Next Article in Journal
In Chronic Lymphocytic Leukemia the JAK2/STAT3 Pathway Is Constitutively Activated and Its Inhibition Leads to CLL Cell Death Unaffected by the Protective Bone Marrow Microenvironment
Previous Article in Journal
Multi-Features Classification of Prostate Carcinoma Observed in Histological Sections: Analysis of Wavelet-Based Texture and Colour Features
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Assessing the Concordance of Genomic Alterations between Circulating-Free DNA and Tumour Tissue in Cancer Patients

by
Leila Jahangiri
1,2,* and
Tara Hurst
1
1
Department of Life Sciences, Birmingham City University, Birmingham B15 3TN, UK
2
Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Lab blocks level 3, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
*
Author to whom correspondence should be addressed.
Cancers 2019, 11(12), 1938; https://doi.org/10.3390/cancers11121938
Submission received: 13 November 2019 / Revised: 28 November 2019 / Accepted: 29 November 2019 / Published: 4 December 2019

Abstract

:
Somatic alterations to the genomes of solid tumours, which in some cases represent actionable drivers, provide diagnostic and prognostic insight into these complex diseases. Spatial and longitudinal tracking of somatic genomic alterations (SGAs) in patient tumours has emerged as a new avenue of investigation, not only as a disease monitoring strategy, but also to improve our understanding of heterogeneity and clonal evolution from diagnosis through disease progression. Furthermore, analysis of circulating-free DNA (cfDNA) in the so-called “liquid biopsy” has emerged as a non-invasive method to identify genomic information to inform targeted therapy and may also capture the heterogeneity of the primary and metastatic tumours. Considering the potential of cfDNA analysis as a translational laboratory tool in clinical practice, establishing the extent to which cfDNA represents the SGAs of tumours, particularly actionable driver alterations, becomes a matter of importance, warranting standardisation of methods and practices. Here, we assess the utilisation of cfDNA for molecular profiling of SGAs in tumour tissue across a broad range of solid tumours. Moreover, we examine the underlying factors contributing to discordance of detected SGAs between cfDNA and tumour tissue.

1. Introduction

Cancer genomes display a plethora of somatic genomic alterations (SGAs), including single nucleotide variations (SNVs), insertions and deletions (indels), focal amplifications, gene fusions, copy number alterations (CNAs) and numerical and segmental chromosomal alterations (NCAs and SCAs). Chromosome aberrations, including gene fusions and translocations are associated with many tumour types and the vast majority of metastatic cancers harbour chromosome arm level CNAs [1,2]. The current gold standard for molecular profiling in clinical practice is the identification of SGAs, specifically actionable driver alterations in tumour tissue, enabling stratification of patients into precise treatment regimens. In a classical view, driver and passenger alterations differ in actionability and a key task is distinguishing these [3]. A challenge of this task is the heterogeneity among somatic alterations, defined as the emergence of distinct subclones with divergent genetic profiles within the tumour, between the primary tumour and its metastases or between sequential tumour samples [4,5,6]. A potential solution to circumvent this issue, though impractical, is to capture a larger spectrum of the genomic landscape by obtaining multiple biopsies of a primary tumour and its metastases throughout the course of the disease. Alternatively, the use of circulating-free DNA (cfDNA), acquired through the so-called ‘liquid biopsy’, represents a rapid and non-invasive method for genomic profiling. The presence of cfDNA in the plasma or serum of cancer patients and the use of this tool for the analysis of SGAs, including actionable alterations, in cancers of tissues such as lung, breast, gastrointestinal tract, nervous system and prostate has been well documented [7,8,9,10,11,12,13,14,15,16,17,18]. These studies bring into focus the emergence of cfDNA as a potential translational tool for clinical practice, particularly in relation to the analysis of primary and metastatic tumour profiles. Furthermore, cfDNA has emerged as a useful surveillance tool for the early detection and prediction of prognosis in several cancers and has displayed correlations with disease burden and treatment response [19,20,21,22,23,24,25,26,27,28,29].
Regarding the potential for the use of cfDNA in routine clinical practice, establishing the extent to which cfDNA reflects the genomic landscape of tumours is significant. However, this task is hampered by numerous technical and biological challenges. Recent studies, though diverse in cohort size and design, have evaluated the feasibility of using cfDNA by measuring the degree of concordance between paired cfDNA and tumour samples and have attempted to dissect the underlying biological or technical factors contributing to discordance. Here we review these studies in the broader context of SGAs in solid tumours.

2. The Concordance Rate of SGAs between cfDNA and Tumour Tissue across Solid Tumours

Mutations in specific oncogenes are frequent signatures in solid and liquid tumours and the presence of these in cfDNA is concordant in variable degrees with that in the tumours. In this section, some studies leveraging the use of cfDNA for detecting SGA in breast, prostate, NSCLC, colorectal, neuroblastoma and oligodendroglioma cancers will be reviewed.
KRAS mutations arise in 50% of metastatic colorectal cancer (mCRC) cases, which can affect the response to EGFR pathway-targeted therapeutics [30]. In multiple studies of mCRC, cohorts of patients were tested for RAS status using standard-of-care PCR and ddPCR (BEAMing) or similar technologies for tissue and cfDNA, yielding 86.4–92% concordance rates [31,32,33]. In excess of 85% of lung cancers are classified as NSCLC, with several actionable alterations of EGFR and ALK contributing to its pathogenesis [34,35]. In a study conducted by Sung et al., 126 cases of NSCLC patient samples were analysed for concordance of cfDNA and tumour tissue using ultra-deep sequencing and tissue genotyping, respectively. Very high overall concordance rates for EGFR mutations (ex19del and L858R) were observed [15,36,37,38,39,40,41].
In the field of breast cancer, circulating tumour cells and cfDNA are promising analytes for prediction of survival and response to therapy [17,20,42,43]. An important cfDNA biomarker of breast cancer, hotspot mutations in ESR1, predicts resistance to endocrine therapy [32]. Takeshita et al., compare ESR1 mutation status of 35 cfDNA and matched tumour tissue in patients with metastatic breast cancer using ddPCR and find an overall concordance rate of 74.3% (26/35) [44]. Further, PIK3CA mutations, frequently detected in cfDNA in breast cancer and an indicator of tumour burden and treatment efficacy have been a subject of interest [45,46,47,48] since they also show high concordance between cfDNA and metastatic tumours [49].
In a recent study on metastatic prostate cancer, the concordance rate of 45 cfDNA and matched tissue biopsies for clinically-relevant genes was determined by targeted sequencing and whole exome sequencing (WES). This group found copy numbers of clinically actionable genes (i.e., AR, BRCA2, PIK3CA) to be 88.9% concordant between cfDNA and tumour DNA. While rearrangements detected in PTEN, APC, BRCA2 displayed 48% concordance [10].
In the field of neuroblastoma, MYCN amplification status, the strongest indicator of poor prognosis and aggressive behaviour [50,51], has been analysed using cfDNA [16]. In addition to MYCN, ALK activating alterations occurring in 10% of NB cases have been assayed in cfDNA using PCR-based methods [52]. Combaret et al., used ddPCR to evaluate the mutational status of ALK hotspots (F1174L exon 23:3520 and 3522, R1275Q exon 25:3824) using cfDNA in a cohort of 114 neuroblastoma patients. Their analysis revealed perfect agreement between cfDNA and tumour tissue for the F1174L ALK mutation (exon 23:3520), while discordance was observed for the other two mutations [52].
A range of SGAs including numerical chromosome alterations (NCAs), segmental chromosome alterations (SCAs) and large SCAs have been investigated in cfDNA in neuroblastoma patients [53,54,55]. Chicard et al., inferred CNAs (including large SCAs, SCAs and NCAs) in cfDNA and matched tumour tissue of neuroblastoma patients. The overall concordance of 97% for dynamic (non-silent) cfDNA and tumour profiles was reported while large SCAs also showed high levels of concordance [53]. In a later publication, this group utilised WES for both cfDNA and tumour tissue and found high concordance of CNAs between cfDNA and primary tumours at diagnosis (151/162) (93%) and with 11/162 (7%) cases of discordance (i.e., 2p gain in tumour only, in case 17) [54]. Good agreement between large structural alterations was also observed by Leary et al., in colorectal and breast cancers. In this study, entire chromosome-level and chromosome arm-level alterations were detected by whole genome sequencing (WGS). Tumour-derived chromosomal copy number changes (1p, 4q loss and 13q gain) and copy number changes of driver alterations including ERBB2 and CDK6 were detected in cfDNA of colorectal and breast cancer patients with good concordance rates when tumour tissue was available for analysis [56]. In a study conducted by Lavon et al., statistically significant concordance rates were detected for loss of heterozygosity (LOH) of 10q and 1p (79% and 62%, respectively) between cfDNA and tumour tissue of oligodendroglioma patients [57].
Standing in contrast to the four former studies, Molparia et al., in a cohort of 24 colorectal cancer patients, detect a lack of concordance between CNAs including deletions of 8p,18 and 9p of cfDNA and tumour tissue, highlighting the subclonal nature of CNAs in colorectal cancer [58]. In conclusion, these studies attest to the feasibility of using cfDNA as a tool for detecting a range of SGAs including structural alterations present in most cancers [2,59].

3. The Underlying Factors Contributing to Perceived Discordance between SGAs Detected in Solid Tumours and cfDNA

The inter-related technical and biological factors that may contribute to discordance between cfDNA and primary and metastatic tumours will be discussed in detail in this section. Figure 1 shows processing of cfDNA and primary and metastatic tumour tissue from sampling to analysis and the summary of contributing factors to discordance rates observed between SGAs in cfDNA and tumour tissue.

3.1. Tumour Fraction and Mutation Allele Frequency (MAF)

The tumour fraction is the proportion of tumour DNA in total cfDNA. Rapidly proliferating subpopulations of the tumour that outgrow their blood supply prior to apoptosis or necrosis would be expected to release DNA of different sizes into the peripheral blood [60,61]. The location, size and vascularity of the tumour can affect the accessibility of tumour DNA to the circulation, and hence impact tumour fraction [62,63,64]. Therefore, these biological factors can affect the release of tumour DNA in the blood, impacting their representation and detectability in cfDNA [31,32]. For instance, García-Foncillas et al., in a study of metastatic colorectal cancer, report that when SGAs (such as RAS mutations) of cfDNA and liver metastasis were compared, a higher concordance rate was obtained than that of cfDNA and lung metastasis [65]. This observation was explained by the higher vascularisation of liver tissue and the greater likelihood of DNA release into the circulation [66]. Furthermore, establishment of the tumour fraction can inform the most appropriate analysis method, especially in the case of alterations presenting at lower MAFs. In a study conducted in a cohort of 520 patients with metastatic prostate or breast cancer, the blood samples of 30% and 40% of breast and prostate cancer patients, respectively, had sufficient tumour fractions for standard depths of WES (i.e., ≥10%) [67]. Regarding MAFs, methods such as ddPCR and NGS with commercial panel designs for molecular profiling, a MAF cut-off value is often introduced [31]. For instance, some ddPCR and NGS platforms have cut-offs in the region of 0.040–.1% and 0.25–%, respectively [68,69], and the commercially-available Guardant360 liquid biopsy assays have almost perfect specificity for SNVs with MAFs of >2% [70]. Therefore, lower concordance is plausible when MAFs fall below the detection cut-off of the method used.
Tumour fraction can be impacted by technical practices used to extract cfDNA from plasma or serum. Guo et al., evaluate the effect of blood sample processing on cfDNA concentration and found that delaying processing beyond four hours significantly decreased detection rate of somatic mutations in cfDNA [71,72]. Release of genomic DNA from white blood cells, resulting in contamination of cfDNA, can be a consequence of delayed processing. The presence of contamination in cfDNA can, however, be accounted for by sequencing white blood cells and filtering somatic mutations attributable to clonal haematopoiesis [73], although this approach will not neutralise the diluting effect of such contamination on the tumour fraction of cfDNA. The necessity of extracting cfDNA from plasma was challenged by a study conducted by Sefrioui et al., The group compared concordance rates in a cohort of 17 mCRC patients and established 93% and 88% mutation detection rates for cfDNA isolated from plasma and crude plasma samples, respectively, suggesting that extraction of cfDNA from plasma may enhance detection by increasing tumour purity [74]. Similarly, increasing cfDNA concentration may also enhance concordance rates, by increasing the tumour fraction [75,76]. For instance, KRAS mutation status across 121 patients with NSCLC, melanoma, breast, uterine, pancreatic cancers was compared between cfDNA and matching tumours [77]. In this study, the initial concordance of 85% was improved to 95% by increasing cfDNA concentration in some discordant cases. Finally, tumour fraction can be impacted by cancer type and stage [10]. In a study of multiple cancers, the cfDNA samples of neuroblastoma patients displayed the highest tumour fraction, including two patients in which it was almost 100% [78,79].
Regarding cancer stage, Bettegwada et al., found that circulating tumour DNA, defined as the fraction of tumour DNA in total circulatory free DNA, was detectable in more than 75% of the patients with advanced stage cancers including pancreatic, ovarian, colorectal, melanoma, hepatocellular and head and neck as opposed to 50% of primary brain, renal, prostate or thyroid cancers [19]. In another study conducted by Namløs et al., on gastrointestinal stromal tumours (GIST), patients with metastatic disease displayed significantly higher frequencies of mutation detection in plasma compared to patients with localised disease which correlated with tumour burden. Specifically, all patients (n = 10) with metastatic disease had detectable mutations, while this rate was lower for patients in less advanced stages of the disease [80]. Consequently, higher tumour fraction in metastatic diseases may contribute to higher concordance rates observed between cfDNA and metastatic tumour compared to cfDNA and primary tumour [72,81], in addition to indicating poor prognosis [82]. Xie et al., tested 35 pairs of NSCLC primary tumour tissues or metastatic tumours and plasma from treatment-naïve patients using targeted sequencing for a custom panel of 56 lung cancer genes. They interrogated similarities between primary and metastatic tumours and matching cfDNA and observed 62% concordance between the trio (67/108 mutations identified). They also found that the concordance rate improved to 77.3% when they limited their analysis to driver alterations. Interestingly, they observed a higher concordance of cfDNA and metastatic profile (73.2 %) compared to cfDNA and primary tumour profiles (68.4%) [83]. Examples of higher concordance rates in metastatic and cfDNA compared to primary tumour and cfDNA, though not statistically significant, has been reported in Table 1.

3.2. Gene Type and the Effect of Drug Therapy

In a classical perspective, driver alterations, which are usually truncal and present from early stages of the disease, are SGAs that provide a selective growth advantage. Passenger alterations, which might be neutral or deleterious, are genetically linked to driver alterations [86]. Current views suggest that environmental and treatment variables lead to a more dynamic status of driver versus passenger alterations; for instance, a passenger alteration can transform into a driver alteration [87]. Driver alterations can be deemed “clinically actionable” if an FDA-approved drug or drug under investigation in a clinical trial could target the protein of interest or its downstream effectors [88]. Discordance observed between cfDNA and tumour tissue may be due to the subclonal presentation of drivers in the tumour in later stages of the disease, which can affect the detectability of these drivers in cfDNA and impact concordance rates [86]. A multitude of studies across the field of solid tumours (i.e., prostate, NSCLC, breast, neuroblastoma, renal, gastrointestinal, pancreatic, thyroid and melanoma) focus on individual (Table 2) or multiple malignancies (Table 3) and report a trend for high agreement between cfDNA and tumour tissue with respect to actionable driver alterations, but with notable exceptions [70,89,90,91,92,93,94]. In Table 2 and Table 3, we have included studies that compare the extent to which cfDNA reflects driver and actionable driver alterations of the primary or metastatic tumours.
Drug therapy may also affect the detection of driver alterations. If a particular clone is resisting therapy through clonal evolution and selection [144,145] and if therapy is stabilising the tumour and suppressing cell turnover, this could potentially affect the representation of clones in the cfDNA pool. Therefore, the selection pressure induced by therapies can lead to heterogeneity within or between tumours, which may, in turn, lead to no or variable representation of subclonal populations within the cfDNA population [5]. In a cohort of 88 NSCLC patients, Schwaederle et al., observed a trend for higher concordance rates of alterations in pre-treatment patients compared to post-treatment patients (64.7% vs. 48.9%) in a subgroup of patients with dynamic (non-flat) cfDNA profiles. Although not statistically significant, differences between the representation of alterations pre- or post-treatment were observed [112]. In a study conducted on HER2-positive metastatic breast cancer patients, fluctuations in HER2 copy number detectable in cfDNA was observed in patients undergoing multiple cycles of therapy. In this cohort of 18 patients, prior to treatment initiation, HER2 amplification was detectable in cfDNA in only 50% of patients (9/18) (despite HER2 amplification detection in tumour tissue in all cases (18/18)). In one particular patient, HER2 copies were not identified in cfDNA prior to treatment and until after cycle 2 of treatment, however, they were detected at an elevated level after cycle 4 of chemotherapy. This level increased further through disease progression after cycle 6 of chemotherapy [146]. This study further supports the effect of cancer treatment on the dynamics of cfDNA release from the tumour.

3.3. Sampling and Processing of Tumour Tissue

Due to logistic and safety limitations of taking multiple biopsies in patients [5], detection of SGAs in the peripheral blood has promising potential as a non-invasive alternative [62]. Regarding biopsy sampling intervals, minimising the collection intervals between cfDNA and tumour tissue enhances concordance rates [32,45,147]. Thompson et al., analysed EGFR mutations in advanced NSCLC patients and found that increasing timing intervals between tumour and cfDNA sampling from less than 2 weeks to more than 6 months, led to significantly lower concordance (p=0 .038) [15]. In a similar study on a cohort of 88 patients with NSCLC, the overall concordance of EGFR mutations varied depending on sampling time, with 88.2% and 64.7% concordance for time intervals of 0.8 months and >0.8 months between blood draw and tissue biopsy, respectively [112].
With regards to tumour tissue processing methods, the use of fresh frozen (FF) samples for tumour tissue instead of formalin-fixed paraffin-embedded (FFPE) marginally increased from 57.1% to 66.7%, suggesting fragmentation of DNA in FFPE processing may be significant, especially when the detection assay relies on amplicon-based amplification [72,100,148].

3.4. Detection Method

Some biomarkers are not easily detectable in the plasma of early stage cancer patients by conventional methods such as ELISA and more sensitive methods such as ddPCR or targeted sequencing may be more appropriate [107,149]. Due to the low abundance of circulating tumour DNA in plasma of early stage cancer patients, using a highly sensitive method while keeping the cost low is challenging. ddPCR assays have high sensitivity and specificity for SNV detection but may not be practical to interrogate a large scope of alterations or unknown alterations, while sequencing approaches including amplicon-based targeted sequencing may profile a broader spectrum of alterations. Demuth and colleagues compared KRAS mutation status of 28 patients with metastatic colorectal cancers between cfDNA and matched tumour samples with targeted sequencing and ddPCR, yielding a concordance rate of 79% and 89% for each method, respectively [75]. Similarly, in a study of 127 patients with advanced NSCLC, assaying for driver and drug-resistance alterations, the use of ultra-deep sequencing of cfDNA and orthogonal ddPCR was compared [89]. This study revealed almost identical findings in relation to EGFR and KRAS mutations by ultra-deep NGS and ddPCR (21/22 cases). In addition, ultra-deep sequencing identified KRAS mutations in 17 cases where tumour tissue was deemed insufficient for genotyping, suggesting that ultra-deep targeted sequencing of cfDNA may be instrumental for the identification of specific SGAs in cfDNA missed in tumour tissue. One limitation of targeted sequencing is that analysis is restricted to pre-defined genomic regions, and therefore only patients who display alterations in the analysed regions can be included [150].
Depth of sequencing may also play a key role in establishing whether SGAs detected are truncal (ancestral mutation shared by all clones) or subclonal. Chicard et al., in a study of neuroblastoma patients, detected 17 suspected relapse-specific SNVs using WES. However, upon deeper targeted sequencing of the primary tumours, these SNVs were identified in minor subclones present at diagnosis [54].
Sequencing depth is also significant in the case of detection of CNAs including chromosome-level copy number or structural changes and rearrangements [56]. In a study of multiple paediatric solid tumours, Klega et al., use ultra-low-pass WGS (ULP-WGS) with a coverage of 0.2x to 1x for detecting CNAs in cfDNA [78]. Another study demonstrated that WGS with a shallow coverage of 0.1x is sufficient for reliable analysis of CNAs [151]. This method was also successfully leveraged in metastatic prostate cancer [18]. This study revealed chromosome arm gains and losses, high level copy number gains, fusions and SNVs indicated in the pathogenesis of prostate cancer, therefore the timely and costly deep coverage WGS may be avoidable. Finally, the use of either the same analysis platform for both cfDNA and tumour or analysing all samples on both platforms may reduce the discordance rates attributed to differences in sensitivity [55].

3.5. Heterogeneity

Subsequent to fine-tuning of methods and practices for detecting technical and biological artefacts in cfDNA and tumour tissue comparative studies, the degree of contribution of tumour heterogeneity and clonal evolution to differences between matched cfDNA and tumour biopsies can be evaluated.
In a study on renal cell carcinoma, it was shown that 65% of SGAs were not detectable in every region of the primary and metastatic tumours. In addition, intratumoural heterogeneity was observed in relation to specific tumour suppressor genes [5]. These results suggest the presence of subclones, within the primary tumour that may compete or collaborate. These subclones may both evolve and expand through disease progression, leading to divergence of genomic landscapes [152], in addition to increasing adaptability to the dynamic microenvironment of the tumour [4].
The presence of spatial and temporal tumour heterogeneity has been detected in studies comparing cfDNA alterations with primary or metastatic tumours. A good example of this is a study conducted in gastroesophageal adenocarcinoma comparing the genomic profiling of primary and metastatic lesions by sequencing across multiple cohorts. This study found extensive differences in SGAs including actionable alterations between primary and metastatic tumours. One key observation was the high concordance rate of 87.5% for actionable alterations between cfDNA and metastatic tissue that were originally found to be discordant between primary and metastatic tumours. This valuable observation may implicate cfDNA in providing a representation of malignant disease, in addition to highlighting heterogeneity between alterations of primary and metastatic tumours [21,153,154], although high agreement between primary and metastatic tumour SGAs has been reported in other studies [155,156].

4. The future of cfDNA in precision oncology

Precision oncology applies tailored treatment to individual characteristics of patients by detecting and monitoring actionable alterations to inform targeted therapy and patient management strategies. The tumour biopsy remains the most efficient diagnostic tool at present but due to the impracticality of obtaining multiple tumour samples to capture a larger scope of spatial and temporal heterogeneity [157], the use of cfDNA has emerged as a viable alternative [158]. The emergence of cfDNA as a clinically relevant, minimally invasive tool to inform disease burden, acquisition of actionable alterations and resistance to therapy has been extensively documented [20,28,42]. Despite these promising prospects, the extent to which cfDNA captures and reflects the SGAs of the tumour and its metastases is not fully dissected. Furthermore, cfDNA as a diagnostic tool poses limitations; for instance, cfDNA cannot replace histologic information obtained from tissue biopsy and the dynamics of cfDNA release may lead to variable representation of important actionable alterations in the cfDNA population [60,61,62,63,64]. Considering these limitations, many studies suggest a “companion role” for cfDNA in the clinical diagnostic setting, in which tumour tissue and cfDNA samples could be considered in parallel to improve the likelihood of early detection of actionable alterations in patients. This approach would provide a window of opportunity for early detection and initiation of targeted therapy, especially in cases where actionable alterations are detected in cfDNA and not tumour tissue due to sampling bias inherent in tissue biopsies [15,36,54,73,89,159]. When tumour biopsies cannot be obtained safely or tumour tissue is not available, plasma sampling could provide valuable information for clinical decision-making.
In addition to the utility of cfDNA as a diagnostic tool, clinical resistance to therapy can be longitudinally monitored in time by analysing serial cfDNA samples [20,160]. Resistance to therapy can emerge from the acquisition of SGAs in genes and pathways targeted by therapy. Understanding the mechanism of resistance by analysis of tumour tissue is challenging due to safety issues of serial tissue biopsies. Hence, the serial mutation profiling based on cfDNA over the duration of the diseas, may permit the real-time appreciation of the efficacy of systemic therapy and detection of disease resistance. In multiple studies, cfDNA has been utilised for monitoring of resistance mutations including EGFR T790M, BRAF, ALK, ERBB2 amplification in NSCLC patients receiving therapy [15,37,41,73,109,128,161,162]. In a study by Sung et al., longitudinal cfDNA analysis lead to the detection of EGFR T790M mutation emergence in 28.6% of NSCLC patients receiving EGFR TKI treatment [36]. Also longitudinal cfDNA analysis has been used in colorectal cancer patients for monitoring of cetuximab resistance through acquisition of secondary KRAS mutations [139] and also resistance to antiangiogenic therapies [163].

5. Conclusions

In conclusion, the importance of this study was the review of the feasibility of using cfDNA for detecting a range of SGAs of tumours across the broad field of solid tumours. Further, concordance rates of actionable driver alterations across solid tumours were examined. Despite some inconsistencies, a trend of high concordance rates for these alterations detected in plasma and tumour tissue was observed. However, due to factors such as clonality and treatment that may affect these rates, we would suggest for each study to be considered in the specific context of cycle of treatment, method, cancer type and stage. Actionable alteration status is critical to targeted therapy decision-making and monitoring treatment response and the promising prospect of leveraging plasma cfDNA for detecting and monitoring these alterations is clinically relevant. In addition, this study drew examples from the literature to interrogate the technical challenges that impact agreement rates between tumours and cfDNA. Fine-tuning of methods and practices is warranted to confidently dissect and distinguish heterogeneity from artefacts. As more sensitive and affordable sequencing technologies become available, deep sequencing of cfDNA can provide insight into tumour evolution and monitoring treatment resistance in several cancers [159].

Author Contributions

L.J. designed the study; L.J. and T.H. collected data and wrote the manuscript.

Funding

This research received no external funding.

Acknowledgments

We would like to thank Ricky Trigg at Functional Genomics Unit at GlaxoSmithKline, Stevenage, UK (previously at the Department of Pathology, Cambridge University) for scientific editing of this manuscript.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Beroukhim, R.; Mermel, C.H.; Porter, D.; Wei, G.; Raychaudhuri, S.; Donovan, J.; Barretina, J.; Boehm, J.S.; Dobson, J.; Urashima, M.; et al. The landscape of somatic copy-number alteration across human cancers. Nature 2010, 463, 899–905. [Google Scholar] [CrossRef] [PubMed]
  2. Mitelman, F.; Johansson, B.; Mertens, F. The impact of translocations and gene fusions on cancer causation. Nat. Rev. Cancer 2007, 7, 233–245. [Google Scholar] [CrossRef] [PubMed]
  3. Nussinov, R.; Jang, H.; Tsai, C.J.; Cheng, F. Review: Precision medicine and driver mutations: Computational methods, functional assays and conformational principles for interpreting cancer drivers. PLoS Comput. Biol. 2019, 15, e1006658. [Google Scholar]
  4. Fisher, R.; Pusztai, L.; Swanton, C. Cancer heterogeneity: Implications for targeted therapeutics. Br. J. Cancer 2013, 108, 479–485. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Gerlinger, M.; Rowan, A.J.; Horswell, S.; Math, M.; Larkin, J.; Endesfelder, D.; Gronroos, E.; Martinez, P.; Matthews, N.; Stewart, A.; et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N. Engl. J. Med. 2012, 366, 883–892. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Hiley, C.; de Bruin, E.C.; McGranahan, N.; Swanton, C. Deciphering intratumor heterogeneity and temporal acquisition of driver events to refine precision medicine. Genome Biol. 2014, 15, 453. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Shapiro, B.; Chakrabarty, M.; Cohn, E.M.; Leon, S.A. Determination of circulating DNA levels in patients with benign or malignant gastrointestinal disease. Cancer 1983, 51, 2116–2120. [Google Scholar] [CrossRef]
  8. Chen, X.; Bonnefoi, H.; Diebold-Berger, S.; Lyautey, J.; Lederrey, C.; Faltin-Traub, E.; Stroun, M.; Anker, P. Detecting tumor-related alterations in plasma or serum DNA of patients diagnosed with breast cancer. Clin. Cancer Res. 1999, 5, 2297–2303. [Google Scholar] [PubMed]
  9. Riviere, P.; Fanta, P.T.; Ikeda, S.; Baumgartner, J.; Heestand, G.M.; Kurzrock, R. The Mutational Landscape of Gastrointestinal Malignancies as Reflected by Circulating Tumor DNA. Mol. Cancer Ther. 2018, 17, 297–305. [Google Scholar] [CrossRef] [Green Version]
  10. Wyatt, A.W.; Annala, M.; Aggarwal, R.; Beja, K.; Feng, F.; Youngren, J.; Foye, A.; Lloyd, P.; Nykter, M.; Beer, T.M.; et al. Concordance of Circulating Tumor DNA and Matched Metastatic Tissue Biopsy in Prostate Cancer. J. Natl. Cancer Inst. 2017, 109. [Google Scholar] [CrossRef] [Green Version]
  11. Kumar, A.; Coleman, I.; Morrissey, C.; Zhang, X.; True, L.D.; Gulati, R.; Etzioni, R.; Bolouri, H.; Montgomery, B.; White, T.; et al. Substantial interindividual and limited intraindividual genomic diversity among tumors from men with metastatic prostate cancer. Nat. Med. 2016, 22, 369–378. [Google Scholar] [CrossRef] [PubMed]
  12. Muluhngwi, P.; Valdes, R., Jr.; Fernandez-Botran, R.; Burton, E.; Williams, B.; Linder, M.W. Cell-free DNA diagnostics: Current and emerging applications in oncology. Pharmacogenomics 2019, 20, 357–380. [Google Scholar] [CrossRef] [PubMed]
  13. Sozzi, G.; Conte, D.; Mariani, L.; Lo Vullo, S.; Roz, L.; Lombardo, C.; Pierotti, M.A.; Tavecchio, L. Analysis of circulating tumor DNA in plasma at diagnosis and during follow-up of lung cancer patients. Cancer Res. 2001, 61, 4675–4678. [Google Scholar] [PubMed]
  14. Leon, S.A.; Shapiro, B.; Sklaroff, D.M.; Yaros, M.J. Free DNA in the serum of cancer patients and the effect of therapy. Cancer Res. 1997, 37, 646–650. [Google Scholar]
  15. Thompson, J.C.; Yee, S.S.; Troxel, A.B.; Savitch, S.L.; Fan, R.; Balli, D.; Lieberman, D.B.; Morrissette, J.D.; Evans, T.L.; Bauml, J.; et al. Detection of Therapeutically Targetable Driver and Resistance Mutations in Lung Cancer Patients by Next-Generation Sequencing of Cell-Free Circulating Tumor DNA. Clin. Cancer Res. 2016, 22, 5772–5782. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Combaret, V.; Audoynaud, C.; Iacono, I.; Favrot, M.C.; Schell, M.; Bergeron, C.; Puisieux, A. Circulating MYCN DNA as a tumor-specific marker in neuroblastoma patients. Cancer Res. 2002, 62, 3646–3648. [Google Scholar]
  17. Cristofanilli, M.; Hayes, D.F.; Budd, G.T.; Ellis, M.J.; Stopeck, A.; Reuben, J.M.; Doyle, G.V.; Matera, J.; Allard, W.J.; Miller, M.C.; et al. Circulating tumor cells: A novel prognostic factor for newly diagnosed metastatic breast cancer. J. Clin. Oncol. 2005, 23, 1420–1430. [Google Scholar] [CrossRef]
  18. Heitzer, E.; Ulz, P.; Belic, J.; Gutschi, S.; Quehenberger, F.; Fischereder, K.; Benezeder, T.; Auer, M.; Pischler, C.; Mannweiler, S.; et al. Tumor-associated copy number changes in the circulation of patients with prostate cancer identified through whole-genome sequencing. Genome Med. 2013, 5, 30. [Google Scholar] [CrossRef] [Green Version]
  19. Bettegowda, C.; Sausen, M.; Leary, R.J.; Kinde, I.; Wang, Y.; Agrawal, N.; Bartlett, B.R.; Wang, H.; Luber, B.; Alani, R.M.; et al. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci. Transl. Med 2014, 6, 224ra24. [Google Scholar] [CrossRef] [Green Version]
  20. Dawson, S.J.; Tsui, D.W.; Murtaza, M.; Biggs, H.; Rueda, O.M.; Chin, S.F.; Dunning, M.J.; Gale, D.; Forshew, T.; Mahler-Araujo, B.; et al. Analysis of circulating tumor DNA to monitor metastatic breast cancer. N. Engl. J. Med. 2013, 368, 1199–1209. [Google Scholar] [CrossRef] [Green Version]
  21. Diaz, L.A. Jr.; Bardelli, A. Liquid biopsies: Genotyping circulating tumor DNA. J. Clin. Oncol. 2014, 32, 579–586. [Google Scholar] [CrossRef] [PubMed]
  22. Gray, E.S.; Rizos, H.; Reid, A.L.; Boyd, S.C.; Pereira, M.R.; Lo, J.; Tembe, V.; Freeman, J.; Lee, J.H.; Scolyer, R.A.; et al. Circulating tumor DNA to monitor treatment response and detect acquired resistance in patients with metastatic melanoma. Oncotarget 2015, 6, 42008–42018. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Newman, A.M.; Bratman, S.V.; To, J.; Wynne, J.F.; Eclov, N.C.; Modlin, L.A.; Liu, C.L.; Neal, J.W.; Wakelee, H.A.; Merritt, R.E.; et al. An ultrasensitive method for quantitating circulating tumor DNA with broad patient coverage. Nat. Med. 2014, 20, 54–548. [Google Scholar] [CrossRef] [PubMed]
  24. De Mattos-Arruda, L.; Caldas, C. Cell-free circulating tumour DNA as a liquid biopsy in breast cancer. Mol. Oncol. 2016, 10, 464–474. [Google Scholar] [CrossRef] [PubMed]
  25. Fleischhacker, M.; Schmidt, B. Circulating nucleic acids (CNAs) and cancer—a survey. Biochim. Biophys. Acta 2007, 1775, 123–181. [Google Scholar] [CrossRef]
  26. Heitzer, E.; Ulz, P.; Geigl, J.B. Circulating tumor DNA as a liquid biopasy for cancer. Clin. Chem. 2015, 61, 23–112. [Google Scholar] [CrossRef]
  27. Montagut, C.; Siravegna, G.; Bardelli, A. Liquid biopsies to evaluate early therapeutic response in colorectal cancer. Ann. Oncol. 2015, 26, 1525–1527. [Google Scholar] [CrossRef]
  28. Murtaza, M.; Dawson, S.J.; Tsui, D.W.; Gale, D.; Forshew, T.; Piskorz, A.M.; Parkinson, C.; Chin, S.F.; Kingsbury, Z.; Wong, A.S.; et al. Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA. Nature 2013, 497, 108–112. [Google Scholar] [CrossRef]
  29. Luke, J.J.; Oxnard, G.R.; Paweletz, C.P.; Camidge, D.R.; Heymach, J.V.; Solit, D.B.; Johnson, B.E.; Cell Free DNA Working Group. Realizing the potential of plasma genotyping in an age of genotypedirected therapies. J. Natl. Cancer Inst. 2014, 106, dju214. [Google Scholar] [CrossRef] [Green Version]
  30. Bokemeyer, C.; Köhne, C.H.; Ciardiello, F.; Lenz, H.J.; Heinemann, V.; Klinkhardt, U.; Beier, F.; Duecker, K.; Krieken van, J.H.; Tejpar, S. FOLFOX4 plus cetuximab treatment and RAS mutations in colorectal cancer. Eur. J. Cancer 2015, 51, 1243–1252. [Google Scholar] [CrossRef]
  31. Grasselli, J.; Elez, E.; Caratù, G.; Matito, J.; Santos, C.; Macarulla, T.; Vidal, J.; Garcia, M.; Viéitez, J.M.; Paéz, D.; et al. Concordance of blood- and tumor-based detection of RAS mutations to guide anti-EGFR therapy in metastatic colorectal cancer. Ann. Oncol. 2017, 28, 1294–1301. [Google Scholar] [CrossRef] [PubMed]
  32. Bando, H.; Kagawa, Y.; Kato, T.; Akagi, K.; Denda, T.; Nishina, T.; Komatsu, Y.; Oki, E.; Kudo, T.; Kumamoto, H.; et al. A multicentre, prospective study of plasma circulating tumour DNA test for detecting RAS mutation in patients with metastatic colorectal cancer. Br. J. Cancer 2019, 120, 982–986. [Google Scholar] [CrossRef] [PubMed]
  33. Schmiegel, W.; Scott, R.J.; Dooley, S.; Lewis, W.; Meldrum, C.J.; Pockney, P.; Draganic, B.; Smith, S.; Hewitt, C.; Philimore, H.; 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 tissuebased RAS testing. Mol. Oncol. 2017, 11, 208–219. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Ettinger, D.S.; Akerley, W.; Borghaei, H.; Chang, A.C.; Cheney, R.T.; Chirieac, L.R.; D’Amico, T.A.; Demmy, T.L.; Govindan, R.; Grannis, F.W.; et al. Nonsmall cell lung cancer, version 2.2013. J. Natl. Compr. Cancer Netw. 2013, 11, 645–653. [Google Scholar] [CrossRef] [Green Version]
  35. Oxnard, G.R.; Binder, A.J.P.A. New targetable oncogenes in non-small-cell lung cancer. J. Clin. Oncol. 2013, 31, 1097–1104. [Google Scholar] [CrossRef] [Green Version]
  36. Sung, J.S.; Chong, H.Y.; Kwon, N.J.; Kim, H.M.; Lee, J.W.; Kim, B.; Lee, S.B.; Park, C.W.; Choi, J.Y.; Chang, W.J.; et al. Detection of somatic variants and EGFR mutations in cell-free DNA from non-small cell lung cancer patients by ultra-deep sequencing using the ion ampliseq cancer hotspot panel and droplet digital polymerase chain reaction. Oncotarget 2017, 8, 106901. [Google Scholar] [CrossRef] [Green Version]
  37. Lee, J.Y.; Qing, X.; Xiumin, W.; Yali, B.; Chi, S.; Bak, S.H.; Lee, H.Y.; Sun, J.M.; Lee, S.H.; Ahn, J.S.; et al. Longitudinal monitoring of EGFR mutations in plasma predicts outcomes of NSCLC patients treated with EGFR TKIs: Korean Lung Cancer Consortium (KLCC-12-02). Oncotarget 2016, 7, 6984–6993. [Google Scholar] [CrossRef] [Green Version]
  38. Stewart, E.L.; Tan, S.Z.; Liu, G.T.M.S. Known and putative mechanisms of resistance to EGFR targeted therapies in NSCLC patients with EGFR mutations-a review. Transl. Lung Cancer Res. 2015, 4, 67–81. [Google Scholar]
  39. Oxnard, G.R.; Paweletz, C.P.; Kuang, Y.; Mach, S.L.; O’Connell, A.; Messineo, M.M.; Luke, J.J.; Butaney, M.; Kirschmeier, P.J.D.M. Noninvasive detection of response and resistance in EGFR-mutant lung cancer using quantitative next-generation genotyping of cell-free plasma DNA. Clin. Cancer Res. Res. 2014, 20, 1698–1705. [Google Scholar] [CrossRef] [Green Version]
  40. Yung, T.K.; Chan, K.C.; Mok, T.S.; Tong, J.; To, K.F.; Lo, Y.M. Single-molecule detection of epidermal growth factor receptor mutations in plasma by microfluidics digital PCR in non-small cell lung cancer patients. Clin. Cancer Res. 2009, 15, 2076–2084. [Google Scholar] [CrossRef] [Green Version]
  41. Sakai, K.; Horiike, A.; Irwin, D.L.; Kudo, K.; Fujita, Y.; Tanimoto, A.; Sakatani, T.; Saito, R.; Kaburaki, K.; Yanagitani, N.; et al. Detection of epidermal growth factor receptor T790M mutation in plasma DNA from patients refractory to epidermal growth factor receptor tyrosine kinase inhibitor. Cancer Sci. 2013, 104, 1198–1204. [Google Scholar] [CrossRef] [PubMed]
  42. Cristofanilli, M.; Budd, G.T.; Ellis, M.J.; Stopeck, A.; Matera, J.; Miller, M.C.; Reuben, J.M.; Doyle, G.V.; Allard, W.J.; Terstappen, L.W.; et al. Circulating tumor cells, disease progression, and survival in metastatic breast cancer. N. Engl. J. Med. 2004, 351, 781–791. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Huang, Z.H.; Li, L.H.; Hua, D. Quantitative analysis of plasma circulating DNA at diagnosis and during follow-up of breast cancer patients. Cancer Lett. 2006, 243, 64–70. [Google Scholar] [CrossRef] [PubMed]
  44. Takeshita, T.; Yamamoto, Y.; Yamamoto-Ibusuki, M.; Tomiguchi, M.; Sueta, A.; Murakami, K.; Omoto, Y.; Iwase, H. Comparison of ESR1 Mutations in Tumor Tissue and Matched Plasma Samples from Metastatic Breast Cancer Patients. Transl. Oncol. 2017, 10, 766–771. [Google Scholar] [CrossRef] [PubMed]
  45. Higgins, M.J.; Jelovac, D.; Barnathan, E.; Blair, B.; Slater, S.; Powers, P.; Zorzi, J.; Jeter, S.C.; Oliver, G.R.; Fetting, J.; et al. Detection of tumor PIK3CA status in metastatic breast cancer using peripheral blood. Clin. Cancer Res. 2012, 18, 3462–3469. [Google Scholar] [CrossRef] [Green Version]
  46. Board, R.E.; Wardley, A.M.; Dixon, J.M.; Armstrong, A.C.; Howell, S.; Renshaw, L.; Donald, E.; Greystoke, A.; Ranson, M.; Hughes, A.; et al. Detection of PIK3CA mutations in circulating free DNA in patients with breast cancer. Breast Cancer Res. Treat. 2010, 120, 461–467. [Google Scholar] [CrossRef]
  47. Garcia-Saenz, J.A.; Ayllon, P.; Laig, M.; Acosta-Eyzaguirre, D.; Garcia-Esquinas, M.; Montes, M.; Sanz, J.; Barquín, M.; Moreno, F.; Garcia-Barberan, V.; et al. Tumor burden monitoring using cell-free tumor DNA could be limited by tumor heterogeneity in advanced breast cancer and should be evaluated together with radiographic imaging. BMC Cancer 2017, 17, 210. [Google Scholar] [CrossRef] [Green Version]
  48. Beaver, J.A.; Jelovac, D.; Balukrishna, S.; Cochran, R.; Croessmann, S.; Zabransky, D.J.; Wong, H.Y.; Toro, P.V.; Cidado, J.; Blair, B.G.; et al. Detection of cancer DNA in plasma of patients with early-stage breast cancer. Clin. Cancer Res. 2014, 20, 2643–2650. [Google Scholar] [CrossRef] [Green Version]
  49. Kodahl, A.R.; Ehmsen, S.; Pallisgaard, N.; Jylling, A.M.B.; Jensen, J.D.; Laenkholm, A.V.; Knoop, A.S.; Ditzel, H.J. Correlation between circulating cell-free PIK3CA tumor DNA levels and treatment response in patients with PIK3CA-mutated metastatic breast cancer. Mol. Oncol. 2018, 12, 925–935. [Google Scholar] [CrossRef] [Green Version]
  50. Di Cataldo, A.; Dau, D.; Conte, M.; Parodi, S.; De Bernardi, B.; Giuliano, M.; Pession, A.; Viscardi, E.; Luksch, R.; Castellano, A.; et al. Diagnostic and prognostic markers in infants with disseminated neuroblastoma: A retrospective analysis from the Italian Cooperative Group for Neuroblastoma. Med. Sci. Monit. 2009, 1, MT11–MT18. [Google Scholar]
  51. Pession, A.; De Bernardi, B.; Perri, P.; Mazzocco, K.; Rondelli, R.; Nigro, M.; Lolascon, A.; Forni, M.; Basso, G.; Conte, M.; et al. The prognostic effect of amplification of MYCN oncogene in neuroblstoma. The preliminary results of the Italian Copperative Group for Neuroblastoma (GCINB). Pediatr. Med. Chir. 1994, 16, 211–218. [Google Scholar] [PubMed]
  52. Combaret, V.; Iacono, I.; Bellini, A.; Bréjon, S.; Bernard, V.; Marabelle, A.; Coze, C.; Pierron, G.; Lapouble, E.; Schleiermacher, G.; et al. Detection of tumor ALK status in neuroblastoma patients using peripheral blood. Cancer Med. 2015, 4, 540–550. [Google Scholar] [CrossRef] [PubMed]
  53. Chicard, M.; Boyault, S.; Colmet Daage, L.; Richer, W.; Gentien, D.; Pierron, G.; Lapouble, E.; Bellini, A.; Clement, N.; Iacono, I.; et al. Genomic Copy Number Profiling Using Circulating Free Tumor DNA Highlights Heterogeneity in Neuroblastoma. Clin. Cancer Res. 2016, 22, 5564–5573. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Chicard, M.; Colmet-Daage, L.; Clement, N.; Danzon, A.; Bohec, M.; Bernard, V.; Baulande, S.; Bellini, A.; Deveau, P.; Pierron, G.; et al. Whole-Exome Sequencing of Cell-Free DNA Reveals Temporo-spatial Heterogeneity and Identifies Treatment-Resistant Clones in Neuroblastoma. Clin. Cancer Res. 2018, 24, 939–949. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Van Roy, N.; Van Der Linden, M.; Menten, B.; Dheedene, A.; Vandeputte, C.; Van Dorpe, J.; Laureys, G.; Renard, M.; Sante, T.; Lammens, T.; et al. Shallow Whole Genome Sequencing on Circulating Cell-Free DNA Allows Reliable Noninvasive Copy-Number Profiling in Neuroblastoma Patients. Clin. Cancer Res. 2017, 23, 6305–6314. [Google Scholar] [CrossRef] [Green Version]
  56. Leary, R.J.; Sausen, M.; Kinde, I.; Papadopoulos, N.; Carpten, J.D.; Craig, D.; O’Shaughnessy, J.; Kinzler, K.W.; Parmigiani, G.; Vogelstein, B.; et al. Detection of chromosomal alterations in the circulation of cancer patients with whole-genome sequencing. Sci. Transl. Med. 2012, 1, 162ra154. [Google Scholar] [CrossRef] [Green Version]
  57. Lavon, I.; Refael, M.; Zelikovitch, B.; Shalom, E.S. Serum DNA can define tumor-specific genetic and epigenetic markers in gliomas of various grades. Neuro. Oncol. 2010, 12, 173–180. [Google Scholar] [CrossRef] [Green Version]
  58. Molparia, B.; Oliveira, G.; Wagner, J.L.; Spencer, E.G.; Torkamani, A. A feasibility study of colorectal cancer diagnosis via circulating tumor DNA derived CNV detection. PLoS. ONE 2018, 13, e0196826. [Google Scholar] [CrossRef]
  59. Campbell, P.J.; Stephens, P.J.; Pleasance, E.D.; O’Meara, S.; Li, H.; Santarius, T.; Stebbings, L.A.; Leroy, C.; Edkins, S.; Hardy, C.; et al. Identification of somatically acquired rearrangements in cancer using genome-wide massively parallel paired-end sequencing. Nat. Genet. 2008, 40, 722–729. [Google Scholar] [CrossRef]
  60. Stroun, M.; Lyautey, J.; Lederrey, C.; Olson-Sand, A.; Anker, P. About the possible origin and mechanism of circulating DNA apoptosis and active DNA release. Clin. Chim. Acta 2001, 313, 139–142. [Google Scholar] [CrossRef]
  61. Snyder, M.W.; Kircher, M.; Hill, A.J.; Daza, R.M.; Shendure, J. Cell-free DNA comprises an in vivo nucleosome footprint that informs its tissues-of-origin. Cell 2016, 164, 57–68. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Jahr, S.; Hentze, H.; Englisch, S.; Hardt, D.; Fackelmayer, F.O.; Hesch, R.D.; Knippers, R. DNA fragments in the blood plasma of cancer patients: Quantitations and evidence for their origin from apoptotic and necrotic cells. Cancer Res. 2001, 61, 1659–1665. [Google Scholar] [PubMed]
  63. Ignatiadis, M.; Dawson, S.J. Circulating tumor cells and circulating tumor DNA for precision medicine: Dream or reality? Ann. Oncol. 2014, 25, 2304–2313. [Google Scholar] [CrossRef] [PubMed]
  64. Aran, D.; Sirota, M.; Butte, A.J. Systematic pan-cancer analysis of tumour purity. Nat. Commun. 2015, 6, 8971. [Google Scholar] [CrossRef] [Green Version]
  65. García-Foncillas, J.; Tabernero, J.; Élez, E.; Aranda, E.; Benavides, M.; Camps, C.; Jantus-Lewintre, E.; López, R.; Muinelo-Romay, L.; Montagut, C.; et al. Prospective multicenter real-world RAS mutation comparison between OncoBEAM-based liquid biopsy and tissue analysis in metastatic colorectal cancer. Br. J. Cancer 2018, 119, 1464–1470. [Google Scholar] [CrossRef]
  66. Thierry, A.R.; El Messaoudi, S.; Mollevi, C.; Raoul, J.L.; Guimbaud, R.; Pezet, D.; Artru, P.; Assenat, E.; Borg, C.; Mathonnet, M.; 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, 2149–2159. [Google Scholar] [CrossRef]
  67. Adalsteinsson, V.A.; Ha, G.; Freeman, S.S.; Choudhury, A.D.; Stover, D.G.; Parsons, H.A.; Gydush, G.; Reed, S.C.; Rotem, D.; Rhoades, J.; et al. Scalable whole-exome sequencing of cell-free DNA reveals high concordance with metastatic tumors. Nat. Commun. 2017, 8, 1324. [Google Scholar] [CrossRef] [Green Version]
  68. Yang, X.; Zhuo, M.; Ye, X.; Bai, H.; Wang, Z.; Sun, Y.; Zhao, J.; An, T.; Duan, J.; Wu, M.; et al. Quantification of mutant alleles in circulating tumor DNA can predict survival in lung cancer. Oncotarget 2016, 7, 20810. [Google Scholar] [CrossRef] [Green Version]
  69. Bartels, S.; Persing, S.; Hasemeier, B.; Schipper, E.; Kreipe, H.; Lehmann, U. Molecular Analysis of Circulating Cell-Free DNA from Lung Cancer Patients in Routine Laboratory Practice: A Cross-Platform Comparison of Three Different Molecular Methods for Mutation Detection. J. Mol. Diagn. 2017, 19, 722–723. [Google Scholar] [CrossRef] [Green Version]
  70. Chae, Y.K.; Davis, A.A.; Jain, S.; Santa-Maria, C.; Flaum, L.; Beaubier, N.; Platanias, L.C.; Gradishar, W.; Giles, F.J.; Cristofanilli, M. Concordance of genomic alterations by next-generation sequencing in tumor tissue versus circulating tumor DNA in breast cancer. Mol. Cancer Ther. 2017, 16, 1412–1420. [Google Scholar] [CrossRef] [Green Version]
  71. Jen, J.; Wu, L.; Sidransky, D. An overview on the isolation and analysis of circulating tumor DNA in plasma and serum. Ann. N. Y. Acad. Sci. 2000, 906, 8–12. [Google Scholar] [CrossRef] [PubMed]
  72. Guo, Q.; Wang, J.; Xiao, J.; Wang, L.; Hu, X.; Yu, W.; Song, G.; Lou, J.; Chen, J. Heterogeneous mutation pattern in tumor tissue and circulating tumor DNA warrants parallel NGS panel testing. Mol. Cancer 2018, 17, 131. [Google Scholar] [CrossRef] [PubMed]
  73. Li, B.T.; Janku, F.; Jung, B.; Hou, C.; Madwani, K.; Alden, R.; Razavi, P.; Reis-Filho, J.S.; Shen, R.; Isbell, J.M.; et al. Ultra-deep next-generation sequencing of plasma cell-free DNA in patients with advanced lung cancers: Results from the Actionable Genome Consortium. Ann. Oncol. 2019, 30, 597–603. [Google Scholar] [CrossRef] [PubMed]
  74. Sefrioui, D.; Beaussire, L.; Perdrix, A.; Clatot, F.; Michel, P.; Frebourg, T.; Di Fiore, F.; Sarafan-Vasseur, N. Direct circulating tumor DNA detection from unpurified plasma using a digital PCR platform. Clin. BioChem. 2017, 50, 963–966. [Google Scholar] [CrossRef] [PubMed]
  75. Demuth, C.; Spindler, K.G.; Johansen, J.S.; Pallisgaard, N.; Nielsen, D.; Hogdall, E.; Vittrup, B.; Sorensen, B.S. Measuring KRAS Mutations in Circulating Tumor DNA by Droplet Digital PCR and Next-Generation Sequencing. Transl. Oncol. 2018, 11, 1220–1224. [Google Scholar] [CrossRef] [PubMed]
  76. Jovelet, C.; Ileana, E.; Le Deley, M.C.; Motté, N.; Rosellini, S.; Romero, A.; Lefebvre, C.; Pedrero, M.; Pata-Merci, N.; Droin, N.; et al. Circulating Cell-Free Tumor DNA Analysis of 50 Genes by Next-Generation Sequencing in the Prospective MOSCATO Trial. Clin. Cancer Res. 2016, 22, 2960. [Google Scholar] [CrossRef] [Green Version]
  77. Janku, F.; Huang, H.J.; Fujii, T.; Shelton, D.N.; Madwani, K.; Fu, S.; Tsimberidou, A.M.; Piha-Paul, S.A.; Wheler, J.J.; Zinner, R.G.; et al. Multiplex KRASG12/G13 mutation testing of unamplified cell-free DNA from the plasma of patients with advanced cancers using droplet digital polymerase chain reaction. Ann. Oncol. 2017, 28, 642–650. [Google Scholar]
  78. Klega, K.; Imamovic-Tuco, A.; Ha, G.; Clapp, A.N.; Meyer, S.; Ward, A.; Clinton, C.; Nag, A.; Van Allen, E.; Mullen, E.; et al. Detection of Somatic Structural Variants Enables Quantification and Characterization of Circulating Tumor DNA in Children With Solid Tumors. JCO Precis. Oncol. 2018, 2, 1–3. [Google Scholar] [CrossRef]
  79. Hemming, M.L.; Klega, K.S.; Rhoades, J.; Ha, G.; Acker, K.E.; Andersen, J.L.; Thai, E.; Nag, A.; Thorner, A.R.; Raut, C.P.; et al. Detection of Circulating Tumor DNA in Patients With Leiomyosarcoma With Progressive Disease. JCO Precis. Oncol. 2019, 3, 1–11. [Google Scholar] [CrossRef]
  80. Namløs, H.M.; Boye, K.; Mishkin, S.J.; Barøy, T.; Lorenz, S.; Bjerkehagen, B.; Stratford, E.W.; Munthe, E.; Kudlow, B.A.; Myklebost, O.; et al. Noninvasive Detection of ctDNA Reveals Intratumor Heterogeneity and Is Associated with Tumor Burden in Gastrointestinal Stromal Tumor. Mol. Cancer Ther. 2018, 17, 2473–2480. [Google Scholar] [CrossRef] [Green Version]
  81. Liu, L.; Liu, H.; Shao, D.; Liu, Z.; Wang, J.; Deng, Q.; Tang, H.; Yang, H.; Zhang, Y.; Qiu, Y.; et al. Development and clinical validation of a circulating tumor DNA test for the identification of clinically actionable mutations in nonsmall cell lung cancer. Genes Chromosom. Cancer 2018, 57, 211–220. [Google Scholar] [CrossRef] [PubMed]
  82. Kumar, S.; Guleria, R.; Singh, V.; Bharti, A.C.; Mohan, A.; Das, B.C. Plasma DNA level in predicting therapeutic efficacy in advanced nonsmall cell lung cancer. Eur. Respir. J. 2010, 36, 885–892. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Xie, F.; Zhang, Y.; Mao, X.; Zheng, X.; Han-Zhang, H.; Ye, J.; Zhao, R.; Zhang, X.; Sun, J. Comparison of genetic profiles among primary lung tumor, metastatic lymph nodes and circulating tumor DNA in treatment-naïve advanced nonsquamous non-small cell lung cancer patients. Lung Cancer 2018, 121, 54–60. [Google Scholar] [CrossRef] [PubMed]
  84. Tzanikou, E.; Markou, A.; Politaki, E.; Koutsopoulos, A.; Psyrri, A.; Mavroudis, D.; Georgoulias, V.; Lianidou, E. PIK3CA hotspot mutations in circulating tumor cells and paired circulating tumor DNA in breast cancer: A direct comparison study. Mol. Oncol. 2019. [Google Scholar] [CrossRef] [Green Version]
  85. Kato, S.; Okamura, R.; Baumgartner, J.M.; Patel, H.; Leichman, L.; Kelly, K.; Sicklick, J.K.; Fanta, P.T.; Lippman, S.M.; Kurzrock, R. Analysis of Circulating Tumor DNA and Clinical Correlates in Patients with Esophageal, Gastroesophageal Junction, and Gastric Adenocarcinoma. Clin. Cancer Res. 2018, 24, 6248–6256. [Google Scholar] [CrossRef] [Green Version]
  86. McGranahan, N.; Favero, F.; de Bruin, E.C.; Birkbak, N.J.; Szallasi, Z.; Swanton, C. Clonal status of actionable driver events and the timing of mutational processes in cancer evolution. Sci. Transl. Med. 2015, 7, 283ra54. [Google Scholar] [CrossRef] [Green Version]
  87. Yap, T.A.; Gerlinger, M.; Futreal, P.A.; Pusztai, L.; Swanton, C. Intratumor heterogeneity: Seeing the wood for the trees. Sci. Transl. Med. 2012, 4, 127ps10. [Google Scholar] [CrossRef] [Green Version]
  88. Stratton, M.; Campbell, P.; Futreal, P. The cancer genome. Nature 2009, 458, 719–724. [Google Scholar] [CrossRef] [Green Version]
  89. Toor, O.M.; Ahmed, Z.; Bahaj, W.; Boda, U.; Cummings, L.S.; McNally, M.E.; Kennedy, K.F.; Pluard, T.J.; Hussain, A.; Subramanian, J.; et al. Correlation of Somatic Genomic Alterations Between Tissue Genomics and ctDNA Employing Next-Generation Sequencing: Analysis of Lung and Gastrointestinal Cancers. Mol. Cancer Ther. 2018, 17, 1123–1132. [Google Scholar] [CrossRef] [Green Version]
  90. Hahn, A.W.; Gill, D.M.; Maughan, B.; Agarwal, A.; Arjyal, L.; Gupta, S.; Streeter, J.; Bailey, E.; Pal, S.K.; Agarwal, N. Correlation of genomic alterations assessed by next-generation sequencing (NGS) of tumor tissue DNA and circulating tumor DNA (ctDNA) in metastatic renal cell carcinoma (mRCC): Potential clinical implications. Oncotarget 2017, 8, 33614–33620. [Google Scholar] [CrossRef] [Green Version]
  91. Kuderer, N.M.; Burton, K.A.; Blau, S.; Rose, A.L.; Parker, S.; Lyman, G.H.; Blau, C.A. Comparison of 2 commercially available next-generation sequencing platforms in oncology. JAMA Oncol. 2017, 3, 996–998. [Google Scholar] [CrossRef] [PubMed]
  92. Barata, P.C.; Koshkin, V.S.; Funchain, P.; Sohal, D.; Pritchard, A.; Klek, S.; Adamowicz, T.; Gopalakrishnan, D.; Garcia, J.; Rini, B.; et al. Next-generation sequencing (NGS) of cell-free circulating tumor DNA and tumor tissue in patients with advanced urothelial cancer: A pilot assessment of concordance. Ann. Oncol. 2017, 28, 2458–2463. [Google Scholar] [CrossRef] [PubMed]
  93. Cohen, J.D.; Javed, A.A.; Thoburn, C.; Wong, F.; Tie, J.; Gibbs, P.; Schmidt, C.M.; Yip-Schneider, M.T.; Allen, P.J.; Schattner, M.; et al. Combined circulating tumor DNA and protein biomarker-based liquid biopsy for the earlier detection of pancreatic cancers. Proc. Natl. Acad. Sci. USA 2017, 114, 10202–11020. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Pishvaian, M.J.; Joseph Bender, R.; Matrisian, L.M.; Rahib, L.; Hendifar, A.; Hoos, W.A.; Mikhail, S.; Chung, V.; Picozzi, V.; Heartwell, C.; et al. A pilot study evaluating concordance between blood-based and patient-matched tumor molecular testing within pancreatic cancer patients participating in the Know Your Tumor (KYT) initiative. Oncotarget 2016, 8, 83446–83456. [Google Scholar] [CrossRef] [Green Version]
  95. Vandekerkhove, G.; Struss, W.J.; Annala, M.; Kallio, H.M.L.; Khalaf, D.; Warner, E.W.; Herberts, C.; Ritch, E.; Beja, K.; Loktionova, Y.; et al. Circulating Tumor DNA Abundance and Potential Utility in De Novo Metastatic Prostate Cancer. Eur. Urol. 2019, 75, 667–675. [Google Scholar] [CrossRef] [Green Version]
  96. Spindler, K.L.; Pallisgaard, N.; Andersen, R.F.; Brandslund, I.; Jakobsen, A. Circulating free DNA as biomarker and source for mutation detection in metastatic colorectal cancer. PLoS ONE 2015, 10, e0108247. [Google Scholar] [CrossRef]
  97. Bachet, J.B.; Bouché, O.; Taieb, J.; Dubreuil, O.; Garcia, M.L.; Meurisse, A.; Normand, C.; Gornet, J.M.; Artru, P.; Louafi, S.; et al. RAS mutation analysis in circulating tumor DNA from patients with metastatic colorectal cancer: The AGEO RASANC prospective multicenter study. Ann. Oncol. Oncol. 2018, 29, 1211–1219. [Google Scholar] [CrossRef]
  98. Vidal, J.; Muinelo, L.; Dalmases, A.; Jones, F.; Edelstein, D.; Iglesias, M.; Orrillo, M.; Abalo, A.; Rodríguez, C.; Brozos, E.; et al. Plasma ctDNA RAS mutation analysis for the diagnosis and treatment monitoring of metastatic colorectal cancer patients. Ann. Oncol. 2017, 28, 1325–1332. [Google Scholar] [CrossRef]
  99. Buim, M.E.; Fanelli, M.F.; Souza, V.S.; Romero, J.; Abdallah, E.A.; Mello, C.A.; Alves, V.; Ocea, L.M.; Mingues, N.B.; Barbosa, P.N.; et al. Detection of KRAS mutations in circulating tumor cells from patients with metastatic colorectal cancer. Cancer Biol. Ther. 2015, 16, 1289–1295. [Google Scholar] [CrossRef] [Green Version]
  100. Wang, B.; Wu, S.; Huang, F.; Shen, M.; Jiang, H.; Yu, Y.; Yu, Q.; Yang, Y.; Zhao, Y.; Zhou, Y.; et al. Analytical and clinical validation of a novel amplicon-based NGS assay for the evaluation of circulating tumor DNA in metastatic colorectal cancer patients. Clin. Chem. Lab. Med. 2019, 57, 1501–1510. [Google Scholar] [CrossRef]
  101. Osumi, H.; Shinozaki, E.; Takeda, Y.; Wakatsuki, T.; Ichimura, T.; Saiura, A.; Yamaguchi, K.; Takahashi, S.; Noda, T.; Zembutsu, H. Clinical relevance of circulating tumor DNA assessed through deep sequencing in patients with metastatic colorectal cancer. Cancer Med. 2019, 8, 408–417. [Google Scholar] [CrossRef] [PubMed]
  102. Germano, G.; Mauri, G.; Siravegna, G.; Dive, C.; Pierce, J.; Di Nicolantonio, F.; D’Incalci, M.; Bardelli, A.; Siena, S.; Sartore-Bianchi, A. Parallel Evaluation of Circulating Tumor DNA and Circulating Tumor Cells in Metastatic Colorectal Cancer. Clin. Colorectal Cancer 2018, 17, 80–83. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Beije, N.; Helmijr, J.C.; Weerts, M.J.A.; Beaufort, C.M.; Wiggin, M.; Marziali, A.; Verhoef, C.; Sleijfer, S.; Jansen, M.P.H.M.; Martens, J.W.M. 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. Mol. Oncol. 2016, 10, 1575–1584. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Kato, S.; Schwaederlé, M.C.; Fanta, P.T.; Okamura, R.; Leichman, L.; Lippman, S.M.; Lanman, R.B.; Raymond, V.M.; Talasaz, A.; Kurzrock, R. 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, 1–16. [Google Scholar] [CrossRef]
  105. Mohamed Suhaimi, N.A.; Foong, Y.M.; Lee, D.Y.; Phyo, W.M.; Cima, I.; Lee, E.X.; Goh, W.L.; Lim, W.Y.; Chia, K.S.; Kong, S.L.; et al. Non-invasive sensitive detection of KRAS and BRAF mutation in circulating tumor cells of colorectal cancer patients. Mol. Oncol. 2015, 9, 850–860. [Google Scholar] [CrossRef] [Green Version]
  106. Kurihara, S.; Ueda, Y.; Onitake, Y.; Sueda, T.; Ohta, E.; Morihara, N.; Hirano, S.; Irisuna, F.; Hiyama, E. Circulating free DNA as non-invasive diagnostic biomarker for childhood solid tumors. J. Pediatr. Surg. 2015, 50, 2094–2097. [Google Scholar] [CrossRef] [Green Version]
  107. Chen, K.Z.; Lou, F.; Yang, F.; Zhang, J.B.; Ye, H.; Chen, W.; Guan, T.; Zhao, M.Y.; Su, X.X.; Shi, R.; et al. Circulating Tumor DNA Detection in Early-Stage Non-Small Cell Lung Cancer Patients by Targeted Sequencing. Sci. Rep. 2016, 6, 31985. [Google Scholar] [CrossRef] [Green Version]
  108. Li, X.; Ren, R.; Ren, S.; Chen, X.; Cai, W.; Zhou, F.; Zhang, Y.; Su, C.; Zhao, C.; Li, J.; et al. Peripheral blood for epidermal growth factor receptor mutation detection in non-small cell lung cancer patients. Transl. Oncol. 2014, 7, 341. [Google Scholar] [CrossRef] [Green Version]
  109. Jin, Y.; Shi, X.; Zhao, J.; He, Q.; Chen, M.; Yan, J.; Ou, Q.; Wu, X.; Shao, Y.W.; Yu, X. Mechanisms of primary resistance to EGFR targeted therapy in advanced lung adenocarcinomas. Lung Cancer 2018, 124, 110–116. [Google Scholar] [CrossRef]
  110. Guo, N.; Lou, F.; Ma, Y.; Li, J.; Yang, B.; Chen, W.; Ye, H.; Zhang, J.B.; Zhao, M.Y.; Wu, W.J.; et al. Circulating tumor DNA detection in lung cancer patients before and after surgery. Sci. Rep. 2016, 6, 33519. [Google Scholar] [CrossRef] [Green Version]
  111. Villaflor, V.; Won, B.; Nagy, R.; Banks, K.; Lanman, R.B.; Talasaz, A.; Salgia, R. Biopsy-free circulating tumor DNA assay identifies actionable mutations in lung cancer. Oncotarget 2016, 7, 66880–66891. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Schwaederlé, M.C.; Patel, S.P.; Husain, H.; Ikeda, M.; Lanman, R.B.; Banks, K.C.; Talasaz, A.; Bazhenova, L.; Kurzrock, R. Utility of Genomic Assessment of Blood-Derived Circulating Tumor DNA (ctDNA) in Patients with Advanced Lung Adenocarcinoma. Clin. Cancer Res. 2017, 23, 5101–5111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Yang, Y.; Shen, X.; Li, R.; Shen, J.; Zhang, H.; Yu, L.; Liu, B.; Wang, L. The detection and significance of EGFR and BRAF in cell-free DNA of peripheral blood in NSCLC. Oncotarget 2017, 8, 49773–49782. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Soria-Comes, T.; Palomar-Abril, V.; Ureste, M.M.; Guerola, M.T.; Maiques, I.C.M. Real-World Data of the Correlation between EGFR Determination by Liquid Biopsy in Non-squamous Non-small Cell Lung Cancer (NSCLC) and the EGFR Profile in Tumor Biopsy. Pathol. Oncol. Res. 2019, 1–7. [Google Scholar] [CrossRef] [PubMed]
  115. Yu, Q.; Huang, F.; Zhang, M.; Ji, H.; Wu, S.; Zhao, Y.; Zhang, C.; Wu, J.; Wang, B.; Pan, B.; et al. Multiplex picoliter-droplet digital PCR for quantitative assessment of EGFR mutations in circulating cell-free DNA derived from advanced non-small cell lung cancer patients. Mol. Med. Rep. 2017, 16, 1157–1166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Mok, T.; Wu, Y.L.; Lee, J.S.; Yu, C.J.; Sriuranpong, V.; Sandoval-Tan, J.; Ladrera, G.; Thongprasert, S.; Srimuninnimit, V.; Liao, M.; et al. Detection and dynamic changes of EGFR mutations from circulating tumor DNA as a predictor of survival outcomes in NSCLC patients treated with first-line intercalated erlotinib and chemotherapy. Clin. Cancer Res. 2015, 21, 3196–3203. [Google Scholar] [CrossRef] [Green Version]
  117. Zhu, Y.J.; Zhang, H.B.; Liu, Y.H.; Zhang, F.L.; Zhu, Y.Z.; Li, Y.; Bai, J.P.; Liu, L.R.; Qu, Y.C.; Qu, X.; et al. Quantitative cell-free circulating EGFR mutation concentration is correlated with tumor burden in advanced NSCLC patients. Lung Cancer 2017, 109, 124–127. [Google Scholar] [CrossRef]
  118. Yao, Y.; Liu, J.; Li, L.; Yuan, Y.; Nan, K.; Wu, X.; Zhang, Z.; Wu, Y.; Li, X.; Zhu, J.; et al. Detection of circulating tumor DNA in patients with advanced non-small cell lung cancer. Oncotarget 2017, 8, 2130–2140. [Google Scholar] [CrossRef] [Green Version]
  119. Cui, S.; Ye, L.; Wang, H.; Chu, T.; Zhao, Y.; Gu, A.; Xiong, L.; Shi, C.; Jiang, L. Use of SuperARMS EGFR Mutation Detection Kit to Detect EGFR in Plasma Cell-free DNA of Patients With Lung Adenocarcinoma. Clin. Lung Cancer 2018, 19, 313–322. [Google Scholar] [CrossRef]
  120. Leighl, N.B.; Page, R.D.; Raymond, V.M.; Daniel, D.B.; Divers, S.G.; Reckamp, K.L.; Villalona-Calero, M.A.; Dix, D.; Odegaard, J.I.; Lanman, R.B.; 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, 4691–4700. [Google Scholar] [CrossRef] [Green Version]
  121. Wu, Z.; Yang, Z.; Li, C.S.; Zhao, W.; Liang, Z.X.; Dai, Y.; Zhu, Q.; Miao, K.L.; Cui, D.H.; Chen, L.A. Differences in the genomic profiles of cell-free DNA between plasma, sputum, urine, and tumor tissue in advanced NSCLC. Cancer Med. 2019, 8, 910–919. [Google Scholar] [CrossRef] [PubMed]
  122. Sim, W.C.; Loh, C.H.; Toh, G.L.; Lim, C.W.; Chopra, A.; Chang, A.Y.C.; Goh, L.L. Non-invasive detection of actionable mutations in advanced non-small-cell lung cancer using targeted sequencing of circulating tumor DNA. Lung Cancer 2018, 124, 154–159. [Google Scholar] [CrossRef] [PubMed]
  123. Xu, S.; Lou, F.; Wu, Y.; Sun, D.Q.; Zhang, J.B.; Chen, W.; Ye, H.; Liu, J.H.; Wei, S.; Zhao, M.Y.; et al. Circulating tumor DNA identified by targeted sequencing in advanced-stage non-small cell lung cancer patients. Cancer Lett. 2016, 370, 324–331. [Google Scholar] [CrossRef] [PubMed]
  124. Reck, M.; Hagiwara, K.; Han, B.; Tjulandin, S.; Grohé, C.; Yokoi, T.; Morabito, A.; Novello, S.; Arriola, E.; Molinier, O.; et al. ctDNA Determination of EGFR Mutation Status in European and Japanese Patients with Advanced NSCLC: The ASSESS Study. J. Thorac. Oncol. Oncol. 2016, 11, 1682–1689. [Google Scholar] [CrossRef] [Green Version]
  125. Jia, J.; Huang, B.; Zhuang, Z.; Chen, S. Circulating tumor DNA as prognostic markers for late stage NSCLC with bone metastasis. Int. J. Biol. Markers 2018, 33, 222–230. [Google Scholar] [CrossRef] [PubMed]
  126. Veldore, V.H.; Choughule, A.; Routhu, T.; Mandloi, N.; Noronha, V.; Joshi, A.; Dutt, A.; Gupta, R.; Vedam, R.; Prabhash, K. Validation of liquid biopsy: Plasma cell-free DNA testing in clinical management of advanced non-small cell lung cancer. Lung Cancer 2018, 9, 1–11. [Google Scholar] [CrossRef] [Green Version]
  127. Ma, M.; Shi, C.; Qian, J.; Teng, J.; Zhong, H.; Han, B. Comparison of plasma and tissue samples in epidermal growth factor receptor mutation by ARMS in advanced non-small cell lung cancer. Gene 2016, 591, 58–64. [Google Scholar] [CrossRef]
  128. Denis, M.G.; Lafourcade, M.P.; Le Garff, G.; Dayen, C.; Falchero, L.; Thomas, P.; Locher, C.; Oliviero, G.; Licour, M.; Reck, M.; et al. Circulating free tumor-derived DNA to detect EGFR mutations in patients with advanced NSCLC: French subset analysis of the ASSESS study. J. Thorac. Dis. 2019, 11, 1370–1378. [Google Scholar] [CrossRef]
  129. Guibert, N.; Hu, Y.; Feeney, N.; Kuang, Y.; Plagnol, V.; Jones, G.; Howarth, K.; Beeler, J.F.; Paweletz, C.P.; Oxnard, G.R. Amplicon-based next-generation sequencing of plasma cell-free DNA for detection of driver and resistance mutations in advanced non-small cell lung cancer. Ann. Oncol. 2018, 29, 49–1055. [Google Scholar] [CrossRef]
  130. Howell, J.; Atkinson, S.R.; Pinato, D.J.; Knapp, S.; Ward, C.; Minisini, R.; Burlone, M.E.; Leutner, M.; Pirisi, M.; Büttner, R.; et al. Identification of mutations in circulating cell-free tumour DNA as a biomarker in hepatocellular carcinoma. Eur. J. Cancer 2019, 116, 56–66. [Google Scholar] [CrossRef]
  131. Bernard, V.; Kim, D.U.; San Lucas, F.A.; Castillo, J.; Allenson, K.; Mulu, F.C.; Stephens, B.M.; Huang, J.; Semaan, A.; Guerrero, P.A.; et al. Circulating Nucleic Acids Are Associated With Outcomes of Patients With Pancreatic Cancer. Gastroenterology 2019, 156, 108–118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Kinugasa, H.; Nouso, K.; Miyahara, K.; Morimoto, Y.; Dohi, C.; Tsutsumi, K.; Kato, H.; Matsubara, T.; Okada, H.; Yamamoto, K. Detection of K-ras gene mutation by liquid biopsy in patients with pancreatic cancer. Cancer 2015, 121, 2271–2280. [Google Scholar] [CrossRef] [PubMed]
  133. Gangadhar, T.C.; Savitch, S.L.; Yee, S.S.; Xu, W.; Huang, A.C.; Harmon, S.; Lieberman, D.B.; Soucier, D.; Fan, R.; Black, T.A.; et al. Feasibility of monitoring advanced melanoma patients using cell-free DNA from plasma. Pigment. Cell Melanoma Res. 2018, 31, 73–81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Haselmann, V.; Gebhardt, C.; Brechtel, I.; Duda, A.; Czerwinski, C.; Sucker, A.; Holland-Letz, T.; Utikal, J.; Schadendorf, D.; Neumaier, M. Liquid Profiling of Circulating Tumor DNA in Plasma of Melanoma Patients for Companion Diagnostics and Monitoring of BRAF Inhibitor Therapy. Clin. Chem. 2018, 64, 830–842. [Google Scholar] [CrossRef] [Green Version]
  135. Tang, H.; Kong, Y.; Si, L.; Cui, C.; Sheng, X.; Chi, Z.; Dai, J.; Yu, S.; Ma, M.; Wu, X.; et al. Clinical significance of BRAFV600E mutation in circulating tumor DNA in Chinese patients with melanoma. Oncol. Lett. 2018, 15, 1839–1844. [Google Scholar] [CrossRef]
  136. Pinzani, P.; Salvianti, F.; Cascella, R.; Massi, D.; De Giorgi, V.; Pazzagli, M.; Orlando, C. Allele specific Taqman-based real-time PCR assay to quantify circulating BRAFV600E mutated DNA in plasma of melanoma patients. Clin. Chim. Acta 2010, 411, 1319–1324. [Google Scholar] [CrossRef]
  137. Calapre, L.; Giardina, T.; Robinson, C.; Reid, A.L.; Al-Ogaili, Z.; Pereira, M.R.; McEvoy, A.C.; Warburton, L.; Hayward, N.K.; Khattak, M.A.; et al. Locus-specific concordance of genomic alterations between tissue and plasma circulating tumor DNA in metastatic melanoma. Mol. Oncol. 2019, 13, 171–184. [Google Scholar] [CrossRef] [Green Version]
  138. Sandulache, V.C.; Williams, M.D.; Lai, S.Y.; Lu, C.; William, W.N.; Busaidy, N.L.; Cote, G.J.; Singh, R.R.; Luthra, R.; Cabanillas, M.E. Real-Time Genomic Characterization Utilizing Circulating Cell-Free DNA in Patients with Anaplastic Thyroid Carcinoma. Thyroid 2017, 27, 81–87. [Google Scholar] [CrossRef] [Green Version]
  139. Kim, S.T.; Lee, W.S.; Lanman, R.B.; Mortimer, S.; Zill, O.A.; Kim, K.M.; Jang, K.T.; Kim, S.H.; Park, S.H.; Park, J.O.; 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, 40360–40369. [Google Scholar] [CrossRef] [Green Version]
  140. Rachiglio, A.M.; Esposito Abate, R.; Sacco, A.; Pasquale, R.; Fenizia, F.; Lambiase, M.; Morabito, A.; Montanino, A.; Rocco, G.; Romano, C.; et al. Limits and potential of targeted sequencing analysis of liquid biopsy in patients with lung and colon carcinoma. Oncotarget 2016, 7, 66595–66605. [Google Scholar] [CrossRef] [Green Version]
  141. Phallen, J.; Sausen, M.; Adleff, V.; Leal, A.; Hruban, C.; White, J.; Anagnostou, V.; Fiksel, J.; Cristiano, S.; Papp, E.; et al. Direct detection of early-stage cancers using circulating tumor DNA. Sci. Transl. Med. 2017, 9, 403. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Baumgartner, J.M.; Raymond, V.M.; Lanman, R.B.; Tran, L.; Kelly, K.J.; Lowy, A.M.; Kurzrock, R. Preoperative Circulating Tumor DNA in Patients with Peritoneal Carcinomatosis is an Independent Predictor of Progression-Free Survival. Ann. Surg Oncol. 2018, 25, 2400–2408. [Google Scholar] [CrossRef] [PubMed]
  143. Perkins, G.; Yap, T.A.; Pope, L.; Cassidy, A.M.; Dukes, J.P.; Riisnaes, R.; Massard, C.; Cassier, P.A.; Miranda, S.; Clark, J.; et al. Multi-purpose utility of circulating plasma DNA testing in patients with advanced cancers. PLoS ONE 2012, 7, E47020. [Google Scholar] [CrossRef] [PubMed]
  144. Aparicio, S.; Caldas, C. The implications of clonal genome evolution for cancer medicine. N. Engl. J. Med. 2013, 368, 842–851. [Google Scholar] [CrossRef] [PubMed]
  145. Cleary, A.S.; Leonard, T.L.; Gestl, S.A.; Gunther, E.J. Tumour cell heterogeneity maintained by cooperating subclones in Wnt-driven mammary cancers. Nature 2014, 508, 113–117. [Google Scholar] [CrossRef]
  146. Ma, F.; Zhu, W.; Guan, Y.; Yang, L.; Xia, X.; Chen, S.; Li, Q.; Guan, X.; Yi, Z.; Qian, H.; et al. ctDNA dynamics: A novel indicator to track resistance in metastatic breast cancer treated with anti-HER2 therapy. Oncotarget 2016, 7, 66020–66031. [Google Scholar] [CrossRef]
  147. Shatsky, R.; Parker, B.A.; Bui, N.Q.; Helsten, T.; Schwab, R.B.; Boles, S.G.; Kurzrock, R. Next-Generation Sequencing of Tissue and Circulating Tumor DNA: The UC San Diego Moores Center for Personalized Cancer Therapy Experience with Breast Malignancies. Mol. Cancer Ther. 2019, 18, 1001–1011. [Google Scholar] [CrossRef] [Green Version]
  148. Perdigones, N.; Murtaza, M. Capturing tumor heterogeneity and clonal evolution in solid cancers using circulating tumor DNA analysis. Pharmacol. Ther. 2017, 174, 22–26. [Google Scholar] [CrossRef]
  149. Molina, R.; Filella, X.; Augé, J.M.; Fuentes, R.; Bover, I.; Rifa, J.; Moreno, V.; Canals, E.; Viñolas, N.; Marquez, A.; et al. Tumor markers (CEA, CA 125, CYFRA 21-1, SCC and NSE) in patients with non-small cell lung cancer as an aid in histological diagnosis and prognosis. Comparison with the main clinical and pathological prognostic factors. Tumour Biol. 2003, 24, 209–218. [Google Scholar] [CrossRef]
  150. Forshew, T.; Murtaza, M.; Parkinson, C.; Gale, D.; Tsui, D.W.; Kaper, F.; Dawson, S.J.; Piskorz, A.M.; Jimenez-Linan, M.; Bentley, D.; et al. Noninvasive identification and monitoring of cancer mutations by targeted deep sequencing of plasma DNA. Sci. Transl. Med. 2012, 4, 136ra68. [Google Scholar] [CrossRef]
  151. Hovelson, D.H.; Liu, C.J.; Wang, Y.; Kang, Q.; Henderson, J.; Gursky, A.; Brockman, S.; Ramnath, N.; Krauss, J.C.; Talpaz, M. Rapid, ultra low coverage copy number profiling of cell-free DNA as a precision oncology screening strategy. Oncotarget 2017, 8, 89848–89866. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Bonavia, R.; Inda, M.M.; Cavenee, W.K.; Furnari, F.B. Heterogeneity maintenance in glioblastoma: A social network. Cancer Res. 2011, 71, 4055–4060. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Pectasides, E.; Stachler, M.D.; Derks, S.; Liu, Y.; Maron, S.; Islam, M.; Alpert, L.; Kwak, H.; Kindler, H.; Polite, B.; et al. Genomic Heterogeneity as a Barrier to Precision Medicine in Gastroesophageal Adenocarcinoma. Cancer Discov. 2018, 8, 37–48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Paik, P.K.; Shen, R.; Won, H.; Rekhtman, N.; Wang, L.; Sima, C.S.; Arora, A.; Seshan, V.; Ladanyi, M.; Berger, M.F.; et al. NextGeneration Sequencing of Stage IV Squamous Cell Lung Cancers Reveals an Association of PI3K Aberrations and Evidence of Clonal Heterogeneity in Patients with Brain Metastases. Cancer Discov. 2015, 5, 610–621. [Google Scholar] [CrossRef] [Green Version]
  155. Kim, M.J.; Lee, H.S.; Kim, J.H.; Kim, Y.J.; Kwon, J.H.; Lee, J.O.; Bang, S.M.; Park, K.U.; Kim, D.W.; Kang, S.B.; et al. Different metastatic pattern according to the KRAS mutational status and site-specific discordance of KRAS status in patients with colorectal cancer. BMC Cancer 2012, 12, 347. [Google Scholar] [CrossRef] [Green Version]
  156. Knijn, N.; Mekenkamp, L.J.; Klomp, M.; Vink-Borger, M.E.; Tol, J.; Teerenstra, S.; Meijer, J.W.; Tebar, M.; Riemersma, S.; van Krieken, J.H.; et al. KRAS mutation analysis: A comparison between primary tumours and matched liver metastases in 305 colorectal cancer patients. Br. J. Cancer 2011, 104, 1020–1026. [Google Scholar] [CrossRef] [Green Version]
  157. Gao, J.; Wang, H.; Zang, W.; Li, B.; Rao, G.; Li, L.; Yu, Y.; Li, Z.; Dong, B.; Lu, Z.; et al. Circulating tumor DNA functions as an alternative for tissue to overcome tumor heterogeneity in advanced gastric cancer. Cancer Sci. 2017, 108, 1881–1887. [Google Scholar] [CrossRef]
  158. Volik, S.; Alcaide, M.; Morin, R.D.; Collins, C. Cell-free DNA (cfDNA): Clinical Significance and Utility in Cancer Shaped by Emerging Technologies. Mol. Cancer Res. 2016, 14, 898–908. [Google Scholar] [CrossRef] [Green Version]
  159. Bosse, K.R.; Maris, J.M. Advances in the translational genomics of neuroblastoma: From improving risk stratification and revealing novel biology to identifying actionable genomic alterations. Cancer 2016, 122, 20–33. [Google Scholar] [CrossRef]
  160. Olsson, E.; Winter, C.; George, A.; Chen, Y.; Howlin, J.; Tang, M.H.; Dahlgren, M.; Schulz, R.; Grabau, D.; van Westen, D.; et al. Serial monitoring of circulating tumor DNA in patients with primary breast cancer for detection of occult metastatic disease. EMBO Mol. Med. 2015, 7, 1034–1047. [Google Scholar] [CrossRef]
  161. Mondaca, S.; Offin, M.; Borsu, L.; Myers, M.; Josyula, S.; Makhnin, A.; Shen, R.; Riely, G.J.; Rudin, C.M.; Ladanyi, M.; et al. Lessons learned from routine, targeted assessment of liquid biopsies for EGFR T790M resistance mutation in patients with EGFR mutant lung cancers. Acta Oncol. 2019, 1634–1639. [Google Scholar] [CrossRef] [PubMed]
  162. Horn, L.; Whisenant, J.G.; Wakelee, H.; Reckamp, K.L.; Qiao, H.; Leal, T.A.; Du, L.; Hernandez, J.; Huang, V.; Blumenschein, G.R.; et al. Monitoring therapeutic response and resistance: Analysis of circulating tumor DNA in patients with ALK+ lung cancer. J. Thorac. Oncol. 2019, 14, 1901–1911. [Google Scholar] [CrossRef] [PubMed]
  163. Toledo, R.A.; Garralda, E.; Mitsi, M.; Pons, T.; Monsech, J.; Vega, E.; Otero, Á.; Albarran, M.I.; Baños, N.; Durán, Y.; et al. Exome Sequencing of Plasma DNA Portrays the Mutation Landscape of Colorectal Cancer and Discovers Mutated VEGFR2 Receptors as Modulators of Antiangiogenic Therapies. Clin. Cancer Res. 2018, 24, 3550–3559. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Analysis of cfDNA and tumour tissue in patients with solid tumours: (A) tissue biopsies and cfDNA samples of patients with primary or metastatic tumours are processed for detection of specific somatic genomic alterations (SGAs). The concordance rate between cfDNA and primary tumour or cfDNA and primary and metastatic tumours can be evaluated (B) A summary of factors affecting concordance rates between cfDNA and tumour tissue, 1o: primary tumour, 2o: secondary (metastatic) tumour.
Figure 1. Analysis of cfDNA and tumour tissue in patients with solid tumours: (A) tissue biopsies and cfDNA samples of patients with primary or metastatic tumours are processed for detection of specific somatic genomic alterations (SGAs). The concordance rate between cfDNA and primary tumour or cfDNA and primary and metastatic tumours can be evaluated (B) A summary of factors affecting concordance rates between cfDNA and tumour tissue, 1o: primary tumour, 2o: secondary (metastatic) tumour.
Cancers 11 01938 g001
Table 1. Example summary data of trends for high concordance rates between somatic GAs of cfDNA and metastatic tumours compared to cfDNA and primary tumour.
Table 1. Example summary data of trends for high concordance rates between somatic GAs of cfDNA and metastatic tumours compared to cfDNA and primary tumour.
Author/Cohort SizeCancer TypeConcordance with Primary or Metastatic TumourDriver and Actionable Driver Alterations Method for Tumour/cfDNA
Thompson/102 [15]NSCLCcfDNA and primary tumour (51%) compared to cfDNA and metastatic tumour (79%) for all alterations50 drivers and 12 resistance alterationsTargeted sequencing
Liu/72 [81]NSCLCcfDNA and primary (50%) compared to cfDNA and metastatic (65%) in 19 patientslung cancer panel including EGFR L858R, L861Q, e19 del, e20INS, KRAS G12X, EML4-ALK, RET-KIF5B, BRAF V600EARMS-PCR and targeted sequencing/Sequencing and ddPCR
Xie/35 [83]NSCLCcfDNA and metastatic tumour (73.2 %), cfDNA and primary tumour (68.4%)56 lung cancer genesTargeted sequencing
Guo/56 [72]NSCLC54.6% of patients in early stage and 80% in late stagelung & colon cancer panel (LV103) and lung cancer panel (L82)Targeted sequencing for both, ddPCR for some cfDNA samples
Garcia- Saenz/49 [47]6 metastatic and 43 localised breast cancer59.1% (overall)
79.8% (for metastatic patients)
PIK3CA mutationsCOBAS PIK3CA Mutation Test/ddPCR using (rare PIK3CA Mutation Assays)
Tzanikou/56 [84]Early and metastatic breast cancer48.2% (27/56) in early breast cancer, 66.6% (18/27) in metastatic breast cancerPIK3CA mutationsCustom method and ddPCR
Chae/12 [70]mCRCFor sequencing approaches, 39% for primary and 55% for metastasis in all panel21 gene panel including TP53, PIK3CA and KRASTargeted sequencing/targeted sequencing, OnTarget assay and ddPCR
Kato/55 [85]Esophageal, gastroesophageal junction, and gastric adenocarcinomaconcordance between ctDNA and primary site vs. cfDNA and metastatic site for TP53: 52.2% vs. 87.5% and for ERBB2: 78.3% vs. 100%54-73 gene panel including KRAS, TP53 and PTENSequencing
NSCLC: non-small cell lung cancer; mCRC: metastatic colorectal cancer. Studies using cfDNA that investigated (1) the mechanism of resistance to drug therapy and (2) the comparison of methods and (3) efficacy of drug therapy have not been included, but studies that compare the extent of which cfDNA and reflect driver and actionable driver alterations of the tumours, primary and metastatic have been included.
Table 2. Comprehensive summary data for driver and actionable driver alterations concordance rates between cfDNA and tumours in individual cancer types.
Table 2. Comprehensive summary data for driver and actionable driver alterations concordance rates between cfDNA and tumours in individual cancer types.
Author/Cohort SizeCancer TypeConcordance InformationPositive Concordance (MUT/MUT)|Negative Concordance (WT/WT)|DiscordanceDriver and Actionable Driver Alterations Method for Tumour/cfDNA
Wyatt/45 [10]MPC 88.9% in clinically actionable genes 72 genes including AR, BRCA2, PTEN, PIK3CA and TP53WES/targeted sequencing
Vandekerkhove/53 [95]MPC 80% in matched samples Panel of genes including TP53 and DNA repair genesTargeted sequencing
Grasselli/146 [31]mCRC89.7% 10.3% (15 cases) concordanceRAS mutationsSoC PCR/ddPCR (BEAMing)
Bando/280 [32] mCRC86.4% (242/280)82.1% (110/134)|90.4% (132/146)|11% (38/280) RAS mutationsddPCR (BEAMing)
Garcia-Foncillas/236 [65]mCRC89% (210/236) improved to 92% by re-analysis86.30%|92.40%|In lung metastasis cases (tissue only)RAS mutationsSoC PCR/OncoBEAM
Schmiegel/98 [33]mCRC91.8% (90/98)90.4% (47/52)|93.5% (43/46)|-RAS mutationsSequencing, SOC, ddPCR (BEAMing)/ddPCR (BEAMing)
Demuth/28 [75] mCRC79% for Ion Torrent seq.- 89% for ddPCR KRAS mutationsGenotyping/Sequencing and ddPCR
Spindler/229 [96]mCRC85% KRASStandard methods/ARMS-qPCR
Bachet/425 [97]mCRC71%- 89% RASStandard methods/sequencing
Vidal/115 [98]mCRC93% RASStandard methods/OncoBEAM
Buim/26 [99]mCRC71% KRASStandard methods/pyrosequencing
Thierry/140 [66]mCRC72%, 74% and 87% for KRAS exon 2, KRAS exon 3–4 and BRAF V600E, respectively 28 mutations including KRAS, BRAF, NRASStandard methods/Q-PCR-based-method (IntPlex V)
Wang/184 [100] mCRC93.33% in pre-treatment cohort KRAS, NRAS, BRAF, PIK3CAARMS-based PCR
/Firefly
Osumi/101 [101]mCRC77.2% (78/101) for RAS23 cases for RAS (discordance)14 CRC- related genes including, APC, TP53 and RASStandard methods/Sequencing
Germano/20 [102]mCRC84.6% (11/13 cases) RAS, BRAF, ERBB2Standard methods/ddPCR
Beije/12 [103]mCRCKRAS, PIK3CA and TP53 for OnTarget assay (80%), digital PCR (93%) 21 CRC gene panel including TP53, PIK3CA and KRASSequencing/Sequencing, OnTarget assay and ddPCR
Kato/94 [104]CRCranging from 63.2% APC to 85.5% BRAF panel including KRAS, TP53 and APCSequencing
Mohamed Suhaimi/44 [105]CRC84.1% for KRAS and 90.9% BRAF KRAS and BRAFGenotyping/sanger sequencing, HRM and ASPCR and pyroseqeuncing
Takeshita/35 [44]MBC74.3% (26/35) 1/35|25/35|9/35ESR1 mutationsddPCR
Beaver/29 [48]Early BC14/15 mutations PIK3CA mutationsSanger sequencing, ddPCR/ddPCR
Higgins/49 and 60 [45] MBC (49 retrospective and 60 prospective)100% in 41 matched retrospectives, 72.5% in 51 prospectives 27.5% in 51 prospective samples (discordance) PIK3CA mutationsSequencing or BEAMing/ddPCR (BEAMing)
Chae/45 [70] BC91.0%–94.2% for all genes10.8%–15.1% (3.5% for CNAs) positive concordance Foundation 1/Guardant360
Board/76 [46] 46 metastatic, 30 localised BC95% in 41 matched samples80%|(47%) discordancePIK3CA mutationsStandard methods/ARMS PCR*
Garcia- Saenz/49 [47]6 Metastatic and 43 localised BC59.1% (overall)
79.8% (for metastatic patients)
PIK3CA mutationsCOBAS PIK3CA Mutation Test/ddPCR using (rare PIK3CA Mutation Assays)
Kodahl/66 [49]PIK3CA- mutated MBC83% (20/24 cases) PIK3CA mutationsddPCR
Combaret/114 [52]NB100% 1/1|1/1|0ALK; F1174L (e23: 3520, T>C)ddPCR and targeted sequencing
55 cases6 cases|49 cases|4 (cfDNA only), 1 (tumour only)ALK, F1174L (e23:3522, C>A)
58 cases12 cases|46 cases|1 (cfDNA only), 1 (tumour only)ALK; R1275Q (e25:3824, G>A)
Kurihara/10 [106]NB100% 2/2|8/8|0MYCNFISH/ddPCR
Chen/58 [107]Stage IA, IB, and IIA NSCLC 50.4% Panel of 50 driver alterations including EGFR, KRAS, PIK3CA and TP53Targeted sequencing
Sung/126 [36]NSCLC90% (ex19del), and 88.33% (L858R) EGFR (ex19del and L858R)Genotyping/Targeted sequencing and ddPCR
Li/164 [108]NSCLC73.6% EGFR mutationsARMS
Lee/81 [37]NSCLC86.2% (ex19del) and 87.9% (L858R) EGFR (ex19del and L858R)Genotyping/ddPCR
Thompson/102 [15]NSCLC79% (19/24) for actionable EGFR mutations
97.5% across all variants
60% across all variants50 drivers, 12 resistance alterationsSequencing
Jin/69 [109]NSCLC88.2% for EGFR mutations EGFR Ex19del, L858R, G719S/C, and L861Q,
TP53 mutations, amp. of RB1, PIK3CA and MYC
Targeted Sequencing
Yang/73 [68]NSCLC74% (54/73)26% (19/73) (discordance)EGFR mutations Sequencing/Sequencing and ddPCR
Guo/41 [110]NSCLC78.1% 50 cancer genes including EGFR, KRAS, and TP53Targeted sequencing
Villaflor/68 [111]NSCLCHigh concordance for truncal oncogenic drivers, 71% for EGFR Driver alterations including EGFRtargeted multiplex testing or tissue- based sequencing/Guardant360
Liu/72 [81]NSCLC54.2% for all clinically actionable alterations,
EGFR L858R (93.1%),
EGFR e19 del (90.3%),
KRAS G12X (96.9%),
ALK rearrang. (96.9%)
MET or HER2 CNA in cfDNA but not tumour (discordance)EGFR L858R,L861Q,e19 del, e20 INS, KRAS G12X, EML4-ALK, RET-KIF5B and BRAF V600EARMS-PCR and sequencing/Sequencing (cfDNA also validated by ddPCR)
Schwaederle/88 [112]NSCLC76.5- 80.8 % for EGFR mutations depending on sampling time7/26 (EGFR mutations)
53% for all alterations|14/26 (EGFR mutations)|5/26 (EGFR mutations)
2 cfDNA only, 3 tumour only
Mutations in TP53, EGFR, MET, KRAS and ALKSequencing or genotyping or no test/Guardant360
Yang/107 [113]NSCLC74.8% (80/107) EGFR
88.8% (95/107) BRAF
EGFR and BRAF mutationsStandard methods/competitive Allele-Specific TaqMan PCR (CastPCR)
Soria- Comes/102 [114]NSCLC87.4% EGFR mutationsCobas EGFR assay
Yu/22 [115]Advanced NSCLCFor 19DEL and L858R (90% and 95%, respectively) EGFR mutations (19DEL and L858R)ARMS/ddPCR
Mok/241 [116]Advanced NSCLC 88% (209/238) EGFR mutationsCobas 4800 FFPET test/Cobas 4800 blood test
Zhu/51 [117]Advanced NSCLC86.73% EGFR mutationsStandard methods/ddPCR
Yao/39 [118]Advanced NSCLC78.21% (30.5/39) for all genes47.43%|30.77%|21.8%Panel of 40 genes including EGFR, KRAS, PIK3CA, ALK and RETTargeted sequencing
Cui/180 [119]Advanced NSCLC87.8%97.3%|85.3%EGFR mutationsStandard methods/SuperARMS
Leighl/282 [120]Advanced NSCLC98.2% for EGFR, ALK, ROS1, BRAF SoC PCR/Guardant360
Wu/50 [121]Advanced NSCLC86% (43/50 cases) Driver alterations including EGFR, TP53, RB1Sequencing
Sim/50 [122] Advanced NSCLC81% for EGFR BRAF, EGFR, ERBB2, KRAS, NRAS, PIK3CASequencing
Xu/42 [123]Advanced NSCLCOverall 76% EGFR, KRAS, PIK3CA, and TP53Targeted sequencing
Reck/1311 [124]Advanced NSCLC89% (in 1162 matched samples) EGFR mutationsStandard methods of local centres
Jia/150 [125]Advanced NSCLC94.7% for EGFR and RAS EGFR and KRAS mutationsStandard methods/ddPCR
Veldore/132 [126]Advanced NSCLC96.96% EGFR mutationsStandard methods/sequencing
Ma/219 [127]Advanced NSCLC82% EGFR mutationsARMS
Denis/1311 [128]Advanced NSCLC96% in 126 matched samples EGFR mutationsStandard methods
Guibert/46 [129]Advanced NSCLCROS1/ALK (8/9), EGFR (9/9), BRAF/MET/HER2 (4/6) EGFR mutations, ROS1, ALK, BRAF/MET/HER2Standard methods/Sequencing and ddPCR
Hahn/19 [90]mRCC8.6% concordanceDNA repair genes (discordance) Foundation 1/Guardant360
Howell/51 [130]HCCmoderate ARID1A AXIN1, ATM, CTNNB1, HNF1A and TP53Targeted sequencing
Bernard/194 [131]PDAC (localised or metastatic)>95% for KRAS in surgically resected tissue KRASddPCR
Cohen/221 [93]PDAC100% KRAS mutationsSequencing
Pishvaian/34 [94]Pancreatic cancerLow concordance Panels including KRAS and TP53 Foundation 1/Guardant360
Kinugasa/75 [132] Pancreatic cancer77.3% (58/75) KRASPCR-PHFA/ddPCR
Gangadhar/25 [133]Advanced melanoma81.8% (9/11) 61 gene panel including BRAF, NRAS and KITStandard methods/Sequencing
Haselmann/634 [134]MelanomaBRAFV600 (92.3%–94.5%) BRAFSoC PCR/BEAMing
Tang/58 [135]Melanoma70.2% BRAFStandard methods/3D ddPCR
Pinzani/55 [136]Melanoma80% BRAFAllele-specific RT-PCR
Calapre/24 [137]Advanced melanoma80% (in a subgroup of 7 matching tissue and cfDNA) 30 melanoma genes including BRAF, NRAS, NF1 and TERTTargeted sequencing (ddPCR for some cfDNA cases)
Sandulache/23 [138]Anaplastic thyroid carcinomahigh for BRAF, PIK3CA, NRAS, and PTEN and moderate for TP53Highest discordance in post-treatment patients50 gene panel for tissue, 70 gene panel for cfDNA, including BRAF, NRAS, TP53 and PIK3CASequencing
MPC: metastatic prostate cancer; mCRC: metastatic colorectal cancer; MBC: metastatic breast cancer; NB: neuroblastoma; NSCLC: non- small cell lung cancer; mRCC: metastatic renal cell carcinoma; HCC: hepatocellular carcinoma; PDAC: pancreatic ductal adenocarcinoma. ARMS: amplification refractory mutation system with scorpion probes. Positive concordance refers to mutant cfDNA/mutant tumour tissue, whereas negative concordance refers to WT cfDNA/WT tumour tissue. Overall concordance includes positive and negative concordance.
Table 3. Comprehensive summary data for driver and actionable driver alterations concordance rates between cfDNA and tumours in studies of multiple cancer types.
Table 3. Comprehensive summary data for driver and actionable driver alterations concordance rates between cfDNA and tumours in studies of multiple cancer types.
Author/Cohort SizeCancer TypeConcordance or Discordance InformationDriver and Actionable Driver Alterations Method for Tumour/cfDNA
Kim/75 [139]CRC, melanoma gastrointestinal stromal tumour, renal cell carcinoma, gastric cancer, sarcoma and 4 other cancers85.9% when all detected mutations considered across all tumour typesPanel of 54 cancer genesSequencing
Rachiglio/79 [140]44 metastatic NSCLC and 35 mCRCHigh concordance for EGFR (17/22) and lower concordance for other driversALK, EGFR, ERBB2, ERBB4, FGFR1, FGFR2, FGFR3, MET, DDR2, KRAS, PIK3CA, BRAF, AKT1, PTEN, NRAS, MAP2K1, STK11, NOTCH1, CTNNB1, SMAD4, FBXW7, TP53Sequencing/Sequencing and ddPCR
Phallen/200 [141] Breast, colorectal, Lung, Ovarian cancerHigh concordance58 cancer related genes including driversSequencing (TEC-Seq)
Riviere/213 [9]colorectal adenocarcinoma, appendiceal adenocarcinoma, hepatocellular carcinoma, pancreatic ductal adenocarcinoma96% KRAS amplification, 94% MYC amplification, 95% KRAS G12V, 91% EGFR amplification
96% overall concordance on gene level
Panel of 68 genes including KRAS amplification, MYC amplification, KRAS G12V, EGFR amplificationGuardant360 panel
Jovelet/334 [76]thoracic, gastrointestinal, breast, head and neck, gynaecologic and urologic cancersOn a gene level only 173/347 mutations corresponded between cfDNA and tumour tissue, 174/347 discordant mutationsPanel of 50 cancer hotspots V2 (CHP2) including TP53, KRAS, PIK3CA, EGFR, APCSequencing
Leary/91 [56]Colorectal or breast cancerGood concordance for cancer driver genes such as ERBB2 and CDK6Chromosomal alterations including rearrangements of CDK6 and ERBB2 lociSequencing
Toor/28 [89]advanced stage gastrointestinal and lung malignancies 7% for lung subgroup, 8% for gastrointestinal subgroup (90% positive concordance), high discordance with respect to driver and actionable alteration Caris or paradigm panels/Guardant360 panel
Baumgartner/80 [142]appendix cancer, colorectal, peritoneal mesothelioma, small bowel, cholangiocarcinoma, ovarian, and testicular cancerOverall, positive, and negative concordance was 96.7%, 35.3%, and 96.6% (in 15 cases with matched samples)Panel of genes including TP53 and KRASSequencing
Kato/55 [85]Esophageal, gastroesophageal junction, and gastric adenocarcinoma61.3% (TP53 alterations) to 87.1% (KRAS alterations)54-73 gene panel
Including KRAS, TP53 and PTEN
Sequencing
Perkins/105 [143]Colorectal, melanoma, breast, prostate, ovarian, NSCLC, mesothelioma, sarcoma, glioblastoma, ACUP, cholangiocarcinoma, and cervical, endometrial, duodenal, esophageal, pancreatic and renal cancersOverall 60% (25/42)BRAF, KRAS, NRAS, HRAS, MET, AKT, PIK3CA, KITStandard methods/Mass Spectrometry TypePLEX and OncoCarta panel (v1.0)
CRC: colorectal cancer; mCRC: mCRC: metastatic colorectal cancer; NSCLC: non-small cell lung cancer; ACUP: adenocarcinoma of unknown primary.

Share and Cite

MDPI and ACS Style

Jahangiri, L.; Hurst, T. Assessing the Concordance of Genomic Alterations between Circulating-Free DNA and Tumour Tissue in Cancer Patients. Cancers 2019, 11, 1938. https://doi.org/10.3390/cancers11121938

AMA Style

Jahangiri L, Hurst T. Assessing the Concordance of Genomic Alterations between Circulating-Free DNA and Tumour Tissue in Cancer Patients. Cancers. 2019; 11(12):1938. https://doi.org/10.3390/cancers11121938

Chicago/Turabian Style

Jahangiri, Leila, and Tara Hurst. 2019. "Assessing the Concordance of Genomic Alterations between Circulating-Free DNA and Tumour Tissue in Cancer Patients" Cancers 11, no. 12: 1938. https://doi.org/10.3390/cancers11121938

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop