Next Article in Journal
An Immune Signature for Risk Stratification and Therapeutic Prediction in Helicobacter pylori-Infected Gastric Cancer
Next Article in Special Issue
Extracellular Vesicles-ceRNAs as Ovarian Cancer Biomarkers: Looking into circRNA-miRNA-mRNA Code
Previous Article in Journal
HBV preS Mutations Promote Hepatocarcinogenesis by Inducing Endoplasmic Reticulum Stress and Upregulating Inflammatory Signaling
Previous Article in Special Issue
Deciphering Tumour Heterogeneity: From Tissue to Liquid Biopsy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Circulating Tumor DNA-Based Genomic Profiling Assays in Adult Solid Tumors for Precision Oncology: Recent Advancements and Future Challenges

1
Project for Development of Liquid Biopsy Diagnosis, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
2
Cancer Precision Medicine, Inc., Kawasaki 213-0012, Japan
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(13), 3275; https://doi.org/10.3390/cancers14133275
Submission received: 11 June 2022 / Revised: 30 June 2022 / Accepted: 2 July 2022 / Published: 4 July 2022
(This article belongs to the Special Issue Liquid Nucleic Acid-Based Biomarkers in Solid Tumors)

Abstract

:

Simple Summary

The use of liquid biopsy for tumor genomic profiling to identify genomic biomarkers for targeted therapies has revolutionized the clinical practice in oncology management. In this review, we have summarized the recent advancements of liquid biopsy-based genomic profiling that have led to their approval for treatment selection in advanced cancer patients and highlighted the major factors that should be considered to choose the most appropriate genomic profiling assay for different patients under different clinical conditions.

Abstract

Genomic profiling using tumor biopsies remains the standard approach for the selection of approved molecular targeted therapies. However, this is often limited by its invasiveness, feasibility, and poor sample quality. Liquid biopsies provide a less invasive approach while capturing a contemporaneous and comprehensive tumor genomic profile. Recent advancements in the detection of circulating tumor DNA (ctDNA) from plasma samples at satisfactory sensitivity, specificity, and detection concordance to tumor tissues have facilitated the approval of ctDNA-based genomic profiling to be integrated into regular clinical practice. The recent approval of both single-gene and multigene assays to detect genetic biomarkers from plasma cell-free DNA (cfDNA) as companion diagnostic tools for molecular targeted therapies has transformed the therapeutic decision-making procedure for advanced solid tumors. Despite the increasing use of cfDNA-based molecular profiling, there is an ongoing debate about a ‘plasma first’ or ‘tissue first’ approach toward genomic testing for advanced solid malignancies. Both approaches present possible advantages and disadvantages, and these factors should be carefully considered to personalize and select the most appropriate genomic assay. This review focuses on the recent advancements of cfDNA-based genomic profiling assays in advanced solid tumors while highlighting the major challenges that should be tackled to formulate evidence-based guidelines in recommending the ‘right assay for the right patient at the right time’.

Graphical Abstract

1. Introduction

Technological advancements and reduction in sequencing costs have enabled genomic profiling of solid tumors to be performed routinely, which promoted the incorporation of precision oncology into the standard of care for advanced cancer patients [1]. Molecular profiling of tumor tissues, either from surgical resections or biopsy specimens, remains the standard approach to identify actionable genomic aberrations for molecular targeted therapies. However, the quality, quantity, and availability of tumor tissues from advanced cancer patients often pose challenges to the implementation of comprehensive genomic profiling (CGP) in clinical settings [2]. Recent multi-institutional studies have shown that 23–26% of collected tissue specimens could not proceed to CGP as a result of insufficient DNA quantity or tumor content [3,4]. Performing re-biopsy at recurrence is often difficult where the procedure is unfeasible in 20–60% of cases [5,6]. Furthermore, re-biopsy is also associated with potential complications and increased turnaround time, delaying treatment initiation [7,8,9]. Thus, the invasiveness of tissue biopsy may preclude re-biopsy at the time of recurrence and impede the identification of resistance mutations [10,11]. Furthermore, genomic profiling of tumor tissue provides a snapshot of a single point in space and time, lacking the ability to capture complex tumor heterogeneity and tumor clonal dynamics [12] (Figure 1).
Liquid biopsies, which involve genomic profiling of tumors using circulating biomarkers in the bodily fluid have emerged as a promising tool to complement and overcome the challenges of tissue-based CGP [13,14,15]. Among the different circulating biomarkers, circulating cell-free DNA (cfDNA) from blood has been most widely studied [16]. The origin and biology of cfDNA have been extensively discussed in previous reviews [17,18,19,20,21,22]. In brief, cfDNA is highly degraded DNA fragments released from apoptosis, necrosis, and secretion from cells [23,24]. The majority of cfDNA in plasma of healthy individuals originate from hematopoietic cells: 55% from white blood cells, 30% from erythroid progenitors, 10% from endothelial and 1% from hepatocytes [25,26,27]. In cancer patients, plasma cfDNA that originates from tumors, or commonly known as circulating tumor DNA (ctDNA), typically represents 0.01–90% of the total cfDNA found in blood [28]. Plasma ctDNA has been found to recapitulate the tumor’s molecular alterations, highlighting its potential to be used as a minimally invasive tumor marker in cancer patients [29]. Despite the low concentration of ctDNA present in blood, recent advancements in sequencing technology and bioinformatics enabled accurate detection of these ctDNA genomic alterations [30,31,32]. The accessibility and minimally invasiveness of blood sampling compared to tumor tissue biopsy allows genomic profiling to be conducted at multiple time points during cancer management, allowing real-time evaluation of treatment response and detecting clonal evolution during disease progression or recurrence [33,34,35]. Furthermore, a single blood sample may allow the capturing of ctDNA released from multiple tumor sites and regions, allowing the detection of inter- and intra-tumor heterogeneity that might be missed from a single-site tissue biopsy, depicting a more comprehensive and complete genomic profile of the tumor [36,37,38] (Figure 1).
The compilation of evidence that supports the use of ctDNA to identify actionable alterations from both analytical and clinical studies in recent years have advocated the FDA approval of several single-gene and multigene assays to be used as companion diagnostics matched to specific targeted therapies [30]. Despite the advantages of using liquid biopsy over tissue biopsy for CGP, limitations and challenges exist. cfDNA often presents at low concentrations (usually below 10 ng or 3000 genome copies per mL of plasma in cancer patients), and only a small fraction of cfDNA is tumor-derived [39]. Moreover, the tumor fraction of cfDNA varies between cancer types and even between metastatic patients with the same cancer type [40,41]. These variabilities and limited input material lead to the detection of ctDNA mutation being highly challenging, generating a higher false-negative rate in ctDNA analysis compared to tissue-based assays [17]. Similarly, false-positive findings as a result of biological factors such as non-tumor-derived clonal-hematopoiesis-related mutations present in plasma are a compelling issue that should be addressed to prevent misinterpretation of results [22,42,43,44]. The advantages and disadvantages of both plasma-based and tissue-based approaches stimulated an ongoing debate among researchers and clinicians as to whether a ‘plasma first’ or ‘tissue first’ approach is the most beneficial and appropriate genomic testing for advanced solid malignancies.
This review summarizes the recent advancements and supporting studies of the use of plasma ctDNA for genomic profiling in patients with advanced solid malignancies. In addition, we will also highlight the major challenges that should be tackled and factors that are to be considered to formulate evidence-based guidelines for the routine use of plasma-based genomic profiling in clinical settings.

2. Recent Advancements in ctDNA-Based Genomic Profiling Assays

Recent developments in sequencing technologies have increased the sensitivity of detecting the minute ctDNA present in plasma cfDNA with a higher level of accuracy and confidence (Table 1). Each method has its advantages and disadvantages. Polymerase chain reaction-based (PCR) methods such as BEAMing and droplet-digital PCR are fast, cost-effective, and simple to perform with extreme sensitivity and specificity of detecting mutations down to an allele frequency of 0.01% [28,45,46]. However, these target-specific approaches with limited multiplexing are only beneficial for the detection of a restricted number of known mutations, making them unsuitable for CGP of tumors. On the other hand, next-generation sequencing-based (NGS) approaches utilize multiplex PCR (amplicon-based) or hybridization capture to enrich and sequence the genomic regions of interest, enabling a more comprehensive analysis of the tumor genomic profile than the PCR-based methods. The larger NGS panel size also allows the evaluation of tumor mutation burden (TMB) and microsatellite instability (MSI), which are putative biomarkers for the response to immunotherapy [47,48]. However, due to the larger number of targets in NGS-based assays, the sensitivity of detection is often lower than the single-target approaches. Furthermore, errors acquired during NGS are one of the key contributing factors in limiting its accuracy and sensitivity to detect rare variants [49]. SafeSeq-S introduced the use of a unique identifier (UID) for error correction to increase the accuracy during sequencing. The UIDs are short sequences that are attached to each DNA template molecule that allows variant alleles present in the original sample to be distinguished from errors introduced during the template preparation and sequencing process [50]. The incorporation of UID has been shown to reduce the error rate by 70-fold [50]. The majority of ctDNA NGS assays available now incorporate unique identifiers, dual-indexing, and error suppression algorithms to increase the calling confidence of rare variants, thereby increasing their sensitivity and specificity [51,52]. These recent advancements, together with their supporting evidence, have encouraged the use of liquid biopsy for genomic profiling in clinical settings and received approval as in vitro companion diagnostics for molecular targeted therapies.

2.1. ctDNA Reflects the General Genomic Landscape of Tumors

Extensive work has been performed to compare the mutation detection concordance between tumor and ctDNA as an approach to evaluate the feasibility of utilizing ctDNA NGS-based assays for CGP of tumors in advanced cancer patients. The reported level of concordance by different studies has been greatly variable, ranging from 8.3% to 93% across different cancer types (Table 2). The differences in cohort size, study design, and definition of concordance may contribute to the variabilities observed. Despite these inconsistencies, the detection of oncogenic driver variants using NGS-based cfDNA assays has consistently demonstrated moderate to high sensitivity (75–93%) across studies in different cancer types [53,54,55,56,57,58,59]. In a large prospective, multicenter study that compared the detection of guideline-recommended biomarkers between a cfDNA-based and tissue-based assay in advanced and treatment-naïve non-small cell lung cancer (NSCLC), 80% of the genomic biomarkers detected from tumor tissues were concordantly detected from the plasma cfDNA [53]. The concordance level was observed to be 98.2% between tumor tissue and plasma for FDA-approved targets (EGFR exon 19 deletions and L858R, ALK fusions, BRAF V600E) [53]. A similar concordance level was observed for advanced breast cancer patients, where an overall concordance of up to 95% (negative and positive) was observed for the detection of mutations from the four major driver genes of breast cancer—PIK3CA, ESR1, AKT1, and HER2 [54]. A high level of concordance was also reported for patients with advanced gastrointestinal (GI) cancer, where the most common treatment-relevant biomarkers in GI cancers—KRAS, NRAS, and BRAF—showed near 100% ctDNA sensitivity compared to tissue-based CGP [59]. In a recent study that evaluated the genomic landscape detected in ctDNA and tissue-based from 837 metastatic castration-resistant prostate cancer (mCRPC) patients, 75.3% of short variants were concordantly detected between the two assays [58]. More importantly, up to 89.7% of BRCA1/2 alterations detected by tissue CGP were also detected from plasma cfDNA, highlighting the clinical utility of liquid biopsy CGP for the detection of clinically actionable alterations [58]. The high level of detection concordance for the major driver genes across different cancer types built the foundation for the use of ctDNA-based assay as an alternative to tissue-based CGP.
In addition to concordance analysis, several large cohort studies have been conducted in recent years to assess whether ctDNA NGS-based assays could benchmark against the current gold standard of tissue-based assays in detecting biomarkers for molecular targeted therapies. The detection of actionable alterations using ctDNA NGS-based assays has been consistent across studies, with approximately 40% of advanced cancer patients harboring at least one actionable target, comparable to tissue-based assays [59,60,61,62] (Table 2). In the NCI-MATCH study and the SHIVA study, actionable alterations were identified in approximately 40% of the evaluated patients using tissue-based CGP [60,62]. In one of the largest ctDNA studies conducted so far with over 10,000 advanced cancer patients, 41.2% of the patients were detected with at least one potential drug-sensitive target using a ctDNA-based CGP assay [40]. A comparable detection rate was observed in another pan-cancer study where 56% of recruited patients harbored clinically actionable alterations [63]. In the same study, the authors also demonstrated that the overall targetable alteration rate from ctDNA was similar to the patient-paired tissue [63]. Nakamura et al. also compared the identification of actionable alterations between two different GI advanced cancer cohorts that were either screened using ctDNA NGS (n = 1687) or using tumor tissues (n = 5621) [59]. The authors indicated that the detection rate was highly comparable in identifying targetable alterations from ctDNA and tissue (57.3% and 54.3% of patients, respectively) [59]. The variable actionable alteration detection rate was observed in different solid tumors (Table 2). Patients with advanced GI cancers showed the highest actionable alteration detection rate of 50%, followed by breast cancer and NSCLC at 38% and 36%, respectively (Table 2). In contrast, 30% of patients with prostate cancer harbor targetable alterations, and only less than 10% of thyroid and ovarian cancer patients were detected with actionable biomarkers using ctDNA-based CGP assays [40,58]. However, studies with larger cohort sizes are required to validate the detection rate of actionable alterations in rare cancer types. Several studies have also suggested that adding plasma NGS testing to the genomic profiling routine could increase targetable mutation detection by 48–75% and improve the delivery of targeted therapy in advanced cancer patients compared to the current standard approach [53,64].

2.2. Promising Clinical Outcomes following ctDNA Profiling for Treatment Selection

Recent clinical trials have incorporated exploratory objectives to evaluate the performance of ctDNA in identifying genomic markers for the prediction of treatment response for molecular targeted therapies. Several studies have shown patients with biomarkers of interest detected from cfDNA tend to exhibit a better prognosis than patients without the biomarker of interest, indicating their predictive value for treatment response [75,76,77,78,79]. In a prospective–retrospective study on archival plasma samples from the SoFEA and PALOMA3 trials, breast cancer patients with detected baseline ESR1 mutations from plasma had improved progression-free survival (PFS) after being treated with fulvestrant (estrogen receptor antagonist) compared with exemestane (aromatase inhibitor), while patients with wildtype ESR1 had similar PFS after receiving either treatment [75]. This was similarly observed in the SOLAR-1 study, where PIK3CA-mutated breast cancer, detected using plasma ctDNA, was associated with a better response to alpelisib plus fulvestrant than the fulvestrant arm [76]. The predictive value and clinical benefits of ctDNA genomic profiling have also been demonstrated in patients with carcinoma of unknown primary (CUP) [80]. CUP represents a heterogenous metastatic disease with an unidentifiable primary tumor where the standard treatments are often empiric chemotherapies with poor prognosis [81]. In the study conducted by Kato et al. 43% of the recruited 1931 CUP patients were detected with actionable alteration using plasma ctDNA CGP [80]. The authors also observed that patients treated with therapies with higher degrees of matching to ctDNA alterations showed better clinical outcomes [80].
The promising results from the exploratory studies suggested the potential of utilizing ctDNA CGP in clinical settings for genomic biomarker identification. The clinical benefits of ctDNA CGP were validated in recent large retrospective studies with cohort sizes of over 1000 patients. No significant differences were observed in the PFS and overall survival (OS) of patients selected based on ctDNA or tissues across several studies [59,66,68]. The clinical outcomes of liquid biopsy CGP compared to tissue CGP in advanced NSCLC patients were assessed in a multi-institutional, retrospective analysis of the real-world data [68]. The clinical and genomic data in this study were collected from a deidentified database where the majority of the patients were treated in a community setting. In this cohort of patients, a targetable genomic alteration was detected in 20% (188/937) of the cases that underwent ctDNA CGP compared to 22% (1215/5582) of tissue CGP cases. PFS for patients who received matched targeted therapy following liquid biopsy and tissue CGP were similar (13.8 vs. 10.6 months, respectively). Similarly, the overall response rate (partial/complete response) to matched targeted therapy was also comparable between post-liquid biopsy and post-tissue CGP (75% vs. 66%, respectively) [68].
In the past 2 years, several ongoing prospective phase II interventional clinical trials that were aimed to assess the accuracy and validity of ctDNA testing to select patients for genomic-directed therapies across different solid tumors have released their early results. All studies have shown over 99% of ctDNA sequencing success rates [54,82,83]. PlasmaMATCH is an open-label, multicohort trial of ctDNA testing in advanced breast cancer patients [54]. Recruited patients were subjected to ctDNA testing by NGS or droplet digital PCR and subsequently recruited into four parallel treatment cohorts matched to mutations (AKT1, ESR1, HER2, and PTEN) identified from plasma. A total of 34% of the sequenced patients had targetable mutations for cohort entry, and 13% of the patients entered one of the treatment arms. The HER2 and AKT1 arms reached the primary end point and exceeded the target number of responses where the response rate achieved by ctDNA-selection was comparable to that observed when guided by tissue testing [54]. However, the ESR1 and PTEN arms did not reach the target number of responses, with only 8% and 11% response rates, respectively, similar to that previously reported [54]. A similar open-label, multicohort study was conducted for advanced NSCLC patients [82]. In the BFAST study, 2219 patients were screened using ctDNA-based NGS for detection of ALK rearrangements [82]. In total, 5.4% of tested patients were ALK-positive, and 3.9% of patients were enrolled and received the mutation-matched treatment alectinib. The ALK-positive cohort met its primary end point with an overall response rate of 87.4%, comparable to previous reports using tissue-based profiling [82]. These results confirmed the clinical application of ctDNA-based CGP as a method to detect genomic biomarkers for treatment selection in advanced solid malignancies, reaching comparable clinical outcomes to tissue-based profiling.

2.3. Shorter Turnaround Time (TAT) with Improved Clinical Trial Enrolment Rate

The overall high sequencing success rate and fast turnaround time (TAT) of ctDNA-based CGP are some of the key advantages of liquid biopsy over tissue profiling. Several studies have compared the TAT from sample collection to reporting results between the two CGP approaches. The median TAT for ctDNA-based NGS is 9 days (ranging 2–15 days) compared to 15 days for tissue CGP (ranging 12–20 days) [54,58,59,67,70,82,84,85]. The additional time required for scheduling tissue biopsy and the procedure itself may contribute to the longer TAT observed with tissue-based CGP compared to an in-clinic, same-day blood collection for plasma ctDNA analysis [86]. The significantly shorter TAT of cfDNA screening may allow earlier initiation of treatments, which can be particularly beneficial for aggressive and fast progression cancer types. Furthermore, the more rapid TAT may also indirectly increase trial enrollment rates compared to tissue-based assays without compromising the treatment efficacy. In the study that evaluated the clinical trial enrollment in advanced GI cancer, ctDNA profiling significantly shortens the screening duration from 33 days to 11 days when compared with using tumor tissues, and the trial enrollment rate was also improved by more than 5% [59]. It has been suggested that more patients in the tissue-profiling cohort would need to start an empirical therapy while waiting for the results, whereas more patients in the ctDNA genotyping cohort had results available in time to inform the selection of molecular targeted therapies, thereby increasing the overall clinical trial enrolment rate [59]. The expected TAT for the currently commercially available ctDNA CGP assays is within 7–10 days [87,88], which coincides with the current observations. However, the current TAT can potentially be further shortened with a more flexible and decentralized sequencing system, which can be placed at the point of care and operated with minimal technical supervision [89,90,91]. Such an automated NGS system would need further analytical and clinical validation for its use in clinical settings.

2.4. FDA Approval of Multigene ctDNA NGS Tests for CGP and as In Vitro Companion Diagnostics

In 2016, the U.S. Food and Drug Administration (FDA) approved the first ctDNA plasma-based genomic testing as a companion diagnostic for the detection of EGFR mutations to identify NSCLC patients that are eligible for erlotinib [92]. The Cobas EGFR Mutation Test v2 utilizes the RT-PCR technology, reaching a detection sensitivity of 0.1–0.8% [93] (Table 3) [93]. The approval of EGFR ctDNA testing provided a rapid and noninvasive method to detect clinically relevant genomic markers for treatment selection and has been proven to be reliable in clinical settings [94,95,96]. However, RT-PCR-based methods limit the number of testing targets and restrict their clinical applications. In 2020, the FDA approved two CGP liquid biopsy tests, Guardant360 CDx and FoundationOne Liquid CDx, for detecting genomic alterations from 55 and 311 genes, respectively (Table 3) [97,98]. Both panels were approved as complementary diagnostics for tumor mutation profiling in patients diagnosed with solid malignancy. The genomic findings from the ctDNA-CGP panels are to be used for treatment selection following professional guidelines [30]. Guardant360 CDx and FoundationOne Liquid CDx also received FDA approval as companion diagnostics for several molecular targeted therapies (Table 3) [99,100]. The number of companion diagnostic indications for both assays has increased since their initial approval and would likely continue to expand with the accumulation of evidence for other targeted therapies. The detection sensitivity for the approved targets using the Guardant360 CDx ranged from 0.2–0.5% [99], and 0.24–0.51% for the FoundationOne Liquid CDx [100]. In particular, to EGFR mutations, both approved ctDNA-CGP panels could not achieve the same level of sensitivity as the RT-PCR-based Cobas system, highlighting the difficulty to maintain the high sensitivity of mutation detection in large multiplexing systems.

3. Limitations and Challenges for the Routine Use of ctDNA-Based CGP for Treatment Selection

As we highlighted in Section 2.1 of this review, the reported level of concordance between tumor and plasma-based NGS analyses across studies has been greatly variable. Technical and biological factors which account for the generation of false-positive and false-negative results may contribute to the discordance observed. These factors remain the key limitations and challenges for the routine use of ctDNA profiling in clinical settings [17,101]. Furthermore, the lack of comprehensive guidelines to recommend the usage of ctDNA CGP also restricts its clinical use.

3.1. Technical Limitations Leading to False Positive and Negative Results

The major technical challenges that ctDNA NGS assays face are the small fragment size of cfDNA (~160 bps) and the low concentrations of ctDNA present in the blood. The detection of rare somatic mutations from such limited genomic material input is highly challenging [39]. Target enrichment, either by hybrid capture or amplicon methods, with extensive PCR amplification, is generally required to successfully capture the tumor genomic profile from the small quantity of cfDNA [102,103]. However, the small size of cfDNA fragments can restrict target enrichment and reduce the accuracy of alignment to the human reference genome [39,104]. These NGS chemical and physical factors can exacerbate biases and sequencing errors, resulting in both false-positive and false-negative results [105]. The majority of the current ctDNA NGS systems incorporate error-suppression strategies such as molecular barcodes or bioinformatic analyses. However, technical false positives remain [39,106]. In the study conducted by Stetson et al. the authors evaluated the false positive (FP) rates of four NGS gene panels using replicate sets of 24 plasma samples [106]. The positive predictive value ranged from 36 to 80% across the four vendors, and the majority of the FP variants occurred at less than variant allele frequency (VAF) of 1% [106]. The FP calls identified in the study were enriched for assay-specific mutational biases and mostly were novel variants not found in somatic variant databases [106]. In the study led by the Oncopanel Sequencing Working group, an average of 1.65–5.3 FP variants were observed per replicate across five leading ctDNA NGS assays [39]. The erroneous variants occurred almost exclusively at VAF less than 0.5% [39]. Despite the FP rate observed, the authors of this study concluded that the sensitivity, rather than precision, was the major determinant of discordance observed [39].
The current reported limit of detection by different ctDNA-targeted NGS platforms ranged from 0.004 to 2% [107]. However, the reproducibility and accuracy of alterations detected at low VAF have been variable across different platforms [39,106]. Similar to the FPs, the sensitivity of ctDNA assays drops significantly for mutations with low VAF [39,106]. Most of the commonly used ctDNA assays, both amplicon-based or hybrid capture, were highly sensitive for variants at high frequencies (over 90% sensitivity for VAF > 0.5%), but the sensitivity drops to 40% for alterations at VAF less than 0.5% [106]. This observation has been similarly reflected in clinical studies. In contrast to the high level of concordance observed in the major driver mutations, several studies have reported that the sensitivity of ctDNA mutation detection is dependent on their clonal fraction in the tumor tissue [42,59,63,108]. In an exploratory study conducted by Razavi et al. 72% of the genomic alterations detected from tumor biopsies were concordantly detected using an NGS-based ctDNA assay with no significant differences in the sensitivity across metastatic breast cancer, NSCLC, or castration-resistant prostate cancer [42]. However, the ctDNA detection rate was significantly higher for clonal mutations (75–90%) than subclonal mutations (20–30%) [42]. A similar disparity was observed in a large-scale study conducted on advanced GI cancer, where the positive predictive value was markedly higher for clonal alterations than for subclonal mutations (80.3 vs. 8.3%) [59]. Growing evidence indicates that subclonal mutations present in a small subset of tumor cells may confer resistance to targeted therapies [109,110,111,112]; therefore, improving the ability to identify these pre-existing resistance mutations from ctDNA is crucial to enhance the implementation of precision medicine. Recent studies suggest the enrichment of shorter fragment lengths, either using in vitro or in silico methods, may improve the detection of alterations with low VAF [113,114,115]. However, most of the studies were conducted with small sample sizes, and further studies are required to confirm the clinical implications of this approach.
The detection of targetable fusions or gene rearrangements from cfDNA remains technically challenging with inconsistent sensitivity across studies owing to the low prevalence of fusions in common solid tumors. To address this, Esagian et al. compiled 38 published studies that assessed the concordance of fusion detection between tumor tissues and plasma cfDNA in NSCLC patients [116]. A total of 1141 patients were included in the systematic review, and less than 60% of the samples with ALK, RET, and ROS1 fusion from tumor tissues were concordantly detected from plasma cfDNA [116]. Most of the fusions arise from inter-chromosomal or intra-chromosomal conjunction of different introns, where the intronic regions can be extremely large with repetitive sequences (especially ROS1 and NTRK) [117,118]. The inclusion of large intronic breakpoints as target regions may improve the detection of gene fusions; however, it may also increase the sequencing costs with reduced sequencing efficiency [117,118]. Current ctDNA-based CGP assays have focused on balancing the overall costs and the number of target probes to optimize the detection rate. FoundationOne Liquid assay included a selected number of introns for 9 of 16 targetable fusion kinase genes and was observed to have an overall concordance rate of 70% compared to tumor tissues from patients with various solid tumors [119]. Previous studies have also shown that assay optimizations such as the use of shorter amplicons or capture probes, primer extension, variant calling, and bioinformatic filtering may enhance the detection of fusions [120,121]. In contrast to cfDNA, cfRNA-based assays are not affected by intronic regions and only identify expressed fusion genes [117]. In a recent exploratory study, the authors demonstrated that a cfRNA-based NGS assay has an overall higher sensitivity to detect ALK, ROS1, and RET fusions than a cfDNA-based NGS system (78% and 33%, respectively) [122]. Furthermore, amplicon-based NGS panels that analyze gene fusions at the ctRNA level and gene alterations at the ctDNA level may also hold promise in improving fusion detection [89,123]. Larger studies are required to evaluate and confirm whether these strategies are beneficial in detecting fusions from plasma cell-free nucleic acids in the clinical setting.

3.2. Biological Limitations: Low Tumor Shedding and Non-Tumoral Origin of cfDNA

Besides the technical limitations, biological factors such as the location, size, and vascularity of the tumor may affect the release of ctDNA into the circulation, thereby compromising its detectability [124]. ctDNA fraction (the fraction of tumor-derived cfDNA) can vary significantly according to the tumor type and even between patients with the same tumor type (ranging from less than 1% to 80%) [40,42,125,126,127]. Tumors from the brain, renal, and thyroid have repeatedly been observed to have a lower ctDNA detection compared to colorectal, lung, and breast cancer, even at an advanced stage [40,125]. Furthermore, several studies have also reported that the detection rate of ctDNA is significantly higher in colorectal cancer patients with liver metastasis compared to nodal or lung metastasis [128,129,130]. The performance of ctDNA NGS assays was found to be highly dependent on the ctDNA fraction, particularly for detecting gene amplification [42,58,131]. For cfDNA samples with high tumor fractions (20–35%), 51–89% of copy number variations (CNVs) from tumor tissues were concordantly detected using ctDNA [58,132]. However, the sensitivity to detect CNVs from ctDNA drops to 28–35% for samples with low ctDNA fraction [58,132]. These biological factors should be taken into consideration when interpreting negative results from ctDNA CGP.
Non-tumoral variants detected from plasma contribute to the false-positive results detected from ctDNA-based CGP. Clonal hematopoiesis (CH) is a normal process of aging with the accumulation of somatic mutations in hematopoietic cells [22]. The detection of these non-tumor-derived CH mutations in plasma has been repeatedly reported as a source of biological noise to ctDNA genomic profiling [42,44,133,134,135]. Previous studies have shown that 15–53% of alterations detected from cfDNA of advanced cancer patients had features consistent with CH [42,43,133,136]. Moreover, a substantial number of CH variants detected from cfDNA are considered to be oncogenic and are indicated for targeted therapies, including mutations from KRAS, EGFR, and PIK3CA [42]. The VAF of CH mutations detected from plasma is indifferent to the tumor-derived mutations [44]. These features highlight the difficulties in distinguishing between CH and tumor-derived mutations and the risk of false findings. The significance of CH mutations in ctDNA CGP has been well recognized and acknowledged by researchers and clinicians. However, currently approved ctDNA-based CGP assays do not differentiate or report the origin of the mutations detected. This should be urgently addressed to prevent the initiation of inappropriate treatments as a result of false findings from ctDNA profiling.

3.3. Lack of Standardized Evidence-Based Guidelines for Tissue or Plasma First Approach

Due to the continuously growing evidence and recognition of the clinical use of ctDNA-based CGP for treatment selection, several leading professional organizations have included recommendations for the use of liquid biopsy in their clinical management guidelines. However, these recommendations were limited to NSCLC, breast cancer and prostate cancer patients. Furthermore, different multidisciplinary bodies also released contradicting suggestions as to whether a ‘plasma-first’ or a ‘tissue-first’ approach should be adopted in clinical settings. For the management of advanced breast cancer, both National Comprehensive Cancer Network (NCCN) and The European Society for Medical Oncology (ESMO) recommend ctDNA profiling as an alternative and complement to tissue CGP, while The American Society of Clinical Oncology (ASCO) recommends cfDNA as the specimen-of-choice for CGP [137,138,139]. Similarly, NCCN, ASCO and ESMO all recommend a ‘tissue-first’ approach during the initial diagnosis of NSCLC patients, while the International Association for the Study of Lung Cancer (IASLC) recommends the use of ctDNA CGP as the assay of choice in their latest consensus statement [140,141,142,143]. Both tissue-based and ctDNA-based CGP have their advantages and disadvantages, making it difficult to adopt a one-size-fits-all approach in the clinical setting. In this section, based on the accumulated evidence and recommendations from multidisciplinary expert panels, we have summarized some of the key factors that should be considered to select the most appropriate approach and proposed a generalized guideline for assay selection under different situations (Figure 2).

3.3.1. Availability of Excision Tumor Tissue, Quality and Quantity of Biopsy; the Presence of Multiple Lesions; Cancer Types

Based on the observations from the current studies, the false-negative rate of plasma samples is higher than that of tissue-based assays [144]. In cases where surgical resection is performed and excision tumor tissues are available, tissue-based CGP would be preferred to overcome the lower sensitivity issue of ctDNA assays. This is also supported by the majority of the expert panels (NCCN, ESMO and ASCO), where ctDNA-based CGP is not recommended in the initial diagnosis setting [141,142,143]. However, other factors should also be considered to choose the most appropriate specimen for CGP. In cases where surgical resection is not feasible and only biopsy samples are available, the timing and condition of the patient are crucial for assay selection. In cases where biopsy samples are difficult to retrieve and the quality and quantity of tissue specimens are insufficient for tissue-based CGP, liquid biopsy should be considered the assay of choice. Furthermore, if patients present with multiple lesions or metastases, liquid biopsy should be preferred due to its ability to detect intertumoral heterogeneity, which could be missed by a single tissue testing [145,146,147]. The detection of spatial tumor heterogeneity was highlighted in a study that sequenced paired primary tumor, metastatic tissue, and plasma cfDNA of breast cancer patients [146]. Plasma cfDNA detected up to 97% of alterations from primary and metastatic tissues, and 13 of the variants in metastatic tumors were exclusively detected from ctDNA, and not in the corresponding primary tumors [146]. However, currently approved ctDNA CGP assays would not be able to identify the origin of the mutations (inter- or intratumoral heterogeneity) without previous knowledge of the tumor genomic profile. Future studies exploring the use of methylation and fragmentomic features of cfDNA may help identify their cellular origins [148,149]. The shorter TAT of cfDNA-based CGP also suggests that liquid biopsy would be more beneficial than tissue profiling for aggressive and fast progression cancers, allowing earlier treatment commencement [144].

3.3.2. Timing: Initial Diagnosis or Recurrence/Progression Disease

At initial diagnosis, tissue-based CGP assays are likely to be more beneficial than ctDNA profiling for treatment-naïve advanced cancer patients with resectable tumors, owing to the lower sensitivity of ctDNA-based assays. However, for treatment selection at the time of recurrence or during disease progression, ctDNA-based genomic profiling should be preferred [144]. Several studies have shown that the longer collection interval between plasma and tissues leads to higher discordance [63,65,70,130,150,151]. The most striking area of discordance between liquid and tissue CGP is often the detection of a range of resistance mutations from liquid biopsy [58,152,153]. In a study that evaluated the detection of androgen receptor (AR)-activating alterations in prostate cancer, for samples that were collected more than 30 days apart and who had previous exposure to AR signaling inhibitors during the collection interval, only 5% of the AR short variants detected from plasma were concordantly detected from tumor tissues. This highlights the ability of liquid biopsy to detect resistance variants that may not be detected from archival tumor tissues and could provide additional ability to identify patients who might benefit from a non-AR signaling inhibitor [58]. Similarly, an increased discordance in the drivers of resistance to anti-EGFR therapy: KRAS, NRAS, and EGFR mutations were observed in metastatic colorectal cancer patients who were treated with anti-EGFR therapy than the treatment-naïve patients (concordance rate of 71% and 94%, respectively) [153]. The clinical benefits of the ctDNA-based assay over tissue-based CGP in detecting resistance mutations at the point of disease progression are also recognized by NCCN, ESMO, and IASLC, where initial use of ctDNA testing for EGFR-T790M alterations is preferred in patients that have developed progression from EGFR tyrosine kinase inhibitors (TKIs) [140,141,143]. The ability of ctDNA profiling to capture temporal tumor heterogeneity highlights the advantage of liquid biopsy over tissue biopsy in patients who have developed recurrence or received previous therapies. However, for cancer types with known low ctDNA tumor fraction and poor ctDNA detection sensitivity (e.g., brain, renal, thyroid, and colorectal cancer with lung metastasis), conventional tissue-based CGP or single-gene cfDNA assays with higher assay sensitivity should be considered instead [144].

4. Future Perspectives of ctDNA-Based CGP to Maximize Its Utilities in Personalizing Oncology Management

The use of liquid biopsy is increasingly being incorporated into the clinical protocol for targeted treatment guidance; however, there are still several areas that require further research. Here, we provide our perspectives on the key challenges that should be attended to optimize the use of ctDNA-based CGP in oncology management.

4.1. Standardizing Methods to Exclude CH Mutations

One of the main challenges for the clinical use of ctDNA-based CGP is the lack of standardized methods to determine the origin of the alterations detected from plasma and the exclusion of CH-related mutations. Most of the studies conducted so far utilize paired-sequencing of the matched white blood cells to a comparable depth as cfDNA to filter out CH mutations [42,44,154]. This approach remains useful; however, it incurs additional costs, which hampers its practicality in clinical settings. Alterations detected from cfDNA may also be validated using white blood cells with single-gene assays such as droplet digital PCR. Single probe assays often have higher sensitivity with lower running costs than NGS; however, additional validation assays would prolong TAT and delay the initiation of treatments. In contrast to validating using white blood cells, recent studies have focused on utilizing cfDNA fragmentomic analysis to differentiate and determine the origin of cfDNA mutations [22,134,135,155,156]. Several studies have shown that ctDNA presents as shorter fragments than CH or non-mutated cfDNA fragments, which might be useful for distinguishing the tumor-derived mutations [63,135,155,156]. More importantly, fragment size distribution can be determined without additional sequencing or validation assays, thereby minimizing costs and time, making it ideal for clinical implementations. However, current observations are based on proof-of-concept studies with small sample sizes. Larger studies are required to confirm the clinical validity. Besides the fragment size of cfDNA, the fragmentation pattern, which includes the nucleotide motifs at the fragment ends, single-stranded jagged ends, and the genomic locations of the fragmentation endpoints, has been suggested to relate to the tissue of origin [149]. It is currently unclear whether these unique characteristic signatures can be employed to distinguish tumor-derived mutations. Future studies should evaluate and determine the most appropriate and economical method to exclude non-tumor-derived alterations.

4.2. Establishing the VAF Threshold for Treatment Initiation

Current technological developments have been mainly focusing on improving the limit of detection to improve the ctDNA detection sensitivity. However, limited research has been conducted to evaluate the clinical outcomes of targeting alterations that are detected at low VAF using ctDNA profiling. There are no cutoff values or thresholds from the guidelines of the approved companion diagnostics to help guide clinicians on whether treatments should be initiated based on the reported VAF. It is unclear whether targeting alterations detected at low VAF from plasma could result in clinical benefits. Two recent exploratory studies have observed no significant differences in treatment response between NSCLC patients detected with EGFR mutations below or above ctDNA VAF of 1% [66,67]. The authors from both studies also reported that a trend of greater clinical benefit was observed in those with a low VAF (<1%), suggesting better disease control in those patients with lower tumor burden as reflected by ctDNA [66,67]. On the other hand, EGFR clonal dominance determined by plasma cfDNA was observed to be independently associated with improved efficacy of EGFR-TKIs in patients with advanced NSCLC [157]. In the study conducted by Ai et al. the authors employed a hierarchical Bayesian clustering method to analyze the clonal structure in ctDNA and evaluated whether the actionable EGFR mutation was the dominant clone across 300 treatment-naïve advanced NSCLC patients [157]. The objective response rate and PFS were significantly higher for patients with EGFR as a dominant clone than those nondominant clones, according to plasma ctDNA NGS results [157]. The authors suggested that the ctDNA VAF normalized using a statistical model might be a more stable parameter for guiding therapeutic strategies based on ctDNA results [157].

4.3. Frequency of ctDNA CGP for Treatment Optimization

A series of observational and interventional clinical trials have demonstrated that monitoring of clonal dynamics and the development of resistance mutations using serial ctDNA analysis may assist in treatment optimization. However, the frequency of ctDNA monitoring and optimal sampling timepoints to achieve maximal clinical benefit remains unclear. Previous studies have shown that ctDNA CGP before the initialization of re-challenge therapy could be effective in predicting clinical benefit [158,159]. In a phase II study, patients with wild-type RAS/BRAF ctDNA before initialization of anti-EGFR re-challenge have a significantly longer OS compared to patients with mutated ctDNA (17.3 and 10.4 months, respectively) [158]. Similarly, in a recent single-arm interventional clinical trial, metastatic colorectal patients who have developed resistance to anti-EGFR monoclonal antibodies were screened for wild-type RAS/BRAF/EGFR using ctDNA for selection of re-challenge therapy [159]. The primary endpoint of the clinical trial was met with an overall disease control rate of 59%, corroborating the effectiveness of ctDNA CGP in selecting patients for re-challenge therapy [159]. Furthermore, Parseghian et al. have demonstrated that ctDNA evaluation at 4.4 months after the cessation of anti-EGFR therapy may be the optimal timing to assess the regression of resistant RAS/BRAF/EGFR clones and to guide the initiation of anti-EGFR re-challenge therapy for maximal clinical benefits [160].
Early detection of resistance mutations through serial ctDNA analysis during treatment has been suggested as an indicator for treatment intervention to prevent or delay tumor progression [161,162,163,164]. The ability of ctDNA to detect the emergence of resistance mutations and prediction of recurrence has been well reported; however, the frequency of ctDNA monitoring has been variable across studies, ranging between fortnightly to every 3 months [161,162,163,164]. The feasibility of preventing or delaying tumor progression via ctDNA monitoring was first evaluated in the recent phase III PADA-1 trial. Metastatic breast cancer patients receiving first-line treatment with palbociclib plus aromatase inhibitor therapy were monitored using ctDNA every 2 months and were switched from an aromatase inhibitor to fulvestrant as soon as an ESR1 mutation became detectable from ctDNA [163]. The early results from the trial indicated that patients who switched to fulvestrant co-treatment showed a 39% reduction in the risk of disease progression or death with a PFS of 11.9 months compared with 5.7 months in patients that maintained the aromatase inhibitor co-treatment [163]. The results from the trial highlighted the clinical benefits of ctDNA monitoring for early detection of resistance mutations to personalize and modify treatment regimens.
The use of a large cfDNA panel or a targeted approach to longitudinally monitor patients with advanced-stage disease should also be further investigated. It has been suggested that large ctDNA CGP panels may be more beneficial in cases where resistance mechanisms of the drugs are not known, while in settings where resistance mechanisms are well described, longitudinal ctDNA monitoring using a targeted approach may be more appropriate and cost-effective [30]. Large studies with health economic benefit assessments are required to facilitate the smooth translation of ctDNA monitoring into the clinical setting.

4.4. ctDNA Biomarkers for Immunotherapy

Tumor mutation burden (TMB) and microsatellite instability (MSI) have shown to be effective genomic biomarkers in identifying patients who are likely to benefit from immune checkpoint inhibitors [165]. However, the insufficiency and poor quality of tissue sampling prevented TMB and MSI testing from being performed regularly in the current clinical setting [166]. ctDNA CGP using large NGS panels may overcome these shortcomings and may serve as a detection tool for prognostic and predictive biomarkers for immunotherapy [47,48]. Modest but consistent level of correlation (mean of R = 0.6) between TMB determined from tissues (tTMB) or ctDNA (bTMB) has been reported across different studies [47,167,168,169,170]. Tumor heterogeneity and low ctDNA tumor fraction from blood may account for the absence of a higher level of concordance between tTMB and bTMB [47,170,171]. Nevertheless, similar to tTMB, bTMB was found to be predictive of immunotherapy outcomes [170,172]. In a meta-analysis study that evaluated the results from 6 randomized clinical trials with a total of 2338 advanced NSCLC patients who were treated with PD-1/PD-L1 inhibitors, patients with high bTMB showed significantly better OS, PFS, and objective response rates from immunotherapy than patients with low bTMB [172]. In contrast to bTMB, ctDNA CGP has shown high sensitivity (78–87%) in detecting MSI compared to tumor tissues [173,174,175]. Patients detected with MSI using ctDNA assays also demonstrated significantly prolonged PFS, confirming their potential clinical validity [173,174,175]. The accumulating observations should be validated in large cohort studies, and future studies should emphasize the standardization of bTMB and MSI assays and determine a validated threshold to accelerate their translation to the clinics.

5. Conclusions

Research developments and the accumulation of analytical and clinical evidence for the use of ctDNA genomic profiling from the past decade have transformed our clinical practice in oncology. The approval of ctDNA-based assays for CGP and as companion diagnostic tools have allowed more cancer patients to gain access to targeted therapies and supported the realization of precision oncology. Developing evidence-based guidelines for the use of ctDNA profiling and addressing the current limitations, such as the exclusion of CH alterations, will further optimize the clinical usage of liquid biopsy for treatment selection. Future studies should focus on expanding the current roles of plasma ctDNA to improve patient access to precision medicine and thereby improve patient outcomes.

Author Contributions

Conceptualization, H.T.C. and S.-K.L.; Writing—original draft preparation, H.T.C.; Writing—review and editing, H.T.C., Y.M.C. and S.-K.L. All authors have read and agreed to the published version of the manuscript.

Funding

This review was supported by the Council for Science, Technology and Innovation (CSTI), cross-ministerial Strategic Innovation Promotion Program (SIP), and “Innovative AI Hospital System” (Funding Agency: National Institute of Biomedical Innovation, Health and Nutrition (NIBIOHN), Grant number: SIPAIH18C03).

Conflicts of Interest

Y.M.C. reported as an employee of Cancer Precision Medicine Inc., Japan. S.-K.L. reported consulting or advisory roles with Cancer Precision Medicine Inc., Japan.

References

  1. Chakravarty, D.; Solit, D.B. Clinical cancer genomic profiling. Nat. Rev. Genet. 2021, 22, 483–501. [Google Scholar] [CrossRef] [PubMed]
  2. Malone, E.R.; Oliva, M.; Sabatini, P.J.B.; Stockley, T.; Siu, L.L. Molecular profiling for precision cancer therapies. Genome Med. 2020, 12, 8. [Google Scholar] [CrossRef] [Green Version]
  3. Al-Kateb, H.; Nguyen, T.T.; Steger-May, K.; Pfeifer, J.D. Identification of major factors associated with failed clinical molecular oncology testing performed by next generation sequencing (NGS). Mol. Oncol. 2015, 9, 1737–1743. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Tomlins, S.A.; Hovelson, D.H.; Suga, J.M.; Anderson, D.M.; Koh, H.A.; Dees, E.C.; McNulty, B.; Burkard, M.E.; Guarino, M.; Khatri, J.; et al. Real-World Performance of a Comprehensive Genomic Profiling Test Optimized for Small Tumor Samples. JCO Precis. Oncol. 2021, 5, 1312–1324. [Google Scholar] [CrossRef] [PubMed]
  5. Uozu, S.; Imaizumi, K.; Yamaguchi, T.; Goto, Y.; Kawada, K.; Minezawa, T.; Okamura, T.; Akao, K.; Hayashi, M.; Isogai, S.; et al. Feasibility of tissue re-biopsy in non-small cell lung cancers resistant to previous epidermal growth factor receptor tyrosine kinase inhibitor therapies. BMC Pulm. Med. 2017, 17, 175. [Google Scholar] [CrossRef] [Green Version]
  6. Chouaid, C.; Dujon, C.; Do, P.; Monnet, I.; Madroszyk, A.; Le Caer, H.; Auliac, J.B.; Berard, H.; Thomas, P.; Lena, H.; et al. Feasibility and clinical impact of re-biopsy in advanced non small-cell lung cancer: A prospective multicenter study in a real-world setting (GFPC study 12-01). Lung Cancer 2014, 86, 170–173. [Google Scholar] [CrossRef] [PubMed]
  7. Overman, M.J.; Modak, J.; Kopetz, S.; Murthy, R.; Yao, J.C.; Hicks, M.E.; Abbruzzese, J.L.; Tam, A.L. Use of research biopsies in clinical trials: Are risks and benefits adequately discussed? J. Clin. Oncol. 2013, 31, 17–22. [Google Scholar] [CrossRef]
  8. Kelly, R.J.; Turner, R.; Chen, Y.-W.; Rigas, J.R.; Fernandes, A.W.; Karve, S. Complications and Economic Burden Associated with Obtaining Tissue for Diagnosis and Molecular Analysis in Patients with Non–Small-Cell Lung Cancer in the United States. J. Oncol. Pract. 2019, 15, e717–e727. [Google Scholar] [CrossRef]
  9. Wu, C.C.; Maher, M.M.; Shepard, J.-A.O. Complications of CT-Guided Percutaneous Needle Biopsy of the Chest: Prevention and Management. Am. J. Roentgenol. 2011, 196, W678–W682. [Google Scholar] [CrossRef]
  10. Nam, B.D.; Yoon, S.H.; Hong, H.; Hwang, J.H.; Goo, J.M.; Park, S. Tissue Adequacy and Safety of Percutaneous Transthoracic Needle Biopsy for Molecular Analysis in Non-Small Cell Lung Cancer: A Systematic Review and Meta-analysis. Korean J. Radiol. 2021, 22, 2082–2093. [Google Scholar] [CrossRef]
  11. Jekunen, A.P. Role of Rebiopsy in Relapsed Non-Small Cell Lung Cancer for Directing Oncology Treatments. J. Oncol. 2015, 2015, 809835. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Russano, M.; Napolitano, A.; Ribelli, G.; Iuliani, M.; Simonetti, S.; Citarella, F.; Pantano, F.; Dell’Aquila, E.; Anesi, C.; Silvestris, N.; et al. Liquid biopsy and tumor heterogeneity in metastatic solid tumors: The potentiality of blood samples. J. Exp. Clin. Cancer Res. 2020, 39, 95. [Google Scholar] [CrossRef] [PubMed]
  13. Alix-Panabières, C. The future of liquid biopsy. Nature 2020, 579, S9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Crocetto, F.; Barone, B.; Ferro, M.; Busetto, G.M.; La Civita, E.; Buonerba, C.; Di Lorenzo, G.; Terracciano, D.; Schalken, J.A. Liquid biopsy in bladder cancer: State of the art and future perspectives. Crit. Rev. Oncol. 2022, 170, 103577. [Google Scholar] [CrossRef]
  15. Crocetto, F.; Cimmino, A.; Ferro, M.; Terracciano, D. Circulating tumor cells in bladder cancer: A new horizon of liquid biopsy for precision medicine. J. Basic Clin. Physiol. Pharmacol. 2021. [Google Scholar] [CrossRef]
  16. Heitzer, E.; Ulz, P.; Geigl, J.B. Circulating Tumor DNA as a Liquid Biopsy for Cancer. Clin. Chem. 2015, 61, 112–123. [Google Scholar] [CrossRef]
  17. Keller, L.; Belloum, Y.; Wikman, H.; Pantel, K. Clinical relevance of blood-based ctDNA analysis: Mutation detection and beyond. Br. J. Cancer 2020, 124, 345–358. [Google Scholar] [CrossRef]
  18. Kustanovich, A.; Schwartz, R.; Peretz, T.; Grinshpun, A. Life and death of circulating cell-free DNA. Cancer Biol. Ther. 2019, 20, 1057–1067. [Google Scholar] [CrossRef] [Green Version]
  19. Schwarzenbach, H.; Hoon, D.S.B.; Pantel, K. Cell-free nucleic acids as biomarkers in cancer patients. Nat. Rev. Cancer 2011, 11, 426–437. [Google Scholar] [CrossRef]
  20. Heitzer, E.; Auinger, L.; Speicher, M.R. Cell-Free DNA and Apoptosis: How Dead Cells Inform About the Living. Trends Mol. Med. 2020, 26, 519–528. [Google Scholar] [CrossRef]
  21. Lui, Y.Y.; Dennis, Y.M. Circulating DNA in plasma and serum: Biology, preanalytical issues and diagnostic applications. Clin. Chem. Lab. Med. 2002, 40, 962–968. [Google Scholar] [CrossRef] [PubMed]
  22. Chan, H.T.; Chin, Y.M.; Nakamura, Y.; Low, S.-K. Clonal Hematopoiesis in Liquid Biopsy: From Biological Noise to Valuable Clinical Implications. Cancers 2020, 12, 2277. [Google Scholar] [CrossRef] [PubMed]
  23. 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]
  24. Anker, P.; Stroun, M.; Maurice, P.A. Spontaneous release of DNA by human blood lymphocytes as shown in an in vitro system. Cancer Res. 1975, 35, 2375–2382. [Google Scholar] [PubMed]
  25. Lui, Y.Y.N.; Chik, K.-W.; Chiu, R.W.K.; Ho, C.-Y.; Lam, C.W.K.; Lo, Y.M.D. Predominant hematopoietic origin of cell-free DNA in plasma and serum after sex-mismatched bone marrow transplantation. Clin. Chem. 2002, 48, 421–427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Moss, J.; Magenheim, J.; Neiman, D.; Zemmour, H.; Loyfer, N.; Korach, A.; Samet, Y.; Maoz, M.; Druid, H.; Arner, P.; et al. Comprehensive human cell-type methylation atlas reveals origins of circulating cell-free DNA in health and disease. Nat. Commun. 2018, 9, 5068. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Lam, W.K.J.; Gai, W.; Sun, K.; Wong, R.S.; Chan, R.W.; Jiang, P.; Chan, N.P.; Hui, W.I.; Chan, A.; Szeto, C.-C.; et al. DNA of Erythroid Origin Is Present in Human Plasma and Informs the Types of Anemia. Clin. Chem. 2017, 63, 1614–1623. [Google Scholar] [CrossRef]
  28. Elazezy, M.; Joosse, S.A. Techniques of using circulating tumor DNA as a liquid biopsy component in cancer management. Comput. Struct. Biotechnol. J. 2018, 16, 370–378. [Google Scholar] [CrossRef]
  29. Bellosillo, B.; Montagut, C. High-accuracy liquid biopsies. Nat. Med. 2019, 25, 1820–1821. [Google Scholar] [CrossRef]
  30. Ignatiadis, M.; Sledge, G.W.; Jeffrey, S.S. Liquid biopsy enters the clinic—Implementation issues and future challenges. Nat. Rev. Clin. Oncol. 2021, 18, 297–312. [Google Scholar] [CrossRef]
  31. Alix-Panabières, C.; Pantel, K. Clinical Applications of Circulating Tumor Cells and Circulating Tumor DNA as Liquid Biopsy. Cancer Discov. 2016, 6, 479–491. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Oliveira, K.C.S.; Ramos, I.B.; Silva, J.M.C.; Barra, W.F.; Riggins, G.J.; Palande, V.; Pinho, C.T.; Frenkel-Morgenstern, M.; Santos, S.E.; Assumpcao, P.P.; et al. Current Perspectives on Circulating Tumor DNA, Precision Medicine, and Personalized Clinical Management of Cancer. Mol. Cancer Res. 2020, 18, 517–528. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Vendrell, J.A.; Quantin, X.; Aussel, A.; Solassol, I.; Serre, I.; Solassol, J. EGFR-dependent mechanisms of resistance to osimertinib determined by ctDNA NGS analysis identify patients with better outcome. Transl. Lung Cancer Res. 2021, 10, 4084–4094. [Google Scholar] [CrossRef]
  34. Nakajima, H.; Kotani, D.; Bando, H.; Kato, T.; Oki, E.; Shinozaki, E.; Sunakawa, Y.; Yamazaki, K.; Yuki, S.; Nakamura, Y.; et al. REMARRY and PURSUIT trials: Liquid biopsy-guided rechallenge with anti-epidermal growth factor receptor (EGFR) therapy with panitumumab plus irinotecan for patients with plasma RAS wild-type metastatic colorectal cancer. BMC Cancer 2021, 21, 674. [Google Scholar] [CrossRef] [PubMed]
  35. Van Helden, E.J.; Angus, L.; Menke-van der Houven van Oordt, C.W.; Heideman, D.V.M.; Boon, E.; van Es, S.C.; Radema, S.A.; van Herpen, C.M.L.; de Groot, D.J.A.; de Vries, E.G.E.; et al. RAS and BRAF mutations in cell-free DNA are predictive for outcome of cetuximab monotherapy in patients with tissue-tested RAS wild-type advanced colorectal cancer. Mol. Oncol. 2019, 13, 2361–2374. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Guo, S.; Ye, Y.; Liu, X.; Gong, Y.; Xu, M.; Song, L.; Liu, H. Intra-Tumor Heterogeneity of Colorectal Cancer Necessitates the Multi-Regional Sequencing for Comprehensive Mutational Profiling. Cancer Manag. Res. 2021, 13, 9209–9223. [Google Scholar] [CrossRef] [PubMed]
  37. Holdhoff, M.; Schmidt, K.; Donehower, R.; Diaz, L. Analysis of Circulating Tumor DNA to Confirm Somatic KRAS Mutations. JNCI J. Natl. Cancer Inst. 2009, 101, 1284–1285. [Google Scholar] [CrossRef] [PubMed]
  38. Murtaza, M.; Dawson, S.-J.; Tsui, D.W.Y.; Gale, D.; Forshew, T.; Piskorz, A.M.; Parkinson, C.; Chin, S.-F.; Kingsbury, Z.; Wong, A.S.C.; et al. Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA. Nature 2013, 497, 108–112. [Google Scholar] [CrossRef]
  39. Deveson, I.W.; Gong, B.; Lai, K.; LoCoco, J.S.; Richmond, T.A.; Schageman, J.; Zhang, Z.; Novoradovskaya, N.; Willey, J.C.; Jones, W.; et al. Evaluating the analytical validity of circulating tumor DNA sequencing assays for precision oncology. Nat. Biotechnol. 2021, 39, 1115–1128. [Google Scholar] [CrossRef]
  40. Zhang, Y.; Yao, Y.; Xu, Y.; Li, L.; Gong, Y.; Zhang, K.; Zhang, M.; Guan, Y.; Chang, L.; Xia, X.; et al. Pan-cancer circulating tumor DNA detection in over 10,000 Chinese patients. Nat. Commun. 2021, 12, 11. [Google Scholar] [CrossRef]
  41. Cohen, J.D.; Li, L.; Wang, Y.; Thoburn, C.; Afsari, B.; Danilova, L.; Douville, C.; Javed, A.A.; Wong, F.; Mattox, A.; et al. Detection and localization of surgically resectable cancers with a multi-analyte blood test. Science 2018, 359, 926–930. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Razavi, P.; Li, B.T.; Brown, D.N.; Jung, B.; Hubbell, E.; Shen, R.; Abida, W.; Juluru, K.; De Bruijn, I.; Hou, C.; et al. High-intensity sequencing reveals the sources of plasma circulating cell-free DNA variants. Nat. Med. 2019, 25, 1928–1937. [Google Scholar] [CrossRef] [PubMed]
  43. Hu, Y.; Ulrich, B.C.; Supplee, J.; Kuang, Y.; Lizotte, P.H.; Feeney, N.B.; Guibert, N.M.; Awad, M.M.; Wong, K.-K.; Jänne, P.A.; et al. False-Positive Plasma Genotyping Due to Clonal Hematopoiesis. Clin. Cancer Res. 2018, 24, 4437–4443. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Chan, H.T.; Nagayama, S.; Chin, Y.M.; Otaki, M.; Hayashi, R.; Kiyotani, K.; Fukunaga, Y.; Ueno, M.; Nakamura, Y.; Low, S. Clinical significance of clonal hematopoiesis in the interpretation of blood liquid biopsy. Mol. Oncol. 2020, 14, 1719–1730. [Google Scholar] [CrossRef]
  45. Li, M.; Diehl, F.; Dressman, D.; Vogelstein, B.; Kinzler, K.W. BEAMing up for detection and quantification of rare sequence variants. Nat. Methods 2006, 3, 95–97. [Google Scholar] [CrossRef]
  46. Bohers, E.; Viailly, P.-J.; Jardin, F. cfDNA Sequencing: Technological Approaches and Bioinformatic Issues. Pharmaceuticals 2021, 14, 596. [Google Scholar] [CrossRef]
  47. Gandara, D.R.; Paul, S.M.; Kowanetz, M.; Schleifman, E.; Zou, W.; Li, Y.; Rittmeyer, A.; Fehrenbacher, L.; Otto, G.; Malboeuf, C.; et al. Blood-based tumor mutational burden as a predictor of clinical benefit in non-small-cell lung cancer patients treated with atezolizumab. Nat. Med. 2018, 24, 1441–1448. [Google Scholar] [CrossRef]
  48. Khagi, Y.; Goodman, A.M.; Daniels, G.A.; Patel, S.P.; Sacco, A.G.; Randall, J.M.; Bazhenova, L.A.; Kurzrock, R. Hypermutated Circulating Tumor DNA: Correlation with Response to Checkpoint Inhibitor–Based Immunotherapy. Clin. Cancer Res. 2017, 23, 5729–5736. [Google Scholar] [CrossRef] [Green Version]
  49. Ma, X.; Shao, Y.; Tian, L.; Flasch, D.A.; Mulder, H.L.; Edmonson, M.N.; Liu, Y.; Chen, X.; Newman, S.; Nakitandwe, J.; et al. Analysis of error profiles in deep next-generation sequencing data. Genome Biol. 2019, 20, 50. [Google Scholar] [CrossRef] [Green Version]
  50. Kinde, I.; Wu, J.; Papadopoulos, N.; Kinzler, K.W.; Vogelstein, B. Detection and quantification of rare mutations with massively parallel sequencing. Proc. Natl. Acad. Sci. USA 2011, 108, 9530–9535. [Google Scholar] [CrossRef] [Green Version]
  51. Newman, A.M.; Lovejoy, A.F.; Klass, D.M.; Kurtz, D.M.; Chabon, J.J.; Scherer, F.; Stehr, H.; Liu, C.L.; Bratman, S.V.; Say, C.; et al. Integrated digital error suppression for improved detection of circulating tumor DNA. Nat. Biotechnol. 2016, 34, 547–555. [Google Scholar] [CrossRef] [PubMed]
  52. De Luca, G.; Dono, M. The Opportunities and Challenges of Molecular Tagging Next-Generation Sequencing in Liquid Biopsy. Mol. Diagn. Ther. 2021, 25, 537–547. [Google Scholar] [CrossRef] [PubMed]
  53. 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] [PubMed] [Green Version]
  54. Turner, N.C.; Kingston, B.; Kilburn, L.S.; Kernaghan, S.; Wardley, A.M.; Macpherson, I.R.; Baird, R.D.; Roylance, R.; Stephens, P.; Oikonomidou, O.; et al. Circulating tumour DNA analysis to direct therapy in advanced breast cancer (plasmaMATCH): A multicentre, multicohort, phase 2a, platform trial. Lancet Oncol. 2020, 21, 1296–1308. [Google Scholar] [CrossRef]
  55. Davis, A.A.; Jacob, S.; Gerratana, L.; Shah, A.N.; Wehbe, F.; Katam, N.; Zhang, Q.; Flaum, L.; Siziopikou, K.P.; Platanias, L.C.; et al. Landscape of circulating tumour DNA in metastatic breast cancer. eBioMedicine 2020, 58, 102914. [Google Scholar] [CrossRef]
  56. Li, B.; Janku, F.; Jung, B.; Hou, C.; Madwani, K.; Alden, R.; Razavi, P.; Reis-Filho, J.; Shen, R.; Isbell, J.; 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]
  57. Odegaard, J.I.; Vincent, J.J.; Mortimer, S.; Vowles, J.V.; Ulrich, B.C.; Banks, K.C.; Fairclough, S.R.; Zill, O.A.; Sikora, M.; Mokhtari, R.; et al. Validation of a Plasma-Based Comprehensive Cancer Genotyping Assay Utilizing Orthogonal Tissue- and Plasma-Based Methodologies. Clin. Cancer Res. 2018, 24, 3539–3549. [Google Scholar] [CrossRef] [Green Version]
  58. Tukachinsky, H.; Madison, R.W.; Chung, J.H.; Gjoerup, O.V.; Severson, E.A.; Dennis, L.; Fendler, B.J.; Morley, S.; Zhong, L.; Graf, R.P.; et al. Genomic Analysis of Circulating Tumor DNA in 3,334 Patients with Advanced Prostate Cancer Identifies Targetable BRCA Alterations and AR Resistance Mechanisms. Clin. Cancer Res. 2021, 27, 3094–3105. [Google Scholar] [CrossRef]
  59. Nakamura, Y.; Taniguchi, H.; Ikeda, M.; Bando, H.; Kato, K.; Morizane, C.; Esaki, T.; Komatsu, Y.; Kawamoto, Y.; Takahashi, N.; et al. Clinical utility of circulating tumor DNA sequencing in advanced gastrointestinal cancer: SCRUM-Japan GI-SCREEN and GOZILA studies. Nat. Med. 2020, 26, 1859–1864. [Google Scholar] [CrossRef]
  60. Flaherty, K.T.; Gray, R.J.; Chen, A.P.; Li, S.; McShane, L.M.; Patton, D.; Hamilton, S.R.; Williams, P.M.; Iafrate, A.J.; Sklar, J.; et al. Molecular Landscape and Actionable Alterations in a Genomically Guided Cancer Clinical Trial: National Cancer Institute Molecular Analysis for Therapy Choice (NCI-MATCH). J. Clin. Oncol. 2020, 38, 3883–3894. [Google Scholar] [CrossRef]
  61. Litchfield, K.; Turajlic, S.; Swanton, C. The GENIE Is Out of the Bottle: Landmark Cancer Genomics Dataset Released. Cancer Discov. 2017, 7, 796–798. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Le Tourneau, C.; Delord, J.-P.; Gonçalves, A.; Gavoille, C.; Dubot, C.; Isambert, N.; Campone, M.; Trédan, O.; Massiani, M.-A.; Mauborgne, C.; et al. Molecularly targeted therapy based on tumour molecular profiling versus conventional therapy for advanced cancer (SHIVA): A multicentre, open-label, proof-of-concept, randomised, controlled phase 2 trial. Lancet Oncol. 2015, 16, 1324–1334. [Google Scholar] [CrossRef]
  63. Brannon, A.R.; Jayakumaran, G.; Diosdado, M.; Patel, J.; Razumova, A.; Hu, Y.; Meng, F.; Haque, M.; Sadowska, J.; Murphy, B.J.; et al. Enhanced specificity of clinical high-sensitivity tumor mutation profiling in cell-free DNA via paired normal sequencing using MSK-ACCESS. Nat. Commun. 2021, 12, 3770. [Google Scholar] [CrossRef] [PubMed]
  64. Aggarwal, C.; Thompson, J.C.; Black, T.A.; Katz, S.I.; Fan, R.; Yee, S.S.; Chien, A.; Evans, T.L.; Bauml, J.M.; Alley, E.W.; et al. Clinical Implications of Plasma-Based Genotyping with the Delivery of Personalized Therapy in Metastatic Non–Small Cell Lung Cancer. JAMA Oncol. 2019, 5, 173–180. [Google Scholar] [CrossRef] [PubMed]
  65. Bieg-Bourne, C.C.; Okamura, R.; Kurzrock, R. Concordance between TP53 alterations in blood and tissue: Impact of time interval, biopsy site, cancer type and circulating tumor DNA burden. Mol. Oncol. 2020, 14, 1242–1251. [Google Scholar] [CrossRef] [Green Version]
  66. Tran, H.T.; Lam, V.K.; Elamin, Y.Y.; Hong, L.; Colen, R.; Elshafeey, N.A.; Hassan, I.S.A.; Altan, M.; Blumenschein, G.R.; Rinsurongkawong, W.; et al. Clinical Outcomes in Non–Small-Cell Lung Cancer Patients Treated With EGFR-Tyrosine Kinase Inhibitors and Other Targeted Therapies Based on Tumor Versus Plasma Genomic Profiling. JCO Precis. Oncol. 2021, 5, 1241–1249. [Google Scholar] [CrossRef]
  67. Park, S.; Olsen, S.; Ku, B.M.; Lee, M.; Jung, H.; Sun, J.; Lee, S.; Ahn, J.S.; Park, K.; Choi, Y.; et al. High concordance of actionable genomic alterations identified between circulating tumor DNA–based and tissue-based next-generation sequencing testing in advanced non–small cell lung cancer: The Korean Lung Liquid Versus Invasive Biopsy Program. Cancer 2021, 127, 3019–3028. [Google Scholar] [CrossRef]
  68. Madison, R.; Schrock, A.B.; Castellanos, E.; Gregg, J.P.; Snider, J.; Ali, S.M.; Miller, V.A.; Singal, G.; Alexander, B.M.; Venstrom, J.M.; et al. Retrospective analysis of real-world data to determine clinical outcomes of patients with advanced non-small cell lung cancer following cell-free circulating tumor DNA genomic profiling. Lung Cancer 2020, 148, 69–78. [Google Scholar] [CrossRef]
  69. Mack, P.C.; Ms, K.C.B.; Ms, C.R.E.; Ms, R.A.B.; Zill, O.A.; Ms, C.E.L.; Riess, J.W.; Mortimer, S.A.; Talasaz, A.; Lanman, R.B.; et al. Spectrum of driver mutations and clinical impact of circulating tumor DNA analysis in non–small cell lung cancer: Analysis of over 8000 cases. Cancer 2020, 126, 3219–3228. [Google Scholar] [CrossRef]
  70. Sabari, J.K.; Offin, M.; Stephens, D.; Ni, A.; Lee, A.; Pavlakis, N.; Clarke, S.; I Diakos, C.; Datta, S.; Tandon, N.; et al. A Prospective Study of Circulating Tumor DNA to Guide Matched Targeted Therapy in Lung Cancers. JNCI J. Natl. Cancer Inst. 2018, 111, 575–583. [Google Scholar] [CrossRef]
  71. Bujak, A.Z.; Weng, C.-F.; Silva, M.J.; Yeung, M.; Lo, L.; Ftouni, S.; Litchfield, C.; Ko, Y.-A.; Kuykhoven, K.; Van Geelen, C.; et al. Circulating tumour DNA in metastatic breast cancer to guide clinical trial enrolment and precision oncology: A cohort study. PLoS Med. 2020, 17, e1003363. [Google Scholar] [CrossRef]
  72. Zhang, M.; Qi, C.; Wang, Z.; Chen, H.; Zhao, X.; Zhang, X.; Zhou, Y.; Gao, C.; Bai, Y.; Jia, S.; et al. Molecular characterization of ctDNA from Chinese patients with advanced gastric adenocarcinoma reveals actionable alterations for targeted and immune therapy. Klin. Wochenschr. 2021, 99, 1311–1321. [Google Scholar] [CrossRef] [PubMed]
  73. Nakamura, Y.; Olsen, S.; Zhang, N.; Liao, J.; Yoshino, T. Comprehensive Genomic Profiling of Circulating Tumor DNA in Patients with Previously Treated Metastatic Colorectal Cancer: Analysis of a Real-World Healthcare Claims Database. Curr. Oncol. 2022, 29, 3433–3448. [Google Scholar] [CrossRef] [PubMed]
  74. Botrus, G.; Kosirorek, H.; Sonbol, M.B.; Kusne, Y.; Uson, P.L.S.; Borad, M.J.; Ahn, D.H.; Kasi, P.M.; Drusbosky, L.M.; Dada, H.; et al. Circulating Tumor DNA-Based Testing and Actionable Findings in Patients with Advanced and Metastatic Pancreatic Adenocarcinoma. Oncologist 2021, 26, 569–578. [Google Scholar] [CrossRef]
  75. Fribbens, C.; O’Leary, B.; Kilburn, L.; Hrebien, S.; Garcia-Murillas, I.; Beaney, M.; Cristofanilli, M.; Andre, F.; Loi, S.; Loibl, S.; et al. Plasma ESR1 Mutations and the Treatment of Estrogen Receptor–Positive Advanced Breast Cancer. J. Clin. Oncol. 2016, 34, 2961–2968. [Google Scholar] [CrossRef]
  76. Andre, F.; Ciruelos, E.M.; Juric, D.; Loibl, S.; Campone, M.; Mayer, I.A.; Rubovszky, G.; Yamashita, T.; Kaufman, B.; Lu, Y.-S.; et al. Alpelisib plus fulvestrant for PIK3CA-mutated, hormone receptor-positive, human epidermal growth factor receptor-2-negative advanced breast cancer: Final overall survival results from SOLAR-1. Ann. Oncol. 2021, 32, 208–217. [Google Scholar] [CrossRef]
  77. Matsubara, N.; de Bono, J.S.; Olmos, D.; Procopio, G.; Kawakami, S.; Urun, Y.; van Alphen, R.J.; Flechon, A.; Carducci, M.A.; Choi, Y.D.; et al. Olaparib efficacy in patients with metastatic castration-resistant prostate cancer (mCRPC) carrying circulating tumor (ct) DNA alterations in BRCA1, BRCA2 or ATM: Results from the PROfound study. J. Clin. Oncol. 2021, 39, 27. [Google Scholar] [CrossRef]
  78. Sharma, P.; Abramson, V.G.; O’Dea, A.P.; Nye, L.; Mayer, I.A.; Pathak, H.B.; Hoffmann, M.; Stecklein, S.R.; Elia, M.; Lewis, S.; et al. Clinical and Biomarker Results from Phase I/II Study of PI3K Inhibitor Alpelisib plus Nab-paclitaxel in HER2-Negative Metastatic Breast Cancer. Clin. Cancer Res. 2021, 27, 3896–3904. [Google Scholar] [CrossRef]
  79. Rothé, F.; Silva, M.J.; Venet, D.; Campbell, C.; Bradburry, I.; Rouas, G.; de Azambuja, E.; Maetens, M.; Fumagalli, D.; Rodrik-Outmezguine, V.; et al. Circulating Tumor DNA in HER2-Amplified Breast Cancer: A Translational Research Substudy of the NeoALTTO Phase III Trial. Clin. Cancer Res. 2019, 25, 3581–3588. [Google Scholar] [CrossRef] [Green Version]
  80. Kato, S.; Weipert, C.; Gumas, S.; Okamura, R.; Lee, S.; Sicklick, J.K.; Saam, J.; Kurzrock, R. Therapeutic Actionability of Circulating Cell-Free DNA Alterations in Carcinoma of Unknown Primary. JCO Precis. Oncol. 2021, 5, 1687–1698. [Google Scholar] [CrossRef]
  81. Kolling, S.; Ventre, F.; Geuna, E.; Milan, M.; Pisacane, A.; Boccaccio, C.; Sapino, A.; Montemurro, F. “Metastatic Cancer of Unknown Primary” or “Primary Metastatic Cancer”? Front. Oncol. 2019, 9, 1546. [Google Scholar] [CrossRef] [PubMed]
  82. Dziadziuszko, R.; Mok, T.; Peters, S.; Han, J.-Y.; Alatorre-Alexander, J.; Leighl, N.; Sriuranpong, V.; Pérol, M.; Junior, G.D.C.; Nadal, E.; et al. Blood First Assay Screening Trial (BFAST) in Treatment-Naive Advanced or Metastatic NSCLC: Initial Results of the Phase 2 ALK-Positive Cohort. J. Thorac. Oncol. 2021, 16, 2040–2050. [Google Scholar] [CrossRef] [PubMed]
  83. Nakamura, Y.; Okamoto, W.; Kato, T.; Esaki, T.; Kato, K.; Komatsu, Y.; Yuki, S.; Masuishi, T.; Nishina, T.; Ebi, H.; et al. Circulating tumor DNA-guided treatment with pertuzumab plus trastuzumab for HER2-amplified metastatic colorectal cancer: A phase 2 trial. Nat. Med. 2021, 27, 1899–1903. [Google Scholar] [CrossRef] [PubMed]
  84. Lee, Y.; Clark, E.W.; Milan, M.S.; Champagne, C.; Michael, K.S.; Awad, M.M.; Barbie, D.A.; Cheng, M.L.; Kehl, K.L.; Marcoux, J.P.; et al. Turnaround Time of Plasma Next-Generation Sequencing in Thoracic Oncology Patients: A Quality Improvement Analysis. JCO Precis. Oncol. 2020, 4, 1098–1108. [Google Scholar] [CrossRef] [PubMed]
  85. Schwaederle, M.; Chattopadhyay, R.; Kato, S.; Fanta, P.T.; Banks, K.C.; Choi, I.S.; Piccioni, D.E.; Ikeda, S.; Talasaz, A.; Lanman, R.B.; et al. Genomic Alterations in Circulating Tumor DNA from Diverse Cancer Patients Identified by Next-Generation Sequencing. Cancer Res. 2017, 77, 5419–5427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Choudhury, Y.; Tan, M.-H.; Shi, J.L.; Tee, A.; Ngeow, K.C.; Poh, J.; Goh, R.R.; Mong, J. Complementing Tissue Testing with Plasma Mutation Profiling Improves Therapeutic Decision-Making for Patients with Lung Cancer. Front. Med. 2022, 9, 758464. [Google Scholar] [CrossRef]
  87. FoundationMedicine. FoundationOne Liquid CDx. 2022. Available online: https://www.foundationmedicine.com/test/foundationone-liquid-cdx#:~:text=FoundationOne%20Liquid%20CDx%20is%20an,liquid%20biopsy%20on%20the%20market (accessed on 16 February 2022).
  88. GuardantHealth. Guardant360 CDx. 2022. Available online: https://guardant360cdx.com/ (accessed on 16 February 2022).
  89. Low, S.-K.K.; Uchibori, K.; Hayashi, R.; Chan, H.T.; Ariyasu, R.; Kitazono, S.; Yanagitani, N.; Nishio, M.; Nakamura, Y. Evaluation of Genexus system that automates specimen-to-report for cancer genomic profiling within a day using liquid biopsy. J. Clin. Oncol. 2020, 38, 3538. [Google Scholar] [CrossRef]
  90. Low, S.-K.; Ariyasu, R.; Uchibori, K.; Hayashi, R.; Chan, H.T.; Chin, Y.M.; Akita, T.; Harutani, Y.; Kiritani, A.; Tsugitomi, R.; et al. Rapid genomic profiling of circulating tumor DNA in non-small cell lung cancer using Oncomine Precision Assay with GenexusTM integrated sequencer. Transl. Lung Cancer Res. 2021, 11, 711–721. [Google Scholar] [CrossRef]
  91. Al Zoughbi, W.; Fox, J.; Beg, S.; Papp, E.; Hissong, E.; Ohara, K.; Keefer, L.; Sigouros, M.; Kane, T.; Bockelman, D.; et al. Validation of a Circulating Tumor DNA-Based Next-Generation Sequencing Assay in a Cohort of Patients with Solid tumors: A Proposed Solution for Decentralized Plasma Testing. Oncologist 2021, 26, e1971–e1981. [Google Scholar] [CrossRef]
  92. FDA. Cobas EGFR Mutation Test v2. 2016. Available online: https://www.fda.gov/drugs/resources-information-approved-drugs/cobas-egfr-mutation-test-v2 (accessed on 17 February 2022).
  93. Roche. Roche Receives FDA Approval for the cobas EGFR Mutation Test v2 as the First Companion Diagnostic Test for Expanded EGFR TKI Therapies in Patients with Non-Small Cell Lung Cancer. 2020. Available online: https://diagnostics.roche.com/global/en/news-listing/2020/fda-class-claim-for-cobas-egfr-mutation-test-v2.html#:~:text=Roche%20receives%20FDA%20approval%20for,non%2Dsmall%20cell%20lung%20cancer (accessed on 17 February 2022).
  94. Heeke, S.; Benzaquen, J.; Hofman, V.; Ilié, M.; Allegra, M.; Long-Mira, E.; Lassalle, S.; Tanga, V.; Salacroup, C.; Bonnetaud, C.; et al. Critical Assessment in Routine Clinical Practice of Liquid Biopsy for EGFR Status Testing in Non–Small-Cell Lung Cancer: A Single-Laboratory Experience (LPCE, Nice, France). Clin. Lung Cancer 2020, 21, 56–65. [Google Scholar] [CrossRef] [Green Version]
  95. Kim, Y.; Shin, S.; Lee, K.-A. A Comparative Study for Detection of EGFR Mutations in Plasma Cell-Free DNA in Korean Clinical Diagnostic Laboratories. BioMed Res. Int. 2018, 2018, 7392419. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Mountzios, G.; Koumarianou, A.; Bokas, A.; Mavroudis, D.; Samantas, E.; Fergadis, E.; Linardou, H.; Katsaounis, P.; Athanasiadis, E.; Karamouzis, M.; et al. A Real-World, Observational, Prospective Study to Assess the Molecular Epidemiology of Epidermal Growth Factor Receptor (EGFR) Mutations upon Progression on or after First-Line Therapy with a First- or Second-Generation EGFR Tyrosine Kinase Inhibitor in EGFR Mutation-Positive Locally Advanced or Metastatic Non-Small Cell Lung Cancer: The ‘LUNGFUL’ Study. Cancers 2021, 13, 3172. [Google Scholar] [CrossRef] [PubMed]
  97. FDA. FoundationOne Liquid CDx—P190032. 2020. Available online: https://www.fda.gov/medical-devices/recently-approved-devices/foundationone-liquid-cdx-p190032 (accessed on 17 February 2022).
  98. FDA. Guardant360 CDx—P200010. 2020. Available online: https://www.fda.gov/medical-devices/recently-approved-devices/guardant360-cdx-p200010 (accessed on 17 February 2022).
  99. Guardant Health, Guardant360 CDx Technical Information. 2021. Available online: https://guardant360cdx.com/wp-content/uploads/guardant360-cdx-technical-information.pdf (accessed on 31 May 2022).
  100. Foundation Medicine, Foundation One Liquid CDx Technical Information. 2021. Available online: https://info.foundationmedicine.com/hubfs/FMI%20Labels/FoundationOne_Liquid_CDx_Label_Technical_Info.pdf (accessed on 31 May 2022).
  101. Larribère, L.; Martens, U.M. Advantages and Challenges of Using ctDNA NGS to Assess the Presence of Minimal Residual Disease (MRD) in Solid Tumors. Cancers 2021, 13, 5698. [Google Scholar] [CrossRef] [PubMed]
  102. Dawson, S.-J. Characterizing the Cancer Genome in Blood. Cold Spring Harb. Perspect. Med. 2018, 9, a026880. [Google Scholar] [CrossRef] [Green Version]
  103. Lin, C.; Liu, X.; Zheng, B.; Ke, R.; Tzeng, C.-M. Liquid Biopsy, ctDNA Diagnosis through NGS. Life 2021, 11, 890. [Google Scholar] [CrossRef]
  104. Ross, M.G.; Russ, C.; Costello, M.; Hollinger, A.; Lennon, N.J.; Hegarty, R.; Nusbaum, C.; Jaffe, D.B. Characterizing and measuring bias in sequence data. Genome Biol. 2013, 14, R51-20. [Google Scholar] [CrossRef] [Green Version]
  105. Willey, J.C.; Morrison, T.B.; Austermiller, B.; Crawford, E.L.; Craig, D.J.; Blomquist, T.M.; Jones, W.D.; Wali, A.; Lococo, J.S.; Haseley, N.; et al. Advancing NGS quality control to enable measurement of actionable mutations in circulating tumor DNA. Cell Rep. Methods 2021, 1, 100106. [Google Scholar] [CrossRef]
  106. Stetson, D.; Ahmed, A.; Xu, X.; Nuttall, B.; Lubinski, T.J.; Johnson, J.H.; Barrett, J.C.; Dougherty, B.A. Orthogonal Comparison of Four Plasma NGS Tests with Tumor Suggests Technical Factors are a Major Source of Assay Discordance. JCO Precis. Oncol. 2019, 3, 1–9. [Google Scholar] [CrossRef]
  107. El Achi, H.; Khoury, J.D.; Loghavi, S. Liquid Biopsy by Next-Generation Sequencing: A Multimodality Test for Management of Cancer. Curr. Hematol. Malign. Rep. 2019, 14, 358–367. [Google Scholar] [CrossRef]
  108. Shu, Y.; Wu, X.; Tong, X.; Wang, X.; Chang, Z.; Mao, Y.; Chen, X.; Sun, J.; Wang, Z.; Hong, Z.; et al. Circulating Tumor DNA Mutation Profiling by Targeted Next Generation Sequencing Provides Guidance for Personalized Treatments in Multiple Cancer Types. Sci. Rep. 2017, 7, 583. [Google Scholar] [CrossRef]
  109. Vaclova, T.; Grazini, U.; Ward, L.; O’Neill, D.; Markovets, A.; Huang, X.; Chmielecki, J.; Hartmaier, R.; Thress, K.S.; Smith, P.D.; et al. Clinical impact of subclonal EGFR T790M mutations in advanced-stage EGFR-mutant non-small-cell lung cancers. Nat. Commun. 2021, 12, 1780. [Google Scholar] [CrossRef] [PubMed]
  110. Loeb, L.A.; Kohrn, B.F.; Loubet-Senear, K.J.; Dunn, Y.J.; Ahn, E.H.; O’Sullivan, J.N.; Salk, J.J.; Bronner, M.P.; Beckman, R.A. Extensive subclonal mutational diversity in human colorectal cancer and its significance. Proc. Natl. Acad. Sci. USA 2019, 116, 26863–26872. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Schmitt, M.W.; Loeb, L.A.; Salk, J.J. The influence of subclonal resistance mutations on targeted cancer therapy. Nat. Rev. Clin. Oncol. 2015, 13, 335–347. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Laurent-Puig, P.; Pekin, D.; Normand, C.; Kotsopoulos, S.K.; Nizard, P.; Perez-Toralla, K.; Rowell, R.; Olson, J.; Srinivasan, P.; Le Corre, D.; et al. Clinical Relevance of KRAS-Mutated Subclones Detected with Picodroplet Digital PCR in Advanced Colorectal Cancer Treated with Anti-EGFR Therapy. Clin. Cancer Res. 2015, 21, 1087–1097. [Google Scholar] [CrossRef] [Green Version]
  113. Mouliere, F.; Chandrananda, D.; Piskorz, A.M.; Moore, E.K.; Morris, J.; Ahlborn, L.B.; Mair, R.; Goranova, T.; Francesco Marass, K.H. Enhanced detection of circulating tumor DNA by fragment size analysis. Sci. Transl. Med. 2018, 10, eaat4921. [Google Scholar] [CrossRef] [Green Version]
  114. Ishida, Y.; Takano, S.; Maekawa, S.; Yamaguchi, T.; Yoshida, T.; Kobayashi, S.; Iwamoto, F.; Kuno, T.; Hayakawa, H.; Matsuda, S.; et al. Fractionated small cell-free DNA increases possibility to detect cancer-related gene mutations in advanced colorectal cancer. JGH Open 2020, 4, 978–986. [Google Scholar] [CrossRef]
  115. Wan, J.C.M.; Heider, K.; Gale, D.; Murphy, S.; Fisher, E.; Mouliere, F.; Ruiz-Valdepenas, A.; Santonja, A.; Morris, J.; Chandrananda, D.; et al. ctDNA monitoring using patient-specific sequencing and integration of variant reads. Sci. Transl. Med. 2020, 12, eaaz8084. [Google Scholar] [CrossRef]
  116. Esagian, S.M.; Grigoriadou, G.I.; Nikas, I.P.; Boikou, V.; Sadow, P.M.; Won, J.-K.; Economopoulos, K.P. Comparison of liquid-based to tissue-based biopsy analysis by targeted next generation sequencing in advanced non-small cell lung cancer: A comprehensive systematic review. J. Cancer Res. Clin. Oncol. 2020, 146, 2051–2066. [Google Scholar] [CrossRef]
  117. Bruno, R.; Fontanini, G. Next Generation Sequencing for Gene Fusion Analysis in Lung Cancer: A Literature Review. Diagnostics 2020, 10, 521. [Google Scholar] [CrossRef]
  118. Schram, A.M.; Chang, M.T.; Jonsson, P.; Drilon, A. Fusions in solid tumours: Diagnostic strategies, targeted therapy, and acquired resistance. Nat. Rev. Clin. Oncol. 2017, 14, 735–748. [Google Scholar] [CrossRef]
  119. Lee, J.K.; Hazar-Rethinam, M.; Decker, B.; Gjoerup, O.; Madison, R.W.; Lieber, D.S.; Chung, J.H.; Schrock, A.B.; Creeden, J.; Venstrom, J.M.; et al. The Pan-Tumor Landscape of Targetable Kinase Fusions in Circulating Tumor DNA. Clin. Cancer Res. 2021, 28, 728–737. [Google Scholar] [CrossRef] [PubMed]
  120. Plagnol, V.; Woodhouse, S.; Howarth, K.; Lensing, S.; Smith, M.; Epstein, M.; Madi, M.; Smalley, S.; Leroy, C.; Hinton, J.; et al. Analytical validation of a next generation sequencing liquid biopsy assay for high sensitivity broad molecular profiling. PLoS ONE 2018, 13, e0193802. [Google Scholar] [CrossRef] [PubMed]
  121. Supplee, J.G.; Milan, M.S.; Lim, L.P.; Potts, K.T.; Sholl, L.M.; Oxnard, G.R.; Paweletz, C.P. Sensitivity of next-generation sequencing assays detecting oncogenic fusions in plasma cell-free DNA. Lung Cancer 2019, 134, 96–99. [Google Scholar] [CrossRef] [Green Version]
  122. Hasegawa, N.; Kohsaka, S.; Kurokawa, K.; Shinno, Y.; Nakamura, I.T.; Ueno, T.; Kojima, S.; Kawazu, M.; Suehara, Y.; Ishijima, M.; et al. Highly sensitive fusion detection using plasma cell-free RNA in non-small-cell lung cancers. Cancer Sci. 2021, 112, 4393–4403. [Google Scholar] [CrossRef] [PubMed]
  123. Papadopoulou, E.; Tsoulos, N.; Tsantikidi, K.; Metaxa-Mariatou, V.; Stamou, P.E.; Kladi-Skandali, A.; Kapeni, E.; Tsaousis, G.; Pentheroudakis, G.; Petrakis, D.; et al. Clinical feasibility of NGS liquid biopsy analysis in NSCLC patients. PLoS ONE 2019, 14, e0226853. [Google Scholar] [CrossRef] [Green Version]
  124. Jahangiri, L.; Hurst, T. Assessing the Concordance of Genomic Alterations between Circulating-Free DNA and Tumour Tissue in Cancer Patients. Cancers 2019, 11, 1938. [Google Scholar] [CrossRef] [Green Version]
  125. 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, 224. [Google Scholar] [CrossRef] [Green Version]
  126. Underhill, H.R. Leveraging the Fragment Length of Circulating Tumour DNA to Improve Molecular Profiling of Solid Tumour Malignancies with Next-Generation Sequencing: A Pathway to Advanced Non-invasive Diagnostics in Precision Oncology? Mol. Diagn. Ther. 2021, 25, 389–408. [Google Scholar] [CrossRef]
  127. Weber, Z.T.; Collier, K.A.; Tallman, D.; Forman, J.; Shukla, S.; Asad, S.; Rhoades, J.; Freeman, S.; Parsons, H.A.; Williams, N.O.; et al. Modeling clonal structure over narrow time frames via circulating tumor DNA in metastatic breast cancer. Genome Med. 2021, 13, 89. [Google Scholar] [CrossRef]
  128. Lee, S.; Park, Y.-S.; Chang, W.-J.; Choi, J.Y.; Lim, A.; Kim, B.; Lee, S.-B.; Lee, J.-W.; Kim, S.-H.; Kim, J.; et al. Clinical Implication of Liquid Biopsy in Colorectal Cancer Patients Treated with Metastasectomy. Cancers 2021, 13, 2231. [Google Scholar] [CrossRef]
  129. Bando, H.; Kagawa, Y.; Kato, T.; Akagi, K.; Denda, T.; Nishina, T.; Komatsu, Y.; Oki, E.; Kudo, T.; Kumamoto, H.; et al. Correction: A multicentre, prospective study of plasma circulating tumour DNA test for detecting RAS mutation in patients with metastatic colorectal cancer. Br. J. Cancer 2020, 122, 1271. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Kagawa, Y.; Elez, E.; García-Foncillas, J.; Bando, H.; Taniguchi, H.; Vivancos, A.; Akagi, K.; García, A.; Denda, T.; Ros, J.; et al. Combined Analysis of Concordance between Liquid and Tumor Tissue Biopsies for RAS Mutations in Colorectal Cancer with a Single Metastasis Site: The METABEAM Study. Clin. Cancer Res. 2021, 27, 2515–2522. [Google Scholar] [CrossRef] [PubMed]
  131. Tsui, D.W.Y.; Cheng, M.L.; Shady, M.; Yang, J.L.; Stephens, D.; Won, H.; Srinivasan, P.; Huberman, K.; Meng, F.; Jing, X.; et al. Tumor fraction-guided cell-free DNA profiling in metastatic solid tumor patients. Genome Med. 2021, 13, 96. [Google Scholar] [CrossRef]
  132. 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. JNCI J. Natl. Cancer Inst. 2017, 109, djx118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Mayrhofer, M.; De Laere, B.; Whitington, T.; Van Oyen, P.; Ghysel, C.; Ampe, J.; Ost, P.; Demey, W.; Hoekx, L.; Schrijvers, D.; et al. Cell-free DNA profiling of metastatic prostate cancer reveals microsatellite instability, structural rearrangements and clonal hematopoiesis. Genome Med. 2018, 10, 85. [Google Scholar] [CrossRef]
  134. Chabon, J.J.; Hamilton, E.G.; Kurtz, D.M.; Esfahani, M.S.; Moding, E.J.; Stehr, H.; Schroers-Martin, J.; Nabet, B.Y.; Chen, B.; Chaudhuri, A.A.; et al. Integrating genomic features for non-invasive early lung cancer detection. Nature 2020, 580, 245–251. [Google Scholar] [CrossRef]
  135. Leal, A.; Van Grieken, N.C.T.; Palsgrove, D.N.; Phallen, J.; Medina, J.E.; Hruban, C.; Broeckaert, M.A.M.; Anagnostou, V.; Adleff, V.; Bruhm, D.C.; et al. White blood cell and cell-free DNA analyses for detection of residual disease in gastric cancer. Nat. Commun. 2020, 11, 525. [Google Scholar] [CrossRef] [Green Version]
  136. Bacon, J.V.; Annala, M.; Soleimani, M.; Lavoie, J.-M.; So, A.; Gleave, M.E.; Fazli, L.; Wang, G.; Chi, K.N.; Kollmannsberger, C.K.; et al. Plasma Circulating Tumor DNA and Clonal Hematopoiesis in Metastatic Renal Cell Carcinoma. Clin. Genitourin. Cancer 2020, 18, 322–331. [Google Scholar] [CrossRef]
  137. Cardoso, F.; Paluch-Shimon, S.; Senkus, E.; Curigliano, G.; Aapro, M.S.; André, F.; Barrios, C.H.; Bergh, J.; Bhattacharyya, G.S.; Biganzoli, L.; et al. 5th ESO-ESMO international consensus guidelines for advanced breast cancer (ABC 5). Ann. Oncol. 2020, 31, 1623–1649. [Google Scholar] [CrossRef]
  138. Burstein, H.J.; Somerfield, M.R.; Barton, D.L.; Dorris, A.; Fallowfield, L.J.; Jain, D.; Johnston, S.R.D.; Korde, L.A.; Litton, J.K.; Macrae, E.R.; et al. Endocrine Treatment and Targeted Therapy for Hormone Receptor-Positive, Human Epidermal Growth Factor Receptor 2-Negative Metastatic Breast Cancer: ASCO Guideline Update. J. Clin. Oncol. 2021, 39, 3959–3977. [Google Scholar] [CrossRef]
  139. National Comprehensive Cancer Network. NCCN Clinical Practice Guidelines in Oncology—Breast Cancer. Version 5.2020; National Comprehensive Cancer Network: Plymouth Meeting, PA, USA, 2020. [Google Scholar]
  140. Rolfo, C.; Mack, P.; Scagliotti, G.V.; Aggarwal, C.; Arcila, M.E.; Barlesi, F.; Bivona, T.; Diehn, M.; Dive, C.; Dziadziuszko, R.; et al. Liquid Biopsy for Advanced NSCLC: A Consensus Statement from the International Association for the Study of Lung Cancer. J. Thorac. Oncol. 2021, 16, 1647–1662. [Google Scholar] [CrossRef] [PubMed]
  141. Planchard, D.; Popat, S.; Kerr, K.; Novello, S.; Smit, E.F.; Faivre-Finn, C.; Mok, T.S.; Reck, M.; Van Schil, P.E.; Hellmann, M.D.; et al. Metastatic non-small cell lung cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2018, 29 (Suppl. 4), iv192–iv237. [Google Scholar] [CrossRef] [PubMed]
  142. Kalemkerian, G.P.; Narula, N.; Kennedy, E.B. Molecular Testing Guideline for the Selection of Lung Cancer Patients for Treatment with Targeted Tyrosine Kinase Inhibitors: American Society of Clinical Oncology Endorsement Summary of the College of American Pathologists/International Association for the Study of Lung Cancer/Association for Molecular Pathology Clinical Practice Guideline Update. J. Oncol. Pract. 2018, 14, 323–327. [Google Scholar] [CrossRef] [PubMed]
  143. Ettinger, D.S.; Wood, D.E.; Aisner, D.L.; Akerley, W.; Bauman, J.R.; Bharat, A.; Bruno, D.S.; Chang, J.Y.; Chirieac, L.R.; D’Amico, T.A.; et al. Non–Small Cell Lung Cancer, Version 3.2022, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Cancer Netw. 2022, 20, 497–530. [Google Scholar] [CrossRef] [PubMed]
  144. Sunami, K.; Bando, H.; Yatabe, Y.; Naito, Y.; Takahashi, H.; Tsuchihara, K.; Toyooka, S.; Mimori, K.; Kohsaka, S.; Uetake, H.; et al. Appropriate use of cancer comprehensive genome profiling assay using circulating tumor DNA. Cancer Sci. 2021, 112, 3911–3917. [Google Scholar] [CrossRef] [PubMed]
  145. Pereira, B.; Chen, C.T.; Goyal, L.; Walmsley, C.; Pinto, C.J.; Baiev, I.; Allen, R.; Henderson, L.; Saha, S.; Reyes, S.; et al. Cell-free DNA captures tumor heterogeneity and driver alterations in rapid autopsies with pre-treated metastatic cancer. Nat. Commun. 2021, 12, 3199. [Google Scholar] [CrossRef]
  146. Moreno, F.; Gayarre, J.; López-Tarruella, S.; del Monte-Millán, M.; Picornell, A.C.; Álvarez, E.; García-Saenz, J.; Jerez, Y.; Márquez-Rodas, I.; Echavarría, I.; et al. Concordance of Genomic Variants in Matched Primary Breast Cancer, Metastatic Tumor, and Circulating Tumor DNA: The MIRROR Study. JCO Precis. Oncol. 2019, 3, 1–16. [Google Scholar] [CrossRef]
  147. Liebs, S.; Eder, T.; Klauschen, F.; Schütte, M.; Yaspo, M.-L.; Keilholz, U.; Tinhofer, I.; Kidess-Sigal, E.; Braunholz, D. Applicability of liquid biopsies to represent the mutational profile of tumor tissue from different cancer entities. Oncogene 2021, 40, 5204–5212. [Google Scholar] [CrossRef]
  148. Barefoot, M.E.; Loyfer, N.; Kiliti, A.J.; McDeed, A.P.I.; Kaplan, T.; Wellstein, A. Detection of Cell Types Contributing to Cancer from Circulating, Cell-Free Methylated DNA. Front. Genet. 2021, 12, 671057. [Google Scholar] [CrossRef]
  149. Lo, Y.M.D.; Han, D.S.C.; Jiang, P.; Chiu, R.W.K. Epigenetics, fragmentomics, and topology of cell-free DNA in liquid biopsies. Science 2021, 372, eaaw3616. [Google Scholar] [CrossRef]
  150. Parikh, A.R.; Leshchiner, I.; Elagina, L.; Goyal, L.; Levovitz, C.; Siravegna, G.; Livitz, D.; Rhrissorrakrai, K.; Martin, E.E.; Van Seventer, E.E.; et al. Liquid versus tissue biopsy for detecting acquired resistance and tumor heterogeneity in gastrointestinal cancers. Nat. Med. 2019, 25, 1415–1421. [Google Scholar] [CrossRef] [PubMed]
  151. Hua, G.; Zhang, X.; Zhang, M.; Wang, Q.; Chen, X.; Yu, R.; Bao, H.; Liu, J.; Wu, X.; Shao, Y.; et al. Real-world circulating tumor DNA analysis depicts resistance mechanism and clonal evolution in ALK inhibitor-treated lung adenocarcinoma patients. ESMO Open 2022, 7, 100337. [Google Scholar] [CrossRef] [PubMed]
  152. Hahn, A.W.; Stenehjem, D.; Nussenzveig, R.; Carroll, E.; Bailey, E.; Batten, J.; Maughan, B.L.; Agarwal, N. Evolution of the genomic landscape of circulating tumor DNA (ctDNA) in metastatic prostate cancer over treatment and time. Cancer Treat. Res. Commun. 2019, 19, 100120. [Google Scholar] [CrossRef] [PubMed]
  153. Gupta, R.; Othman, T.; Chen, C.; Sandhu, J.; Ouyang, C.; Fakih, M. Guardant360 Circulating Tumor DNA Assay Is Concordant with FoundationOne Next-Generation Sequencing in Detecting Actionable Driver Mutations in Anti-EGFR Naive Metastatic Colorectal Cancer. Oncol. 2019, 25, 235–243. [Google Scholar] [CrossRef] [Green Version]
  154. Abbosh, C.; Swanton, C.; Birkbak, N. Clonal haematopoiesis: A source of biological noise in cell-free DNA analyses. Ann. Oncol. 2019, 30, 358–359. [Google Scholar] [CrossRef]
  155. Marass, F.; Stephens, D.; Ptashkin, R.; Zehir, A.; Berger, M.F.; Solit, D.B.; A Diaz, L.; Tsui, D.W.Y. Fragment Size Analysis May Distinguish Clonal Hematopoiesis from Tumor-Derived Mutations in Cell-Free DNA. Clin. Chem. 2020, 66, 616–618. [Google Scholar] [CrossRef] [Green Version]
  156. Cristiano, S.; Leal, A.; Phallen, J.; Fiksel, J.; Adleff, V.; Bruhm, D.C.; Jensen, S.Ø.; Medina, J.E.; Hruban, C.; White, J.R.; et al. Genome-wide cell-free DNA fragmentation in patients with cancer. Nature 2019, 570, 385–389. [Google Scholar] [CrossRef]
  157. Ai, X.; Cui, J.; Zhang, J.; Chen, R.; Lin, W.; Xie, C.; Liu, A.; Zhang, J.; Yang, W.; Hu, X.; et al. Clonal Architecture of EGFR Mutation Predicts the Efficacy of EGFR-Tyrosine Kinase Inhibitors in Advanced NSCLC: A Prospective Multicenter Study (NCT03059641). Clin. Cancer Res. 2021, 27, 704–712. [Google Scholar] [CrossRef]
  158. Martinelli, E.; Martini, G.; Famiglietti, V.; Troiani, T.; Napolitano, S.; Pietrantonio, F.; Ciardiello, D.; Terminiello, M.; Borrelli, C.; Vitiello, P.P.; et al. Cetuximab Rechallenge Plus Avelumab in Pretreated Patients with RAS Wild-type Metastatic Colorectal Cancer: The Phase 2 Single-Arm Clinical CAVE Trial. JAMA Oncol. 2021, 7, 1529–1535. [Google Scholar] [CrossRef]
  159. Sartore-Bianchi, A.; Pietrantonio, F.; Lonardi, S.; Mussolin, B.; Rua, F.; Fenocchio, E.; Amatu, A.; Corallo, S.; Manai, C.; Tosi, F.; et al. Phase II study of anti-EGFR rechallenge therapy with panitumumab driven by circulating tumor DNA molecular selection in metastatic colorectal cancer: The CHRONOS trial. J. Clin. Oncol. 2021, 39, 3506. [Google Scholar] [CrossRef]
  160. Parseghian, C.; Loree, J.; Morris, V.; Liu, X.; Clifton, K.; Napolitano, S.; Henry, J.; Pereira, A.; Vilar, E.; Johnson, B.; et al. Anti-EGFR-resistant clones decay exponentially after progression: Implications for anti-EGFR re-challenge. Ann. Oncol. 2019, 30, 243–249. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  161. Siena, S.; Sartore-Bianchi, A.; Garcia-Carbonero, R.; Karthaus, M.; Smith, D.; Tabernero, J.; Van Cutsem, E.; Guan, X.; Boedigheimer, M.; Ang, A.; et al. Dynamic molecular analysis and clinical correlates of tumor evolution within a phase II trial of panitumumab-based therapy in metastatic colorectal cancer. Ann. Oncol. 2017, 29, 119–126. [Google Scholar] [CrossRef] [PubMed]
  162. Chin, Y.M.; Shibayama, T.; Chan, H.T.; Otaki, M.; Hara, F.; Kobayashi, T.; Kobayashi, K.; Hosonaga, M.; Fukada, I.; Inagaki, L.; et al. Serial circulating tumor DNA monitoring of CDK4/6 inhibitors response in metastatic breast cancer. Cancer Sci. 2022, 113, 1808–1820. [Google Scholar] [CrossRef] [PubMed]
  163. Bidard, F.-C.; Hardy-Bessard, A.-C.; Bachelot, T.; Pierga, J.-Y.; Canon, J.-L.; Clatot, F.; Andre, F.; De La Motte Rouge, T.; Pistilli, B.; Dalenc, F.; et al. Fulvestrant-palbociclib vs. continuing AI-palbociclib upon detection of circulating ESR1 mutation in HR+ HER2–metastatic breast cancer patients: Results of PADA-1, a UCBG-GINECO randomized phase 3 trial. In Proceedings of the 2021 San Antonio Breast Cancer Symposium, San Antonio, TX, USA, 7–10 December 2021. [Google Scholar]
  164. 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] [PubMed]
  165. Palmeri, M.; Mehnert, J.; Silk, A.; Jabbour, S.; Ganesan, S.; Popli, P.; Riedlinger, G.; Stephenson, R.; de Meritens, A.; Leiser, A.; et al. Real-world application of tumor mutational burden-high (TMB-high) and microsatellite instability (MSI) confirms their utility as immunotherapy biomarkers. ESMO Open 2021, 7, 100336. [Google Scholar] [CrossRef]
  166. Snyder, A.; Morrissey, M.P.; Hellmann, M.D. Use of Circulating Tumor DNA for Cancer Immunotherapy. Clin. Cancer Res. 2019, 25, 6909–6915. [Google Scholar] [CrossRef] [Green Version]
  167. Kim, S.T.; Cristescu, R.; Bass, A.J.; Kim, K.-M.; Odegaard, J.I.; Kim, K.; Liu, X.Q.; Sher, X.; Jung, H.; Lee, M.; et al. Comprehensive molecular characterization of clinical responses to PD-1 inhibition in metastatic gastric cancer. Nat. Med. 2018, 24, 1449–1458. [Google Scholar] [CrossRef]
  168. Wang, Z.; Duan, J.; Cai, S.; Han, M.; Dong, H.; Zhao, J.; Zhu, B.; Wang, S.; Zhuo, M.; Sun, J.; et al. Assessment of Blood Tumor Mutational Burden as a Potential Biomarker for Immunotherapy in Patients with Non–Small Cell Lung Cancer with Use of a Next-Generation Sequencing Cancer Gene Panel. JAMA Oncol. 2019, 5, 696–702. [Google Scholar] [CrossRef]
  169. Rizvi, N.A.; Cho, B.C.; Reinmuth, N.; Lee, K.H.; Ahn, M.-J.; Luft, A.; van den Heuvel, M.; Cobo, M.; Smolin, A.; Vicente, D.; et al. Durvalumab With or Without Tremelimumab vs. Standard Chemotherapy in First-line Treatment of Metastatic Non-Small Cell Lung Cancer: The MYSTIC Phase 3 Randomized Clinical Trial. JAMA Oncol. 2020, 6, 661–674. [Google Scholar] [CrossRef] [Green Version]
  170. Si, H.; Kuziora, M.; Quinn, K.J.; Helman, E.; Ye, J.; Liu, F.; Scheuring, U.; Peters, S.; Rizvi, N.A.; Brohawn, P.Z.; et al. A Blood-based Assay for Assessment of Tumor Mutational Burden in First-line Metastatic NSCLC Treatment: Results from the MYSTIC Study. Clin. Cancer Res. 2020, 27, 1631–1640. [Google Scholar] [CrossRef]
  171. Stadler, J.-C.; Belloum, Y.; Deitert, B.; Sementsov, M.; Heidrich, I.; Gebhardt, C.; Keller, L.; Pantel, K. Current and Future Clinical Applications of ctDNA in Immuno-Oncology. Cancer Res. 2022, 82, 349–358. [Google Scholar] [CrossRef] [PubMed]
  172. Ba, H.; Liu, L.; Peng, Q.; Chen, J.; Zhu, Y.-D. The relationship between blood-based tumor mutation burden level and efficacy of PD-1/PD-L1 inhibitors in advanced non-small cell lung cancer: A systematic review and meta-analysis. BMC Cancer 2021, 21, 1220. [Google Scholar] [CrossRef] [PubMed]
  173. Wang, Z.; Zhao, X.; Gao, C.; Gong, J.; Wang, X.; Gao, J.; Li, Z.; Wang, J.; Yang, B.; Wang, L.; et al. Plasma-based microsatellite instability detection strategy to guide immune checkpoint blockade treatment. J. Immunother. Cancer 2020, 8, e001297. [Google Scholar] [CrossRef] [PubMed]
  174. Willis, J.; Lefterova, M.I.; Artyomenko, A.; Kasi, P.M.; Nakamura, Y.; Mody, K.; Catenacci, D.V.; Fakih, M.; Barbacioru, C.; Zhao, J.; et al. Validation of Microsatellite Instability Detection Using a Comprehensive Plasma-Based Genotyping Panel. Clin. Cancer Res. 2019, 25, 7035–7045. [Google Scholar] [CrossRef] [PubMed]
  175. Georgiadis, A.; Durham, J.N.; Keefer, L.A.; Bartlett, B.R.; Zielonka, M.; Murphy, D.; White, J.R.; Lu, S.; Verner, E.L.; Ruan, F.; et al. Noninvasive Detection of Microsatellite Instability and High Tumor Mutation Burden in Cancer Patients Treated with PD-1 Blockade. Clin. Cancer Res. 2019, 25, 7024–7034. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Advantages and disadvantages of tumor genomic profiling using tumor tissues and plasma cfDNA.
Figure 1. Advantages and disadvantages of tumor genomic profiling using tumor tissues and plasma cfDNA.
Cancers 14 03275 g001
Figure 2. Generalized guidelines for CGP assay selection under different clinical situations. The schema was devised based on recommended guidelines from NCCN, ESMO, ASCO and IASLC together with existing literature. Patients with advanced solid tumors with known FDA-approved targeted therapies should undergo tissue-based genomic profiling for treatment selection. In cases where biopsy and tumor tissues are insufficient or unfeasible, patients present with multiple lesions or patients with aggressive, fast-progression cancers, cfDNA-based genomic profiling should be considered. CGP using cfDNA-based assays should be considered the assay of choice for patients who have developed recurrence or disease progression after targeted therapies. However, for patients with cancer types known to be low shedding of ctDNA and with contemporary tissues available, a tissue-based assay should be preferred. Patients with negative ctDNA results should reflex to tissue testing in all cases.
Figure 2. Generalized guidelines for CGP assay selection under different clinical situations. The schema was devised based on recommended guidelines from NCCN, ESMO, ASCO and IASLC together with existing literature. Patients with advanced solid tumors with known FDA-approved targeted therapies should undergo tissue-based genomic profiling for treatment selection. In cases where biopsy and tumor tissues are insufficient or unfeasible, patients present with multiple lesions or patients with aggressive, fast-progression cancers, cfDNA-based genomic profiling should be considered. CGP using cfDNA-based assays should be considered the assay of choice for patients who have developed recurrence or disease progression after targeted therapies. However, for patients with cancer types known to be low shedding of ctDNA and with contemporary tissues available, a tissue-based assay should be preferred. Patients with negative ctDNA results should reflex to tissue testing in all cases.
Cancers 14 03275 g002
Table 1. Common ctDNA detection platforms.
Table 1. Common ctDNA detection platforms.
MethodNameExampleNumber of TargetsLOD
PCR-basedqPCRCOLD-PCR10.1–1%
dPCRBEAMing1–20 targets0.01–0.1%
dPCRddPCRup to 5 targets0.01–0.1%
MassSpec PCRUltraSeekMultigenes0.1–1%
NGS-basedAmplicon-basedIonTorrent-OncomineMultigenes0.1%
Safe-SeqS; Plasma-SeqSenseiMultigenes0.04–0.2%
Hybrid captureAvenio, TruSight 500Multigenes0.5%
CAPP-Seq; Guardant360; FoundationOne LiquidMultigenes0.02%
PCR: Polymerase Chain Reaction; ddPCR: droplet digital PCR; qPCR: quantitative PCR; NGS: Next-generation sequencing; COLD-PCR: Co-amplification at lower denaturation temperature PCR; BEAMing: Beads, emulsion, amplification, magnetics; LOD: Limit of detection.
Table 2. Summary of recently published studies on the mutation detection concordance between tumor tissues and ctDNA.
Table 2. Summary of recently published studies on the mutation detection concordance between tumor tissues and ctDNA.
Cancer TypeSample SizeMethodNumber of GenesTypes of VariantsDetection RateDetection of Actionable Mutations from ctDNACH EliminationTissue Plasma Concordance *CommentsReference
Pan Cancer11,525Customized hybridization capture NGS1021SNVs, Indels, CNVs, Fusion, bTMB73.50%41.2%YesN.D [40]
Pan Cancer681MSK-ACCESS hybridization capture NGS129SNVs, Indels, CNVs, Fusion, bTMB73%56.0%Yes59.0%Variable collection interval between tissue and plasma[63]
Pan Cancer433Guardant 360 hybridization capture NGS73SNVs, Indels, CNVs, Fusion37%N.D.No45.0%Only examined TP53; variable collection interval between tissue and plasma[65]
Pan Cancer161Customized hybridization capture NGS (GRAIL)508SNVs, Indels, CNVs, Fusion84%N.D.Yes72.0% [42]
Pan Cancer10,593Guardant 360 hybridization capture NGS73SNVs, Indels, CNVs, Fusion86%72.0%No92.0%Concordance based on 543 patients, in 7 genes[57]
Lung1971Guardant 360 hybridization capture NGS73SNVs, Indels, CNVs, Fusion87.30%26.70%NoN.D. [66]
Lung262Guardant 360 hybridization capture NGS73SNVs, Indels, CNVs, Fusion60.40%60.40%No67.70%Initial diagnosis/treatment naïve; only examined 6 genes (EGFR, ALK, MET, ROS1, RET, KRAS)[67]
Lung934FoundationLiquid/FoundationACT NGS62/70SNVs, Indels, CNVs, Fusion90.00%20.00%NoN.D.ctDNA: 937 patients; Tissue: 5582 patients[68]
Lung8388Guardant 360 hybridization capture NGS73SNVs, Indels, CNVs, Fusion86.00%48.00%NoN.D. [69]
Lung282Guardant 360 hybridization capture NGS/ddPCR73SNVs, Indels, CNVs, Fusion-27.30%No80.0% [53]
Lung127Customized hybridization capture NGS (GRAIL)37SNVs, Indels, CNVs, Fusion--Yes75.0% [56]
Lung210ResBio ctDx-Lung amplicon-based NGS21SNVs, Indels, CNVs, Fusion64.30%21.90%No60.6%A subset of patients subjected to treatment at the time of plasma collection[70]
Lung323Guardant 360 hybridization capture NGS73SNVs, Indels, CNVs, Fusion-33.00%NoN.D. [64]
Breast162Customized amplicon-based NGS39SNVs92.50%39.00%NoN.D. [71]
Breast1044Guardant 360 hybridization capture NGS/ddPCRddPCR: 4; NGS: 73SNVs, Indels, CNVs, Fusion51.10%34.50%No93%Concordance is based on 77 patients in 4 genes (AKT1, HER2, ESR1, PIK3CA). Negative concordance was included.[54]
Breast255Guardant 360 hybridization capture NGS73SNVs, Indels, CNVs, Fusion89.00%26.00%No79–91%Actionable alterations in PIK3CA, ESR1, ERBB2[55]
Gastrointestinal1687Guardant 360 hybridization capture NGS73SNVs, Indels, CNVs, Fusion91.40%57.30%No8.3–80.3%
(<0.3 vs. >0.3 clonality)
Concordance is based on 287 patients[59]
Gastrointestinal200Customized amplicon-based NGS150SNVs, Indels, CNVs, Fusion, bTMB, bMSI84.05%45.50%NoN.D. [72]
Gastrointestinal1064Guardant 360 hybridization capture NGS73SNVs, Indels, CNVs, Fusion93.70%47.70%NoN.D.Only included metastatic colorectal cancer patients[73]
Gastrointestinal282Guardant 360 hybridization capture NGS73SNVs, Indels, CNVs, Fusion75.00%48.00%No50–86% [74]
Prostate3334FoundationLiquid/FoundationACT NGS62/70SNVs, Indels, CNVs, Fusion79.50%30% (DDR gene alteration)Yes75.3% (SNVs); 70.3% (rearrangements); 27.5% (CNVs)DDR alterations: BRCA1/2; CDK12; MSI-H[58]
* % of mutations detected from tumor tissues also detected from plasma cfDNA, unless stated. CH: clonal hematopoiesis; ddPCR: droplet digital PCR; N.D.: not determined; SNVs: single nucleotide variants; CNVs: copy number variations; bTMB: blood tumor mutation burden; DDR: DNA damage response and repair; MSI-H: microsatellite instability high.
Table 3. Summary of FDA-approved diagnostic plasma ctDNA assays.
Table 3. Summary of FDA-approved diagnostic plasma ctDNA assays.
Approved Diagnostic ToolTechnologyNumber of GenesInput (ng)DiseaseDrugBiomarkerLOD
Cobas EGFRRT-PCR1Undefined; (2 mL of plasma)NSCLCErlotinib & GefitinibEGFR Exon 19 deletions; L858RExon 19 deletions: 0.1–0.5%; L858R: 0.4–0.8%
OsimertinibEGFR T790MExon 19 deletions: 0.1–0.5%; L858R: 0.4–0.8%;
T790M: 0.4–0.8%
TherascreenRQT-PCR1Undefined; (2 mL of plasma)BreastAlpelisibPIK3CA (C420R, E542K, E545A, E545D [1635G > T only], E545G, E545K, Q546E, Q546R; and H1047L, H1047R, and H1047Y)1.82–7.07%
FoundationOne Liquid CDxNGS-hybridization enrichment324 (311 FDA approved)20NSCLCAlectinibALK rearrangements: ALK-EML4ALK-EML4: 0.24%
Osimertinib & ErlotinibEGFR Exon 19 deletions; L858RExon 19 deletions: 0.27%; L858R: 0.34%
CapmatinibMET SNVs & Indels that lead to MET exon 14 skippingSubstitutions: 0.4%;
Indels: 0.28%
ProstateOlaparibBRAC1Substitutions: 0.34%;
Indels: 0.38%
BRCA2Substitutions: 0.37%;
Indels: 0.36%;
ATM alterationsIndels: 0.51%
RucaparibBRCA1Substitutions: 0.34%;
Indels: 0.38%
BRCA2Substitutions: 0.37%;
Indels: 0.36%
OvarianRucaparibBRCA1Substitutions: 0.34%;
Indels: 0.38%
BRCA2Substitutions: 0.37%;
Indels: 0.36%
BreastAlpelisibPIK3CA (C420R, E542K, E545A, E545D [1635G > T only], E545G, E545K, Q546E, Q546R; and H1047L, H1047R, and H1047Y)Substitutions: 0.34%
Guardant360 CDxNGS- hybridization enrichment74 (55 FDA approved)30NSCLCOsimertinibEGFR Exon 19 deletions; L858R; T790M0.20%
Amivantamab-vmjwEGFR exon 20 insertions0.30%
SotorasibKRAS G12C0.50%
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Chan, H.T.; Chin, Y.M.; Low, S.-K. Circulating Tumor DNA-Based Genomic Profiling Assays in Adult Solid Tumors for Precision Oncology: Recent Advancements and Future Challenges. Cancers 2022, 14, 3275. https://doi.org/10.3390/cancers14133275

AMA Style

Chan HT, Chin YM, Low S-K. Circulating Tumor DNA-Based Genomic Profiling Assays in Adult Solid Tumors for Precision Oncology: Recent Advancements and Future Challenges. Cancers. 2022; 14(13):3275. https://doi.org/10.3390/cancers14133275

Chicago/Turabian Style

Chan, Hiu Ting, Yoon Ming Chin, and Siew-Kee Low. 2022. "Circulating Tumor DNA-Based Genomic Profiling Assays in Adult Solid Tumors for Precision Oncology: Recent Advancements and Future Challenges" Cancers 14, no. 13: 3275. https://doi.org/10.3390/cancers14133275

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