92) | 0.99 (0.98–1.00) | 66.25 (27.32–160.69) | 0.21 (0.12–0.38) | 331.93 (107.84–1021.68) | 0.9889 |
| Digital PCR | 6 | 0.80 (0.72–0.87) | 0.94 (0.89–0.97) | 9.61 (1.19–77.69) | 0.23 (0.15–0.35) | 37.22 (5.52–250.91) | 0.8516 |
| Conventional PCR | 5 | 0.63 (0.55–0.70) | 0.96 (0.93–0.98) | 17.59 (5.08–60.88) | 0.37 (0.22–0.61) | 51.62 (12.05–221.04) | 0.2550 |
| Techniques used for plasma sample versus tissue sample | |||||||
| Matched | 30 | 0.63 (0.58–0.67) | 0.98 (0.97–0.99) | 15.39 (9.15–25.86) | 0.41 (0.31–0.54) | 51.25 (26.39–101.47) | 0.9193 |
| Unmatched | 23 | 0.75 (0.71–0.79) | 0.97 (0.96–0.98) | 17.10 (7.71–37.92) | 0.29 (0.20–0.40) | 61.07 (28.03–133.07) | 0.8702 |
AUC = area under curve, DOR = diagnostic odds ratio, NGS = next generation sequencing, NLR = negative likelihood ratio, PLR = positive likelihood ratio, SROC = summary receiver operating characteristic.
Subgroup analysis was also performed on techniques used for plasma sample. MassARRAY was excluded due to limited number of studies. In the rest 3 types of techniques (NGS, digital PCR, and conventional PCR), digital PCR showed the highest pooled sensitivity (0.78 [95% CI: 0.72–0.82]), and NGS showed the highest specificity (0.99 [95% CI: 0.98–0.99]), PLR (23.61 [95% CI: 14.29–39.02]), DOR (65.90 [95%CI: 33.24–122.83]), and AUC of SROC curve (0.9336).
Considering the different techniques used for paired tissue samples among the studies, we further analyzed the performance of the 4 techniques in plasma sample when a certain technique was used for tissue sample. When standard of care was used for tissue sample, NGS also had the best performance by showing the highest pooled sensitivity (0.82 [95% CI: 0.66–0.92]), specificity (0.99 [95% CI: 0.98–1.00]), PLR (66.25 [95% CI: 27.32–160.69]), DOR (331.93 [95% CI: 107.84–1021.68]), and AUC of SROC curve (0.9889), compared to digital PCR and conventional PCR (Table 3). MassARRAY was excluded from the analysis due to limited number of studies. When NGS was used for tissue sample, all of the studies (15/15) used NGS for plasma sample, and further analysis was not applicable. Similarly, when conventional PCR was used for tissue sample, majority of the studies (10/12) used conventional PCR for plasma sample, and further analysis was not performed due to limited number of studies using other techniques. For the rest techniques (digital PCR, MassARRAY, Sanger sequencing, and pyrosequencing), further analysis was also not performed due to limited number of studies.
Furthermore, we also divided the studies into 2 groups based on whether the study used the same technique in plasma and tissue samples (matched/unmatched). However, limited difference was observed in the performance of BRAF mutation testing in plasma sample between the matched and unmatched groups.
Deek funnel plot asymmetry test was used to evaluate publication bias since our study is investigating diagnostic accuracy. The test results showed no significant publication bias (P = .43, Fig. 3).
Figure 3.
Deek funnel plot.
4. Discussion
Precise measurement of BRAF mutation status in tumor is essential for the success of anti-BRAF targeted therapy, for example, Vemurafenib and Dabrafenib.[9] Tumor tissue samples (resection or biopsy) are commonly used for tumor genotyping, which is abundant in tumor-derived DNA.[10] When tumor tissue samples are not available (e.g., in recurrent or metastatic cancer), liquid biopsy samples (e.g., plasma, urine, and etc) are mostly used as an alternative to determine the mutation status in tumor.[12] However, the reliability of tumor genotyping using liquid biopsy samples needs to be validated. In this systemic review and meta-analysis, we investigated the accuracy of BRAF mutation detection using plasma sample, compared to paired tumor tissue sample.
In many previous studies, the accuracy of BRAF mutation detection in plasma samples has been validated using tissue sample as reference. In all, we involved 53 eligible studies in our systemic review and meta-analysis after database searching and screening. After pooling, BRAF mutation detection using plasma sample showed a moderate sensitivity (69%) and a high specificity (98%) as compared to tissue sample. The DOR, an important indicator of diagnostic test, was also quite high (55.78), and AUC of SROC curve was 0.9435. Those results indicated an overall high accuracy of BRAF mutation detection using plasma sample. Esagian et al compared tumor genotyping results using NGS in paired liquid biopsy and tissue biopsy samples of NSCLC patients, and reported a positive percent agreement of 53.9% for BRAF.[69] Since the study by Esagian et al only involved studies using NGS as the detection method and did not report sensitivity and specificity,[69] it is difficult to compare their results with findings of our meta-analysis.
During the data pooling, we observed significant inter-study heterogeneity. Therefore, we performed diagnostic threshold analysis and meta-regression. The analysis results did not shown significant threshold effect, and meta-regression also showed no significant association between inter-study heterogeneity and the covariates (cancer type, technique used for plasma sample, technique used for tissue sample, and region of the study). We further performed subgroup analysis based on cancer type and techniques used for plasma sample. For subgroup analysis on cancer type, we separated and pooled the results among melanoma, CRC, and thyroid carcinoma. Among the 3 types of cancer, CRC showed the highest specificity (99%), PLR (32.79), and DOR (89.17), indicating that an overall higher accuracy of plasma testing for BRAF mutation in CRC, although melanoma showed the highest sensitivity (74%) and thyroid carcinoma had the highest AUC of SROC curve (0.9896). Among the different techniques used for plasma sample, NGS showed the highest specificity (99%), PLR (23.61), DOR (63.90), and AUC of SROC curve (0.9336), while digital PCR had the highest sensitivity (78%). In addition, in studies using standard of care for tissue samples, NGS also showed the highest sensitivity (82%), specificity (99%), PLR (66.25), and DOR (331.93), and AUC of SROC curve (0.9889) for the detection of BRAF mutation in plasma samples, compared to digital PCR and conventional PCR. Those results indicate an overall higher accuracy of NGS in BRAF mutation testing using plasma sample. The differences in diagnostic accuracy among the subgroups might partially explain the inter-study heterogeneity observed in data pooling. Publication bias was also investigated using Deek funnel plot asymmetry test, and results indicated no significant publication bias.
In all, our study results indicated moderate sensitivity and high specificity and DOR of BRAF mutation testing using plasma sample. Overall, the testing of BRAF status using plasma sample showed high accuracy compared to paired tumor tissue sample of cancer patients, and could be used as an alternative when tissue sample is not available. Among the cancer types which most frequently carry BRAF mutation (melanoma, CRC, thyroid carcinoma), plasma sample showed the highest accuracy in CRC. Among different techniques used for plasma sample, NGS showed the highest accuracy and is more recommended for BRAF mutation testing using plasma sample. On the other hand, digital PCR showed the highest sensitivity and therefore is recommended if high sensitivity is expected. Limitation of this study may include the small number of studies in some subgroups (thyroid carcinoma) which should be treated carefully. In addition, although the performance of BRAF mutation testing between different techniques does not differ much in tissue sample due to high abundance of tumor DNA, difference in technique may cause potential bias. Large prospective studies are needed to further validate the accuracy of BRAF mutation testing using plasma sample.
Author contributions
Conceptualization: Peng Ye, Jie Zhang.
Data curation: Peng Ye, Peiling Cai.
Formal analysis: Peng Ye, Peiling Cai, Jing Xie.
Funding acquisition: Peng Ye.
Supervision: Jie Zhang.
Writing – original draft: Peng Ye.
Writing – review & editing: Peng Ye, Peiling Cai, Jing Xie, Jie Zhang.
Footnotes
Abbreviations: AUC = area under curve, BRAF = B-Raf proto-oncogene, CRC = colorectal cancer, ctDNA = circulating tumor DNA, DOR = diagnostic odds ratio, NGS = next-generation sequencing, NLR = negative likelihood ratio, NSCLC = non-small cell lung cancer, PCR = polymerase chain reaction, PLR = positive likelihood ratio, SROC = summary receiver operating characteristic.
How to cite this article: Ye P, Cai P, Xie J, Zhang J. Reliability of BRAF mutation detection using plasma sample: a systematic review and meta-analysis. Medicine. 2021;100:51(e28382).
All data generated or analyzed during this study are included in this published article [and its supplementary information files].
The authors have no conflicts of interest to disclose.
Supplemental digital content is available for this article. Direct URL citations appear in the printed text and are provided in the HTML and PDF versions of this article on the journal's Website (www.md-journal.com).
References
- [1].Kumar S, Gerstein M. Cancer genomics: less is more in the hunt for driver mutations. Nature 2017;547:40–1. [DOI] [PubMed] [Google Scholar]
- [2].Rheinbay E, Parasuraman P, Grimsby J, et al. Recurrent and functional regulatory mutations in breast cancer. Nature 2017;547:55–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [3].Zaman A, Wu W, Bivona TG. Targeting oncogenic BRAF: past, present, and future. Cancers 2019;11:1197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [4].FDA approves vemurafenib for treatment of metastatic melanoma. Oncology (Williston Park) 2011;25:906. [PubMed] [Google Scholar]
- [5].Ballantyne AD, Garnock-Jones KP. Dabrafenib: first global approval. Drugs 2013;73:1367–76. [DOI] [PubMed] [Google Scholar]
- [6].Long GV, Stroyakovskiy D, Gogas H, et al. Dabrafenib and trametinib versus dabrafenib and placebo for Val600 BRAF-mutant melanoma: a multicentre, double-blind, phase 3 randomised controlled trial. Lancet 2015;386:444–51. [DOI] [PubMed] [Google Scholar]
- [7].Ascierto PA, McArthur GA, Dreno B, et al. Cobimetinib combined with vemurafenib in advanced BRAF(V600)-mutant melanoma (coBRIM): updated efficacy results from a randomised, double-blind, phase 3 trial. Lancet Oncol 2016;17:1248–60. [DOI] [PubMed] [Google Scholar]
- [8].Planchard D, Smit EF, Groen HJM, et al. Dabrafenib plus trametinib in patients with previously untreated BRAF(V600E)-mutant metastatic non-small-cell lung cancer: an open-label, phase 2 trial. Lancet Oncol 2017;18:1307–16. [DOI] [PubMed] [Google Scholar]
- [9].Thompson CA. New melanoma drug requires gene mutation test. Am J Health Syst Pharm 2011;68:1764. [DOI] [PubMed] [Google Scholar]
- [10].Tran TV, Dang KX, Pham QH, et al. Evaluation of the expression levels of BRAF(V600E) mRNA in primary tumors of thyroid cancer using an ultrasensitive mutation assay. BMC cancer 2020;20:368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [11].Li X, Cai W, Yang G, et al. Comprehensive analysis of EGFR-mutant abundance and its effect on efficacy of EGFR TKIs in advanced NSCLC with EGFR mutations. J Thorac Oncol 2017;12:1388–97. [DOI] [PubMed] [Google Scholar]
- [12].Luchini C, Veronese N, Nottegar A, et al. Liquid biopsy as surrogate for tissue for molecular profiling in pancreatic cancer: a meta-analysis towards precision medicine. Cancers 2019;11:1152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [13].Crowley E, Di Nicolantonio F, Loupakis F, Bardelli A. Liquid biopsy: monitoring cancer-genetics in the blood. Nat Rev Clin Oncol 2013;10:472–84. [DOI] [PubMed] [Google Scholar]
- [14].Molparia B, Oliveira G, Wagner JL, Spencer EG, Torkamani A. A feasibility study of colorectal cancer diagnosis via circulating tumor DNA derived CNV detection. PloS one 2018;13:e0196826. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [15].Gupta R, Othman T, Chen C, Sandhu J, Ouyang C, Fakih M. Guardant360 circulating tumor DNA assay is concordant with foundation one next-generation sequencing in detecting actionable driver mutations in anti-EGFR naive metastatic colorectal cancer. Oncologist 2020;25:235–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [16].Tzanikou E, Haselmann V, Markou A, et al. Direct comparison study between droplet digital PCR and a combination of allele-specific PCR, asymmetric rapid PCR and melting curve analysis for the detection of BRAF V600E mutation in plasma from melanoma patients. Clin Chem Lab Med 2020;58:1799–807. [DOI] [PubMed] [Google Scholar]
- [17].Lin JK, Lin PC, Lin CH, et al. Clinical relevance of alterations in quantity and quality of plasma DNA in colorectal cancer patients: based on the mutation spectra detected in primary tumors. Ann Surg Oncol 2014;21: (Suppl 4): S680–6. [DOI] [PubMed] [Google Scholar]
- [18].Whiting PF, Rutjes AW, Westwood ME, et al. QUADAS-2: a revised tool for the quality assessment of diagnostic accuracy studies. Ann Intern Med 2011;155:529–36. [DOI] [PubMed] [Google Scholar]
- [19].Iyer PC, Cote GJ, Hai T, et al. Circulating BRAF V600E Cell-Free DNA as a biomarker in the management of anaplastic thyroid carcinoma. JCO Precis Oncol 2018;2:01–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [20].Choi IS, Kato S, Fanta PT, et al. Genomic profiling of blood-derived circulating tumor dna from patients with colorectal cancer: implications for response and resistance to targeted therapeutics. Mol Cancer Therap 2019;18:1852–62. [DOI] [PubMed] [Google Scholar]
- [21].Leighl NB, Page RD, Raymond VM, et al. Clinical utility of comprehensive cell-free DNA analysis to identify genomic biomarkers in patients with newly diagnosed metastatic non-small cell lung cancer. Clin Cancer Res 2019;25:4691–700. [DOI] [PubMed] [Google Scholar]
- [22].Sandulache VC, Williams MD, Lai SY, et al. Real-time genomic characterization utilizing circulating cell-free DNA in patients with anaplastic thyroid carcinoma. Thyroid 2017;27:81–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [23].Kim ST, Lee WS, Lanman RB, 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;6:40360–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [24].Kato S, Schwaederle MC, Fanta PT, et al. Genomic assessment of blood-derived circulating tumor DNA in patients with colorectal cancers: correlation with tissue sequencing, therapeutic response, and survival. JCO Precis Oncol 2019;3:PO.18.00158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [25].Janku F, Diamond EL, Goodman AM, et al. Molecular profiling of tumor tissue and plasma cell-free DNA from patients with non-langerhans cell histiocytosis. Mol Cancer Ther 2019;18:1149–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [26].Nguyen HT, Tran DH, Ngo QD, et al. Evaluation of a liquid biopsy protocol using ultra-deep massive parallel sequencing for detecting and quantifying circulation tumor DNA in colorectal cancer patients. Cancer Investig 2020;38:85–93. [DOI] [PubMed] [Google Scholar]
- [27].Diefenbach RJ, Lee JH, Strbenac D, et al. Analysis of the whole-exome sequencing of tumor and circulating tumor DNA in metastatic melanoma. Cancers 2019;11:1905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [28].Mas L, Bachet JB, Taly V, et al. BRAF mutation status in circulating tumor DNA from patients with metastatic colorectal cancer: extended mutation analysis from the AGEO RASANC study. Cancers 2019;11:998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [29].Jovelet C, Ileana E, Le Deley MC, et al. Circulating cell-free tumor DNA analysis of 50 genes by next-generation sequencing in the prospective MOSCATO trial. Clin Cancer Res Research 2016;22:2960–8. [DOI] [PubMed] [Google Scholar]
- [30].Mithraprabhu S, Khong T, Ramachandran M, et al. Circulating tumour DNA analysis demonstrates spatial mutational heterogeneity that coincides with disease relapse in myeloma. Leukemia 2017;31:1695–705. [DOI] [PubMed] [Google Scholar]
- [31].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;59:54–8. [DOI] [PubMed] [Google Scholar]
- [32].Wong SQ, Raleigh JM, Callahan J, et al. Circulating tumor DNA analysis and functional imaging provide complementary approaches for comprehensive disease monitoring in metastatic melanoma. JCO Precis Oncol 2017;1:01–14. [DOI] [PubMed] [Google Scholar]
- [33].Sakai K, Tsurutani J, Yamanaka T, et al. Extended RAS and BRAF mutation analysis using next-generation sequencing. PloS One 2015;10:e0121891. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [34].Gangadhar TC, Savitch SL, Yee SS, et al. Feasibility of monitoring advanced melanoma patients using cell-free DNA from plasma. Pigment Cell Melanoma Res 2018;31:73–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [35].Wang Z, Cheng G, Han X, et al. Application of single-molecule amplification and resequencing technology for broad surveillance of plasma mutations in patients with advanced lung adenocarcinoma. J Mol Diagn 2017;19:169–81. [DOI] [PubMed] [Google Scholar]
- [36].Kidess-Sigal E, Liu HE, Triboulet MM, 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;7:85349–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [37].Jin CE, Koo B, Lee TY, et al. Simple and low-cost sampling of cell-free nucleic acids from blood plasma for rapid and sensitive detection of circulating tumor DNA. Adv Sci (Weinh) 2018;5:1800614. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [38].Kidess E, Heirich K, Wiggin M, et al. Mutation profiling of tumor DNA from plasma and tumor tissue of colorectal cancer patients with a novel, high-sensitivity multiplexed mutation detection platform. Oncotarget 2015;6:2549–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [39].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 Med 2019;8:3761–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [40].Mohrmann L, Huang HJ, Hong DS, et al. Liquid biopsies using plasma exosomal nucleic acids and plasma cell-free DNA compared with clinical outcomes of patients with advanced cancers. Clin Cancer Res 2018;24:181–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [41].Sclafani F, Chau I, Cunningham D, et al. KRAS and BRAF mutations in circulating tumour DNA from locally advanced rectal cancer. Sci Rep 2018;8:1445. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [42].Andersen RF, Jakobsen A. Screening for circulating RAS/RAF mutations by multiplex digital PCR. Clin Chim Acta 2016;458:138–43. [DOI] [PubMed] [Google Scholar]
- [43].Oxnard GR, Paweletz CP, Kuang Y, et al. Noninvasive detection of response and resistance in EGFR-mutant lung cancer using quantitative next-generation genotyping of cell-free plasma DNA. Clin Cancer Res 2014;20:1698–705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [44].Burjanivova T, Malicherova B, Grendar M, et al. Detection of BRAFV600E mutation in melanoma patients by digital PCR of circulating DNA. Genet Test Mol Biomarkers 2019;23:241–5. [DOI] [PubMed] [Google Scholar]
- [45].Hyman DM, Diamond EL, Vibat CR, et al. Prospective blinded study of BRAFV600E mutation detection in cell-free DNA of patients with systemic histiocytic disorders. Cancer Discov 2015;5:64–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [46].Garcia-Romero N, Carrion-Navarro J, Areal-Hidalgo P, et al. BRAF V600E detection in liquid biopsies from pediatric central nervous system tumors. Cancers 2019;12:66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [47].Li H, Zhao J, Zhang J, et al. Detection of ctDNA in the plasma of patients with papillary thyroid carcinoma. Exp Ther Med 2019;18:3389–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [48].Tang H, Kong Y, Si L, et al. Clinical significance of BRAF(V600E) mutation in circulating tumor DNA in Chinese patients with melanoma. Oncol Lett 2018;15:1839–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [49].Haselmann V, Gebhardt C, Brechtel I, et al. Liquid profiling of circulating tumor DNA in plasma of melanoma patients for companion diagnostics and monitoring of BRAF inhibitor therapy. Clin Chem 2018;64:830–42. [DOI] [PubMed] [Google Scholar]
- [50].Janku F, Angenendt P, Tsimberidou AM, et al. Actionable mutations in plasma cell-free DNA in patients with advanced cancers referred for experimental targeted therapies. Oncotarget 2015;6:12809–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [51].Maurel J, Alonso V, Escudero P, et al. Clinical impact of circulating tumor RAS and BRAF Mutation dynamics in patients with metastatic colorectal cancer treated with first-line chemotherapy plus anti–epidermal growth factor receptor therapy. JCO Precis Oncol 2019;3:01–16. [DOI] [PubMed] [Google Scholar]
- [52].Long-Mira E, Ilie M, Chamorey E, et al. Monitoring BRAF and NRAS mutations with cell-free circulating tumor DNA from metastatic melanoma patients. Oncotarget 2018;9:36238–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [53].Janku F, Huang HJ, Claes B, et al. BRAF mutation testing in cell-free DNA from the plasma of patients with advanced cancers using a rapid, automated molecular diagnostics system. Mol Cancer Ther 2016;15:1397–404. [DOI] [PubMed] [Google Scholar]
- [54].Aung KL, Donald E, Ellison G, et al. Analytical validation of BRAF mutation testing from circulating free DNA using the amplification refractory mutation testing system. J Mol Diagn 2014;16:343–9. [DOI] [PubMed] [Google Scholar]
- [55].Spindler KG, Appelt AL, Pallisgaard N, Andersen RF, Jakobsen A. KRAS-mutated plasma DNA as predictor of outcome from irinotecan monotherapy in metastatic colorectal cancer. Br J Cancer 2013;109:3067–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [56].Khatami F, Larijani B, Heshmat R, et al. Hypermethylated RASSF1 and SLC5A8 promoters alongside BRAF(V600E) mutation as biomarkers for papillary thyroid carcinoma. J Cell Physiol 2020;235:6954–68. [DOI] [PubMed] [Google Scholar]
- [57].Liu XN, Tian Z, Wei XF, et al. Combined detection of KRAS, NRAS, BRAF and PIK3CA mutations in the plasma and tumor tissues of colorectal cancer patients. Zhonghua Bing Li Xue Za Zhi 2019;48:373–7. [DOI] [PubMed] [Google Scholar]
- [58].Solit DB, Osman I, Polsky D, et al. Phase II trial of 17-allylamino-17-demethoxygeldanamycin in patients with metastatic melanoma. Clin Cancer Res 2008;14:8302–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [59].Yancovitz M, Yoon J, Mikhail M, et al. Detection of mutant BRAF alleles in the plasma of patients with metastatic melanoma. J Mol Diagn 2007;9:178–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [60].Cradic KW, Milosevic D, Rosenberg AM, Erickson LA, McIver B, Grebe SK. Mutant BRAF(T1799A) can be detected in the blood of papillary thyroid carcinoma patients and correlates with disease status. J Clin Endocrinol Metab 2009;94:5001–9. [DOI] [PubMed] [Google Scholar]
- [61].Lilleberg SL, Durocher J, Sanders C, Walters K, Culver K. High sensitivity scanning of colorectal tumors and matched plasma DNA for mutations in APC, TP53, K-RAS, and BRAF genes with a novel DHPLC fluorescence detection platform. Ann N Y Acad Sci 2004;1022:250–6. [DOI] [PubMed] [Google Scholar]
- [62].Gonzalez-Cao M, Mayo-de-Las-Casas C, Molina-Vila MA, et al. BRAF mutation analysis in circulating free tumor DNA of melanoma patients treated with BRAF inhibitors. Melanoma Res 2015;25:486–95. [DOI] [PubMed] [Google Scholar]
- [63].Thierry AR, El Messaoudi S, Mollevi C, et al. Clinical utility of circulating DNA analysis for rapid detection of actionable mutations to select metastatic colorectal patients for anti-EGFR treatment. Ann Oncol 2017;28:2149–59. [DOI] [PubMed] [Google Scholar]
- [64].Thierry AR, Mouliere F, El Messaoudi S, et al. Clinical validation of the detection of KRAS and BRAF mutations from circulating tumor DNA. Nat Med 2014;20:430–5. [DOI] [PubMed] [Google Scholar]
- [65].Yang Y, Shen X, Li R, et al. The detection and significance of EGFR and BRAF in cell-free DNA of peripheral blood in NSCLC. Oncotarget 2017;8:49773–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [66].Arnold L, Alexiadis V, Watanaskul T, Zarrabi V, Poole J, Singh V. Clinical validation of qPCR Target Selector assays using highly specific switch-blockers for rare mutation detection. J Clin Pathol 2020;73:648–55. [DOI] [PubMed] [Google Scholar]
- [67].Perkins G, Yap TA, Pope L, et al. Multi-purpose utility of circulating plasma DNA testing in patients with advanced cancers. PloS One 2012;7:e47020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [68].Gray ES, Witkowski T, Pereira M, et al. Genomic analysis of circulating tumor DNA using a melanoma-specific UltraSEEK oncogene panel. J Mol Diagn 2019;21:418–26. [DOI] [PubMed] [Google Scholar]
- [69].Esagian SM, Grigoriadou G, Nikas IP, et al. Comparison of liquid-based to tissue-based biopsy analysis by targeted next generation sequencing in advanced non-small cell lung cancer: a comprehensive systematic review. J Cancer Res Clin Oncol 2020;146:2051–66. [DOI] [PMC free article] [PubMed] [Google Scholar]



