Open Access

Liquid biopsy: Comprehensive overview of circulating tumor DNA (Review)

  • Authors:
    • Qian Ge
    • Zhi-Yun Zhang
    • Suo-Ni Li
    • Jie-Qun Ma
    • Zheng Zhao
  • View Affiliations

  • Published online on: September 13, 2024     https://doi.org/10.3892/ol.2024.14681
  • Article Number: 548
  • Copyright: © Ge et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Traditional tumor diagnosis methods rely on tissue biopsy, which can be invasive and unsuitable for long‑term monitoring of tumor dynamics. The advent of liquid biopsy has notably improved the overall management of patients with cancer. Liquid biopsy techniques primarily involve detection of circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA). The present review focuses on ctDNA because of its significance in tumor diagnosis, monitoring and treatment. The use of ctDNA‑based liquid biopsy offers several advantages, including non‑invasive or minimally invasive collection methods, the ability to conduct repeated assessment and comprehensive insights into tumor biology. It serves crucial roles in disease management by facilitating screening of high‑risk patients, dynamically monitoring therapeutic responses and diagnosis. Furthermore, ctDNA can be used to demonstrate pseudo‑progression, monitor postoperative tumor status and guide adaptive treatment plans. The present study provides a comprehensive review of ctDNA, exploring its origins, metabolism, detection methods, clinical role and the current challenges associated with its application.

Introduction

Currently, pathological tissue biopsy is the gold standard for diagnosing and monitoring a number of malignant tumors, such as lung cancer, stomach cancer, colorectal cancer. This method allows rapid assessment of the extent and nature of lesions, provides relatively accurate pathological classification and facilitates early detection and diagnosis. In addition, tissue biopsies conducted during treatment can reflect disease progression and treatment efficacy and provide clinicians with valuable information to tailor treatment plans (1). However, use of insufficient or inadequate tissue samples may lead to diagnostic bias, which may be exacerbated by tumor heterogeneity (2). Furthermore, repeated invasive procedures can cause discomfort for patients, particularly as the disease advances.

The emergence of liquid biopsy has effectively addressed several of these challenges. Liquid biopsy involves the molecular analysis of liquid (non-tissue) samples to evaluate physiological states (3). While blood samples are the most commonly used, other bodily fluids such as cerebrospinal fluid (CSF), saliva, pleural effusion, bile, abdominal fluid and urine can also be utilized (4). Liquid biopsy primarily focuses on analyzing circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), as well as circulating cell-free (cf)RNA, extracellular vesicles and tumor-inducing platelets (5). ctDNA has emerged as a pivotal element in clinical practice as it serves a key role in cancer diagnosis, monitoring and treatment. In clinical treatment, physicians are able manage treatment strategies by reference to ctDNA results. The detection of ctDNA pre-treatment and pre- and post-operation guide patient treatment plans (6). However, there are limitations to ctDNA analysis in liquid biopsy, including challenges in evaluating tumor pathology, detection sensitivity and the absence of standardized protocols.

Overview of ctDNA-based liquid biopsy

cfDNA

Circulating cfDNA refers to highly fragmented DNA released from cells into the bloodstream that circulates freely in human blood (7). First discovered in 1948 (7), cfDNA has become a major focus of medical research (810). It is composed of both double- and single-stranded fragments (1113), typically 120–220 bp in length, with an average length of ~167 bp, which is associated with the nucleosome (14).

In the bloodstream, cfDNA exists in three primary forms: Free, bound to protein (such as nucleosomes and lipoproteins) or associated with extracellular vesicles, such as exosomes, apoptotic bodies and microvesicles (15,16). The majority of plasma cfDNA is found in exosomes (17). The sources of cfDNA have been a topic of debate and can be generally divided into two main categories: Cellular destruction and active cellular release (18,19). Potential sources of cfDNA include cellular byproducts released during normal physiological processes, exogenous DNA originating from dietary intake, blood transfusions or infections, release from the nervous system, secretion into the blood circulation due to factors such as stress, hereditary conditions, degeneration or disease, fetal cellular material transferred to the mother during pregnancy and systemic release due to obesity and aging (19). Plasma cfDNA from healthy individuals predominantly originates from white blood cells (55%), red blood cell progenitors (30%) and vascular endothelial (10%) and liver cells (1%) (19). In patients with cancer, however, cfDNA is mainly derived from tumor tissue and surrounding cells (18).

cfDNA is a key biomarker for various physiological and pathological conditions and is associated with factors such as aging (20) and physical or psychological stress (21). In addition, cfDNA serves as a key biomarker in several types of cancer, including non-small cell lung cancer (NSCLC) (22), liver (23), breast (24,25), pancreatic (26), oral (27) and colorectal cancer (28,29). Previous studies suggest that cfDNA may also be associated with xenotransplantation (30,31).

cfDNA encompasses short and long DNA fragments and analysis of these fragments can provide valuable insight into a health status. Increasing evidence shows that cfDNA is vital for immune regulation, tumor-associated inflammation and the maintenance of cell homeostasis (32,33). cfDNA can impact cellular function and transformation, contribute to tumor growth and metastasis and holds promise for early disease detection (32,33). In healthy individuals, there is a dynamic balance between production and clearance of cfDNA. cfDNA is primarily cleared by the liver, spleen and kidney (34,35). However, in patients with chronic inflammation or tumors, cfDNA levels significantly increase due to impaired clearance and subsequent accumulation (32).

ctDNA

ctDNA is a subset of cfDNA that consists of DNA fragments that originate from tumor tissue and potentially other sources, such as shedding cells from normal tissue, characterized by genetic alterations that mirror those of the tumor (18,36). The increase in cfDNA levels in patients with cancer is primarily attributable to the elevated levels of ctDNA (37). Typically, ctDNA fragments are ~140 bp in length and have a half-life >2 h, which makes their dynamics variable (38,39). The detection rate of ctDNA in plasma ranging from 0.01% to a majority in the cfDNA, which reflects notable heterogeneity among different types of tumor (40,41). Despite this variability, the quantity of ctDNA detected is usually high compared with that of CTCs (42). The ability to detect ctDNA is influenced by characteristics of the tumor. For example, smaller solid tumors or those with low metabolic activity may be more challenging to detect, which may lead to false-negative results (6). Notably, ctDNA levels decrease rapidly following radical tumor resection if there is no or minimal residual tumor (43).

ctDNA detection in biological samples: Blood vs. non-blood sources

Blood

Blood is the most widely used sample type for detection of ctDNA (44), primarily due to the minimally invasive collection process and reproducibility. However, the precise mechanisms by which ctDNA enters the bloodstream remain unclear. It is has been hypothesized that ctDNA in blood originates from three primary sources: Apoptotic or necrotic and viable tumor cells and CTCs (45). Plasma is considered the optimal sample for ctDNA analysis, as serum (excluding clotting factors) contains an increased proportion of DNA from leukocyte lysis (46). For example, Heger et al (47) developed a molecular prognostic index for central nervous system (CNS) lymphoma using plasma ctDNA and demonstrated that is was effective in predicting patient outcomes. This ultra-sensitive method can detect CNSL-derived mutations in plasma ctDNA that are highly consistent with CSF and tumor tissue. Plasma ctDNA undetectable at baseline was associated with favorable outcomes. However, detecting ctDNA in white blood cells poses challenges due to high levels of cfDNA in these cells, which leads to notable dilution of the ctDNA (48). Compared with blood samples, ctDNA is easier to detect in non-blood samples.

CSF

Acquiring brain tissue for diagnostic purposes is both challenging and high-risk due to the unique structure and function of the brain. Therefore, CSF serves an irreplaceable role. While magnetic resonance imaging (MRI) is commonly used to monitor CNS diseases (49), its predictive capability is limited. Liquid biopsy that involves the detection of ctDNA in CSF may serve as a novel detection method for CNS diseases. The unique composition of CSF, along with the protective blood-brain barrier, decreases interference from cfDNA, which results in increased concentration of ctDNA and mutated allele frequencies (MAF) in CSF. This improves the sensitivity and accuracy of ctDNA mutation detection. Consequently, CSF biopsy may be a promising diagnostic tool for the detection and monitoring of brain tumors and CNS metastases (5052).

A study has shown that carcinoembryonic antigen and CSF ctDNA are effective biomarkers for distinguishing patients with and without brain parenchymal tumor or CNS metastases (53). CSF ctDNA analysis has demonstrated distinct mutational profiles in patients with bone marrow metastasis (53). In a 2022 case report (54), a patient with lung adenocarcinoma experienced neurological symptoms, including headache, nausea, aphasia, limb restlessness and sudden blindness during treatment. While initial MRI and CSF cytology did not indicate the presence of brain metastasis, CSF ctDNA analysis identified the same EGFR mutation as that detected in the lung tumor of the patient. Follow-up MRI scans 9 months later confirmed brain metastasis, which suggested earlier MRI and CSF cytology results were false negatives. This case underscores the potential of CSF ctDNA as an early diagnostic biomarker as it may detect brain metastasis before cytological or MRI evidence emerges.

To the best of our knowledge, studies of ctDNA in CSF have predominantly focused on adults (5558), with relatively few investigating its application in children (5961). Pages et al (62) assessed tumor reliability by analyzing ctDNA in peripheral blood, CSF and urine samples from children with brain tumors; ctDNA detection in this demographic was limited by low tumor fraction (TF). In numerous cases, the actual TF was >1%, with TF >0.1% being undetectable. Furthermore, only a small percentage of high-grade tumors were available. Therefore, uncertainty persists regarding the application of liquid biopsy for pediatric tumors, indicating the need for further research.

Saliva and sputum

The clinical relevance of saliva as a diagnostic tool is uncertain. A study from 2019 (63) suggested that saliva may not serve as an adequate alternative sample for quantitative cfDNA testing due to insufficient cfDNA concentration for diagnosis of NSCLC. However, it was proposed that saliva may be beneficial as a complementary method to cytology. Wang et al (64) made notable advancements by optimizing the extraction technique for sputum samples to overcome limitations posed by large amounts of mucus components and the low yield of cancer cells. Super-amplification refractory mutation system was used to analyze EGFR mutation status in cfDNA derived from sputum samples; sputum could be a promising sample type for detecting EGFR mutations, although its use for diagnosing lung adenocarcinoma may be limited. Further research by Wang et al (65) highlighted saliva from patients with lung adenocarcinoma as a valuable alternative source for detecting the EGFR exon 20 p.T790M mutation, which is linked to resistance to EGFR targeted therapy (65). In addition, a previous study investigating head and neck cancer reported a high concordance (93%) in ctDNA detection between saliva and blood samples, as well as efficacy of ctDNA in saliva in predicting patient outcomes (66). A meta-analysis of 64 cases of malignant salivary gland carcinoma found increased levels of ctDNA and CTCs in malignant cases (67). According to the 2024 Expert Consensus, ctDNA extracted from saliva, along with serum or plasma, provides meaningful insight into tumor genetics and dynamics (68).

Pleural, peritoneal and pericardial effusion

Tumor supernatant, such as pleural, peritoneal and pericardial effusion, are in proximity to tumors and may provide distinct advantages over blood for ctDNA detection; for example, it is easier to detect ctDNA of abdominal tumor with abdominal fluid A previous study that compared mutant allele scores from 30 supernatant samples with those from paired formalin-fixed paraffin-embedded cell blocks reported a variant concordance up to 90% (69), and similar results were detected in both supernatant and FFPE samples in 74% of cases. This suggests that supernatant may serve as a viable alternative to traditional tissue biopsy (69).

Yang et al (70) used high-throughput next generation sequencing (NGS) to analyze cfDNA in 15 pleural, five abdominal and one pericardial effusion; they identified key pathogenic mutations in malignant fluid from 13 patients with metastatic tumors, potentially malignant fluid from two cases and benign fluid from one case. In another study that focused on peritoneal cancer, malignant ascites or peritoneal lavage fluid was collected for microdroplet digital PCR (ddPCR) analysis; peritoneal effusion cfDNA could predict the tumor load of peritoneal cancer and assess patient eligibility for cytoreductive surgery by calculating the MAF (71). Compared with blood, fluids such as pleural and pericardial effusion and ascites exhibit increased sensitivity for ctDNA testing and may serve as predictive biomarkers for responses to EGFR inhibitors (48).

Pancreatic fluid and bile

In pancreaticobiliary tract tumors, obtaining tissue biopsies can be challenging due to the occult nature of the disease. Given their direct contact with tumor tissue, bile and pancreatic fluid are ideal samples for liquid biopsies. Kinugasa et al (72) compared levels of ctDNA in tumor tissue with those in bile from 49 patients with gallbladder cancer; ctDNA isolation from bile was a valuable approach for diagnosing gallbladder cancer. The sensitivity of ctDNA testing (58.3%) was higher compared with that of cytology (45.8%), and there was a high mutation concordance between the two methods. Further study has demonstrated consistent KRAS mutations in ctDNA from bile, plasma and formalin-fixed paraffin-embedded samples from patients with bile duct tumors (39). Notably, only 18.8% of plasma ctDNA samples test positive for KRAS mutations, compared with a detection rate of 48.0% in bile ctDNA (39). Moreover, patients with KRAS mutations detected in bile ctDNA exhibit significantly lower survival rates compared with those with wild-type KRAS (39).

In early-stage pancreatic ductal adenocarcinoma, identification of mutations in plasma ctDNA is often challenging (73). A study in 2023 compared the detection rates of ctDNA sourced from pancreatic fluid with that in plasma; DNA concentrations and the ratios of Alu247/Alu115 were higher in pancreatic fluid compared with plasma, however, there was no difference in the mutation detection rate between pancreas and plasma (74). This limitation may be attributable to the small sample size and influence of enzymes present in pancreatic fluid, which underscores the need for further investigation.

Urine

In 1995, ctDNA was detected in urine (75), marking the beginning of research interest in this non-invasive biomarker. Thus far, two primary sources of ctDNA in urine have been identified, including debris shed directly from tumor cells within the urinary system and CTCs that are filtered through the kidney (76,77). The latter source of ctDNA tends to have a smaller molecular weight, restricted by pore size of the glomerular barrier (76). In 2008, a comparative study examined KRAS mutations in both blood and urine samples from patients with colon cancer; Although the study had a small sample size, mutation rates were similar in both fluids. However, as the sample size increased, mutation rates in urine became significantly higher compared with those in blood (78), highlighting potential of urine as a tumor marker.

A recent prospective multi-center study reported that measurement of DNA methylation in urine effectively differentiates pathological types of bladder cancer and predicts 180-day recurrence-free survival with 100% accuracy (79). This represents a breakthrough in use of urine DNA methylation for differentiation of pathological cancer types. In addition, a study on ctDNA in urine during neoadjuvant chemotherapy for bladder cancer demonstrated that monitoring tumor DNA dynamics in urine, supernatant and plasma predicts treatment response and outcome (80). Urinary ctDNA has also been shown to detect the recurrence of upper urinary tract urothelial carcinoma up to 60 days earlier than cystoscopy (81). Kim et al (82) reported that binding urinary ctDNA improves detection rate of hepatocellular carcinoma from 62 to 92% (82). Increased DNA methylation levels in urine have also been reported in patients with NSCLC.

Urine sampling is a non-invasive procedure that can be performed by non-professionals. Patients can conveniently collect samples at home, which can be sent to testing laboratories. Unlike blood, which is subject to buffering and regulatory mechanisms that may alter its properties (83), urine is excreted and may better reflect bodily abnormality. In addition, urine has a lower presence of contaminating proteins compared with blood, which simplifies DNA extraction processes (76). The ability to test large volumes of urine repeatedly enhances sensitivity of ctDNA detection (84), which makes it a promising tool in cancer diagnostics.

Semen

To the best of our knowledge, prostate-specific antigen is the only well-established tumor marker for prostate cancer. However, its specificity is limited, necessitating exploration of additional diagnostic tools. ctDNA testing has emerged as a valuable adjunct in diagnosis of prostate cancer (8587). While ctDNA is commonly detected in advanced cancer such as pancreatic, ovarian, colorectal, bladder, gastroesophageal, breast, hepatocellular and head and neck cancer and melanoma, it is present in <50% of cases of primary brain, kidney, prostate and thyroid cancer (40).

Semen may serve as a potential sample for prostate cancer diagnostics. Significant differences in cfDNA levels have been observed (83) in semen samples from patients with prostate cancer, individuals with benign prostatic hyperplasia and healthy individuals (88). Patients with prostate cancer exhibit higher concentrations of cfDNA than healthy individuals in semen alongside a distinctive size distribution of cfDNA fragments. Notably, longer cfDNA fragments are significantly more prevalent in semen from patients with prostate cancer compared with those with benign hyperplasia or healthy individuals (69). Size and concentration of cfDNA fragments in semen is associated with tumor burden and treatment response (88). As a direct source of prostate disease-specific molecules, semen represents a promising body fluid for identification of prostate cancer biomarkers and may provide important insight for early diagnosis (Fig. 1) (89).

Detection methods

ctDNA provides insight for cancer diagnosis and treatment. To harness the potential of ctDNA, accurate detection methods are key. The main techniques for ctDNA detection include DNA sequencing, PCR-based methods and DNA-based hybridization strategies (9092). At present there is no universally accepted standard of detection (93).

DNA sequencing
Directed error correction sequencing

Phallen et al (94) developed targeted error correction sequencing (TEC-Seq), which enables ultra-sensitive evaluation of sequence changes in ctDNA through large-scale parallel sequencing. TEC-Seq uses targeted capture of multiple genomic regions combined with deep sequencing of DNA fragments, facilitating the detection of 58 cancer-associated genes spanning 81 kb. A plasma analysis conducted on 44 healthy individuals reported that 16% of asymptomatic individuals exhibit genomic changes associated with clonal hematopoiesis, although none show alterations in driver genes linked to solid tumors (94). CancerSEEK assay (multicancer early detection blood test) demonstrates an accurate tissue-of-origin prediction in 83% of cases, while TEC-Seq yields cancer detection rates of 59–71%, depending on the type of cancer assessed (95). Achieving effective sensitivity in ctDNA analysis using TEC-Seq presents a notable technical challenge (94), which reflects the need for continued refinement of this approach to enhance its applicability in clinical settings.

NGS

Since US Food and Drug Administration (FDA) approval of the Guardant360® CDx (Guardant Health, Inc.) for use of third-generation tyrosine kinase inhibitor osimertinib in patients with NSCLC and EGFR mutations, the NGS approach has rapidly evolved (96). NGS is the preferred liquid biopsy technique for metastatic NSCLC according to the current European Society for Medical Oncology guidelines (97). This high-throughput sequencing method enables comprehensive analysis of DNA and RNA, which allows examination of the entire sequence of target genes and facilitates detection of a broader spectrum of mutations, including previously unknown variants (97,98). NGS can simultaneously sequence multiple genomes on a single platform, even when working with low concentrations of tumor DNA derived from plasma or other liquid biopsy samples (99). NGS can be tailored to analyze a variable number of regions, from a few loci to the entire exome or genome (100). Broadly, NGS targeting ctDNA uses two main approaches. The first approach involves deep sequencing of specific regions containing relevant mutations, which offers high sensitivity and specificity suitable for targeted clinical application. The second approach entails whole-exome or whole-genome sequencing, which can uncover novel genomic changes and is more suitable to basic research (98). NGS is characterized by high throughput, sensitivity and specificity, rendering NGS-based ctDNA mutation spectrum analysis superior to other technologies (98) such as TEC-Seq. However, there are challenges associated with NGS, including the potential for mislocalisation of mutations (97). In addition, this technology requires extensive data analysis, incurs notable costs and has a turnaround time of 7–14 days (101,102).

Targeted amplicon sequencing (TAm-Seq)

TAm-Seq is a labeled amplicon deep sequencing method that integrates efficient library preparation with advanced statistical analysis. This technique enables the sequencing of ~6,000 nucleotides and in-depth analysis (103). A notable implementation of TAm-Seq is InVisionFirst® (Neogenomics Laboratories).

Liquid biopsy platform, which is designed to detect both hotspot mutations and entire coding regions across 35 cancer-associated genes. Leveraging enhanced TAm-Seq techniques, it identifies low-frequency mutations in ctDNA by amplifying highly fragmented DNA (104). Further improvements of Tm-Seq involves optimization of the amplification process by splitting it into two steps. The initial step involves limited cycle pre-amplification using all primer sets to capture the starting molecules present in the template. This is followed by a single amplification step to purify and isolate the target sequence. This refined approach enables detection of cancer mutations in ctDNA at allelic frequencies as low as 2% with sensitivity and specificity of >97% (105). TAm-Seq method demonstrates its utility in clinical settings by routinely detecting ctDNA not only at the time of diagnosis, but also post-treatment (106).

Deep sequencing personalized cancer analysis (CAPP-Seq)

Developed by Newman et al (107), CAPP-Seq is an economical and ultra-sensitive method for quantifying ctDNA. This method integrates a low DNA initiation mass library preparation strategy with a multiphase bioinformatics approach to generate a ‘selector’ of biotinylated DNA oligonucleotides designed to target tumor regions with recurrent mutations. To monitor ctDNA, the selector is initially applied to tumor DNA to identify unique cancer-specific genetic aberrations. Subsequently, it is used on ctDNA to quantify these aberrations (107). Among the various NGS-based ctDNA analysis methods, CAPP-Seq has the lowest background error rate and detection limit (107), demonstrating greater sensitivity compared with TAm-Seq (106). Originally intended for analysis of NSCLC (107), CAPP-Seq has since been successfully applied to a variety of other types of cancer, including esophageal (108) and ovarian cancer (109), mantle cell lymphoma (110), bladder cancer (111), head and neck squamous cell carcinoma (112) and melanoma (113). However, despite its high sensitivity, CAPP-Seq has higher cost compared with TAm-Seq and may not offer advantages for routine screening and surveillance (106).

PCR

Real-time quantitative PCR (qPCR) qPCR can be used for endometrial cancer (114), non-small cell lung cancer (115), colorectal cancer (116). qPCR-based tests have received approval from US FDA and European Medicines Agency for detection, activation or identification of resistance to EGFR targeted therapies in NSCLC (117). Despite its widespread use, the simple nature of these tests may increase the risk of false positives when qPCR is used in isolation (97).

Microdroplet ddRCR

Microdroplet ddPCR, also known as third-generation PCR, uses sample allocation, restricted dilution and statistical data processing based on Poisson distributions to accurately and reliably quantify nucleic acids. This technique divides mixed nucleic acid molecules and PCR solution into small droplets. By using microfluidic loops and surfactant chemistry, sample DNA is randomly assigned to isolated droplets, generating 20,000 droplets. The template is amplified and product is detected based on specific fluorescent labeling (97,118,119). ddPCR is well-suited for studying specific single-gene hotspot mutations that may be found in CSF samples, achieving a limit of detection as low as 0.01%/reaction (119,120). It can measure mutations that constitute 0.01% of a sample (39), offering greater sensitivity compared with qPCR (97). However, compared with NGS, ddPCR has narrower reference range (90.0% of operable mutations) and does not cover certain variants, such as estrogen receptor 1 mutation (121). In addition, ddPCR requires specialized personnel to operate, which adds to overall complexity and operational costs (39,118,122).

Beads, emulsion, amplification, magnetics (BEAMing)

BEAMing is a digital PCR method that enhances the capability of ddPCR by incorporating pre-amplifications of DNA using conventional PCR and target-specific primers (97). PCR products amplified by BEAMing molecules are linked to single magnetic beads and the mutation sites extended via fluorescent probes or primers. By counting fluorescently labelled beads, BEAMing allows the quantitative detection of mutant alleles (119,123). Taniguchi et al (123) used BEAMing to monitor disease progression in patients with lung cancer undergoing EGFR targeted therapy, which effectively determined the proportion of T790M-positive alleles in cancer cells, regardless of potential contamination from normal cell DNA.

Thermal coupling index expansion

An ultra-fast monitoring method known as thermal coupling exponential amplification test has been recently reported (124). This technique combines exponential amplification reaction (EXPAR) with Thermus thermophilus argonaute-coupling), from the thermophilic bacterium Thermus thermophilus, to quickly and accurately detect ctDNA in ~16 min (124). A previous study on tumor threshold changes in mouse models (seven Kirsten rat sarcoma-2 virus (KRAS) point mutations) indicated that this method holds significant potential for monitoring tumor load and evaluating chemotherapy response (121). TtAgo-CEAR assay leverages rapid, specific cleavage function of TtAgo and the high amplification efficiency of EXPAR to identify common hotspot mutations in KRAS (124).

DNA hybridization

Traditional methods for detecting and quantifying ctDNA, such as PCR and NGS, are well-established but have limitations (125127). These methods are not suitable for detecting short ctDNA fragments (<100 bp). Moreover, they can be costly, require complex instrumentation, time-consuming due to multiple reaction steps and prone to false positives (128). By contrast, hybrid chain reaction is an isothermal, enzyme-free amplification technique that allows indefinite amplification of signals. This method provides advantages for the detection of small molecules and shows potential for ctDNA detection (129). In 2021, researchers successfully employed a hydrogel-based hybrid chain reaction to amplify small amounts of exosomal microRNA from urine samples, achieving a 35-fold increase in detection sensitivity and effectively distinguishing patients with prostate cancer from normal controls (130). A novel device known as the hybrid chain reactor-driven laboratory fiber optic device has been introduced for ultra-fast and sensitive detection of ctDNA in whole blood. This method is time-efficient, straightforward and cost-effective, as it enables real-time monitoring of ctDNA changes (128) and represents a promising direction for advancing detection capability.

Clinical role of ctDNA

Screening and management of patients with cancer

ctDNA serves a key role in the screening and early diagnosis of various solid types of tumors, particularly among asymptomatic individuals. Phallen et al (94) demonstrated a strong correlation between plasma somatic mutations and tumor changes in patients with stage I or II colorectal, ovarian and breast cancer. This suggested that ctDNA analysis may be instrumental in both early detection and ongoing disease management. In a study of esophageal adenocarcinoma, baseline ctDNA levels were used to identify patients with locally advanced disease at higher risk of relapse (106). This highlights potential of ctDNA not only as a biomarker for early diagnosis but also as a prognostic tool for tailoring treatment and monitoring disease progression.

Dynamic longitudinal monitoring to evaluate prognosis

The role of ctDNA as a prognostic marker has gained recognition in recent years (131133). ctDNA is detected in various solid tumors, with its concentration associated with the stage of the disease (134). In a prospective phase II clinical trial, ctDNA was dynamically monitored every three treatment cycles in five patients with solid tumors undergoing immune checkpoint inhibitor (ICI) treatment. The ctDNA levels were associated with tumor status, demonstrating predictive value both at baseline and following treatment (135). The presence of ctDNA following surgery is strongly indicative of tumor recurrence (6). Gale et al (136) demonstrated that ctDNA could identify residual lesions and predict recurrence in patients with NSCLC. Pre- and post-treatment ctDNA testing is shown to identify patients with NSCLC at high risk for recurrence (136). Monitoring ctDNA levels at baseline, during neoadjuvant and adjuvant therapy and after radical therapy allows clinicians to assess drug response and refine treatment regimens. This dynamic longitudinal monitoring ultimately improves prognostic evaluation and informs clinical decision-making, contributing to improved patient management and outcome.

Effectively identifying false advances

Response Evaluation Criteria in Solid Tumors (RECIST1.1) guidelines are key for assessing tumor progression (137). However, they also have limitations, particularly concerning pseudo-progression, which refers to the transient appearance of space-occupying lesions and edema following treatment. This can often mimic disease progression on radiographical imaging but typically resolves or changes after 4–8 weeks of follow-up (138). In clinical practice, clinicians rely on RECIST guidelines to evaluate disease progression and make treatment decisions. However, ctDNA detection can offer earlier and more accurate indication of disease status compared with imaging, potentially reducing follow-up time and offering better guidance for clinical treatment. Similarly, the emerging concept of ‘hyperprogression’ describes a rapid acceleration in tumor growth that can be induced by ICIs (139). To the best of our knowledge, no studies have reported the association between ctDNA and hyperprogressive disease. Future research should explore this association and its implications.

Detecting molecular residual disease (MRD)

Detection of ctDNA provides extensive information and enables analysis of minimal residual lesions. Early detection of residual ctDNA following local radical treatment can indicate MRD and identify patients at higher risk of recurrence or metastasis (140). Due to limited sensitivity of CTC detection, it is rarely used for MRD evaluation (141). Over the past two decades, early detection of MRD in children with acute lymphoblastic leukemia has significantly improved risk stratification, enhanced treatment for high-risk patients and decreased treatment intensity for those at low risk (142). The potential for MRD detection is established in other malignancies, including acute myeloid (143,144) and chronic lymphocytic leukemia (145), NSCLC (146,147), multiple myeloma (148151), breast cancer (152), melanoma (153), head and neck squamous cell carcinoma (154), follicular lymphoma (155), urothelial carcinoma (156) and colorectal cancer (157).

Guiding treatment escalation and de-escalation

A promising application of ctDNA is its ability to inform decisions regarding treatment escalation and de-escalation (158). Patients with a positive MRD result may be candidates for intensified adjuvant therapy, while those with a negative MRD result may potentially benefit from a reduction in treatment intensity (43). It has been suggested that a key treatment endpoint for colorectal cancer should be complete clearance of ctDNA (43). Currently, a multicenter, prospective, randomized clinical trial is underway to evaluate efficacy of ctDNA-guided adjuvant chemotherapy strategies compared to standard care (158). This aims to assess whether ctDNA-guided treatment adjustments yield superior outcomes in terms of three-year disease-free survival for patients with high-risk stage II and III colorectal cancer (Fig. 2) (158).

Limitations

A notable challenge associated with ctDNA-based liquid biopsy is limited detection capability. The mutation abundance of ctDNA is often lower compared with that in localized tumor tissues, and its detectability is influenced by factors including tumor type and load, anatomical location, cellular turnover and disease stage (141). In the context of early cancer detection, ctDNA levels are particularly low, often causing MAF to fall below detection limits of current methods (159). Thus, improving sensitivity of existing detection methods is key. Employing a combination of DNA analysis from liquid biopsy and tissue samples may improve the overall sensitivity and diagnostic accuracy (160).

Discrepancies in results can arise from the diverse standards and interpretations employed by different ctDNA testing methods and laboratories. There is need for the establishment of standardized testing protocols and interpretative guidelines. It is also important to select the appropriate sampling methods, as improper collection of cfDNA from body fluids can lead to missed detection of ctDNA even with appropriate testing methods (100).

Another barrier is the high cost associated with ctDNA detection technologies and equipment, which hampers comprehensive clinical monitoring and may affect treatment decisions. Furthermore, there is currently no evidence to suggest that ctDNA can fully replace traditional pathological testing. Addressing these challenges is key for refining the role of ctDNA in clinical practice.

Outlook

ctDNA may serve a key role tumor treatment. Despite existing challenges, ongoing research may advance the utility of ctDNA in clinical practice. As improvements in detection sensitivity, standardization of testing protocols and cost reduction are realized, ctDNA may enhance patient care by guiding treatment decisions, monitoring therapeutic response and improving outcomes.

Acknowledgements

Not applicable.

Funding

The present study was supported by Shaanxi Provincial Cancer Hospital, grant no. 2022SF-282, cphcf-2022-218); and 24YXYJ0179).

Availability of data and materials

Not applicable.

Authors' contributions

QG and ZYZ conceived the study. QG, ZYZ, SL, JM and ZZ wrote and reviewed the manuscript. Data authentication is not applicable. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Schoop R, Roode LM, de Boer LL and Dashtbozorg B: Framework for deep learning based Multi-Modality image registration of snapshot and pathology images. IEEE J Biomed Health Inform. Aug 16–2024.(Epub ahead of print). View Article : Google Scholar : PubMed/NCBI

2 

Pritzker K and Nieminen HJ: Needle biopsy adequacy in the era of precision medicine and value-based health care. Arch Pathol Lab Med. 143:1399–1415. 2019. View Article : Google Scholar : PubMed/NCBI

3 

Nikanjam M, Kato S and Kurzrock R: Liquid biopsy: Current technology and clinical applications. J Hematol Oncol. 15:1312022. View Article : Google Scholar : PubMed/NCBI

4 

Tivey A, Church M, Rothwell D, Dive C and Cook N: Circulating tumour DNA-looking beyond the blood. Nat Rev Clin Oncol. 19:600–612. 2022. View Article : Google Scholar : PubMed/NCBI

5 

Alix-Panabieres C and Pantel K: Liquid biopsy: From discovery to clinical application. Cancer Discov. 11:858–873. 2021. View Article : Google Scholar : PubMed/NCBI

6 

Cohen SA, Liu MC and Aleshin A: Practical recommendations for using ctDNA in clinical decision making. Nature. 619:259–268. 2023. View Article : Google Scholar : PubMed/NCBI

7 

Mandel P and Metais P: Nuclear acids in human blood plasma. C R Seances Soc Biol Fil. 142:241–243. 1948.(In French). PubMed/NCBI

8 

Hobbs KJ, Cooper BL, Dembek K and Sheats MK: Investigation of extracted plasma cell-free DNA as a biomarker in foals with sepsis. Vet Sci. 11:3462024.PubMed/NCBI

9 

Zhang LX, Jiang YZ, Qiu LJ and Huang DP: Quantitative detection and integrality analysis of plasma circulating Cell-free DNA in multiple myeloma. Zhongguo Shi Yan Xue Ye Xue Za Zhi. 32:1106–1111. 2024.(In Chinese). PubMed/NCBI

10 

Joshi J, Raval A, Desai U, Upadhyay V, Bhavsar M, Shah K, Rawal R, Panchal H and Shah F: EGFR mutation analysis in Non-small cell lung carcinoma patients: A liquid biopsy approach. Indian J Clin Biochem. 36:51–58. 2021. View Article : Google Scholar : PubMed/NCBI

11 

Volik S, Alcaide M, Morin RD and Collins C: Cell-free DNA (cfDNA): Clinical significance and utility in cancer shaped by emerging technologies. Mol Cancer Res. 14:898–908. 2016. View Article : Google Scholar : PubMed/NCBI

12 

Tan EM, Schur PH, Carr RI and Kunkel HG: Deoxybonucleic acid (DNA) and antibodies to DNA in the serum of patients with systemic lupus erythematosus. J Clin Invest. 45:1732–1740. 1966. View Article : Google Scholar : PubMed/NCBI

13 

Koffler D, Agnello V, Winchester R and Kunkel HG: The occurrence of single-stranded DNA in the serum of patients with systemic lupus erythematosus and other diseases. J Clin Invest. 52:198–204. 1973. View Article : Google Scholar : PubMed/NCBI

14 

Giacona MB, Ruben GC, Iczkowski KA, Roos TB, Porter DM and Sorenson GD: Cell-free DNA in human blood plasma: Length measurements in patients with pancreatic cancer and healthy controls. Pancreas. 17:89–97. 1998. View Article : Google Scholar : PubMed/NCBI

15 

Szilagyi M, Pos O, Marton E, Buglyo G, Soltesz B, Keseru J, Penyige A, Szemes T and Nagy B: Circulating cell-free nucleic acids: Main characteristics and clinical application. Int J Mol Sci. 21:68272020. View Article : Google Scholar : PubMed/NCBI

16 

Biro O, Fothi A, Alasztics B, Nagy B, Orban TI and Rigo JJ: Circulating exosomal and Argonaute-bound microRNAs in preeclampsia. Gene. 692:138–144. 2019. View Article : Google Scholar : PubMed/NCBI

17 

Fernando MR, Jiang C, Krzyzanowski GD and Ryan WL: New evidence that a large proportion of human blood plasma cell-free DNA is localized in exosomes. PLoS One. 12:e01839152017. View Article : Google Scholar : PubMed/NCBI

18 

Aucamp J, Bronkhorst AJ, Badenhorst C and Pretorius PJ: The diverse origins of circulating cell-free DNA in the human body: A critical re-evaluation of the literature. Biol Rev Camb Philos Soc. 93:1649–1683. 2018. View Article : Google Scholar : PubMed/NCBI

19 

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. 9:50682018. View Article : Google Scholar : PubMed/NCBI

20 

Teo YV, Capri M, Morsiani C, Pizza G, Faria A, Franceschi C and Neretti N: Cell-free DNA as a biomarker of aging. Aging Cell. 18:e128902019. View Article : Google Scholar : PubMed/NCBI

21 

Hummel EM, Hessas E, Muller S, Beiter T, Fisch M, Eibl A, Wolf OT, Giebel B, Platen P, Kumsta R and Moser DA: Cell-free DNA release under psychosocial and physical stress conditions. Transl Psychiatry. 8:2362018. View Article : Google Scholar : PubMed/NCBI

22 

Ai B, Liu H, Huang Y and Peng P: Circulating cell-free DNA as a prognostic and predictive biomarker in non-small cell lung cancer. Oncotarget. 7:44583–44595. 2016. View Article : Google Scholar : PubMed/NCBI

23 

Kim DY, Cho EH, Kim JS, Chie EK and Kang HC: Plasma Circulating Cell-free DNa in advanced hepatocellular carcinoma patients treated with radiation therapy. In Vivo. 37:2306–2313. 2023. View Article : Google Scholar : PubMed/NCBI

24 

Gianni C, Palleschi M, Merloni F, Di Menna G, Sirico M, Sarti S, Virga A, Ulivi P, Cecconetto L, Mariotti M and De Giorgi U: Cell-Free DNA Fragmentomics: A promising biomarker for diagnosis, prognosis and prediction of response in breast cancer. Int J Mol Sci. 23:141972022. View Article : Google Scholar : PubMed/NCBI

25 

Al SN, Messaoudi SA, Babu SR, Chaudhary AB, Alsharm AA, Alrefaei AF, Kadasah S, Abu-Elmagd M, Assidi M, Buhmeida A, et al: Utility of circulating Cell-free DNA in assessing microsatellite instability and loss of Heterozygosity in breast cancer using human identification approach. Genes (Basel). 13:5902022. View Article : Google Scholar

26 

Bahado-Singh RO, Turkoglu O, Aydas B and Vishweswaraiah S: Precision oncology: Artificial intelligence, circulating cell-free DNA, and the minimally invasive detection of pancreatic cancer-A pilot study. Cancer Med. 12:19644–19655. 2023. View Article : Google Scholar : PubMed/NCBI

27 

Lin LH, Chang KW, Kao SY, Cheng HW and Liu CJ: Increased plasma circulating Cell-Free DNA could be a potential marker for oral cancer. Int J Mol Sci. 19:33032018. View Article : Google Scholar : PubMed/NCBI

28 

Fang Q, Yuan Z, Hu H, Zhang W, Wang G and Wang X: Genome-wide discovery of circulating cell-free DNA methylation biomarkers for colorectal cancer detection. Clin Epigenetics. 15:1192023. View Article : Google Scholar : PubMed/NCBI

29 

Eskander NS, Mansour L, Abdelaal A, Saad E and Mohamed D: Circulating cell free DNA integrity index as a biomarker for response to chemotherapy in patients with metastatic colorectal carcinoma. Asian Pac J Cancer Prev. 23:339–348. 2022. View Article : Google Scholar : PubMed/NCBI

30 

Heidrich I and Pantel K: Liquid biopsy: Blood-based analyses of circulating cell-free DNA in xenografts. EMBO Mol Med. 14:e163262022. View Article : Google Scholar : PubMed/NCBI

31 

Kroeze A, Cornelissen AS, Pascutti MF, Verheij M, Bulder I, Klarenbeek S, Ait SA, Hazenberg MD, Nur E, van der Schoot CE, et al: Cell-free DNA levels are increased in acute graft-versus-host disease. Eur J Haematol. 109:271–281. 2022. View Article : Google Scholar : PubMed/NCBI

32 

Kustanovich A, Schwartz R, Peretz T and Grinshpun A: Life and death of circulating cell-free DNA. Cancer Biol Ther. 20:1057–1067. 2019. View Article : Google Scholar : PubMed/NCBI

33 

Stewart CM and Tsui D: Circulating cell-free DNA for non-invasive cancer management. Cancer Genet. 228–229. 169–179. 2018.

34 

Yu SC, Lee SW, Jiang P, Leung TY, Chan KC, Chiu RW and Lo YM: High-resolution profiling of fetal DNA clearance from maternal plasma by massively parallel sequencing. Clin Chem. 59:1228–1237. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Butler TM, Spellman PT and Gray J: Circulating-tumor DNA as an early detection and diagnostic tool. Curr Opin Genet Dev. 42:14–21. 2017. View Article : Google Scholar : PubMed/NCBI

36 

Jahr S, Hentze H, Englisch S, Hardt D, Fackelmayer FO, Hesch RD and Knippers R: DNA fragments in the blood plasma of cancer patients: Quantitations and evidence for their origin from apoptotic and necrotic cells. Cancer Res. 61:1659–1665. 2001.PubMed/NCBI

37 

Miller AM and Karajannis MA: Current role and future potential of CSF ctDNA for the diagnosis and clinical management of pediatric central nervous system tumors. J Natl Compr Canc Netw. 20:1363–1369. 2022.PubMed/NCBI

38 

Diehl F, Schmidt K, Choti MA, Romans K, Goodman S, Li M, Thornton K, Agrawal N, Sokoll L, Szabo SA, et al: Circulating mutant DNA to assess tumor dynamics. Nat Med. 14:985–990. 2008. View Article : Google Scholar : PubMed/NCBI

39 

Han JY, Ahn KS, Kim TS, Kim YH, Cho KB, Shin DW, Baek WK, Suh SI, Jang BC and Kang KJ: Liquid biopsy from Bile-Circulating tumor DNA in patients with biliary tract cancer. Cancers (Basel). 13:45812021. View Article : Google Scholar : PubMed/NCBI

40 

Bettegowda C, Sausen M, Leary RJ, Kinde I, Wang Y, Agrawal N, Bartlett BR, Wang H, Luber B, Alani RM, et al: Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci Transl Med. 6:224ra242014. View Article : Google Scholar : PubMed/NCBI

41 

Diehl F, Li M, Dressman D, He Y, Shen D, Szabo S, Diaz LJ, Goodman SN, David KA, Juhl H, et al: Detection and quantification of mutations in the plasma of patients with colorectal tumors. Proc Natl Acad Sci USA. 102:16368–16373. 2005. View Article : Google Scholar : PubMed/NCBI

42 

Dawson SJ, Tsui DW, Murtaza M, Biggs H, Rueda OM, Chin SF, Dunning MJ, Gale D, Forshew T, Mahler-Araujo B, et al: Analysis of circulating tumor DNA to monitor metastatic breast cancer. N Engl J Med. 368:1199–1209. 2013. View Article : Google Scholar : PubMed/NCBI

43 

Sato S, Nakamura Y, Oki E and Yoshino T: Molecular residual Disease-guided adjuvant treatment in resected colorectal cancer: Focus on CIRCULATE-Japan. Clin Colorectal Cancer. 22:53–58. 2023. View Article : Google Scholar : PubMed/NCBI

44 

Yi K, Wang X, Filippov SK and Zhang H: Emerging ctDNA detection strategies in clinical cancer theranostics. Smart Med. 2:e202300312023. View Article : Google Scholar : PubMed/NCBI

45 

Cheng F, Su L and Qian C: Circulating tumor DNA: A promising biomarker in the liquid biopsy of cancer. Oncotarget. 7:48832–48841. 2016. View Article : Google Scholar : PubMed/NCBI

46 

Lee TH, Montalvo L, Chrebtow V and Busch MP: Quantitation of genomic DNA in plasma and serum samples: Higher concentrations of genomic DNA found in serum than in plasma. Transfusion. 41:276–282. 2001. View Article : Google Scholar : PubMed/NCBI

47 

Heger JM, Mattlener J, Schneider J, Godel P, Sieg N, Ullrich F, Lewis RI, Bucaciuc-Mracica T, Schwarz RF, Ruess D, et al: Entirely noninvasive outcome prediction in central nervous system lymphomas using circulating tumor DNA. Blood. 143:522–534. 2023. View Article : Google Scholar

48 

Werner B, Warton K and Ford CE: Transcending Blood-Opportunities for alternate liquid biopsies in oncology. Cancers (Basel). 14:13092022. View Article : Google Scholar : PubMed/NCBI

49 

Seyhan AA: Circulating liquid biopsy biomarkers in glioblastoma: Advances and challenges. Int J Mol Sci. 25:79742024. View Article : Google Scholar : PubMed/NCBI

50 

Zheng MM, Li YS, Jiang BY, Tu HY, Tang WF, Yang JJ, Zhang XC, Ye JY, Yan HH, Su J, et al: Clinical utility of cerebrospinal fluid Cell-Free DNA as liquid biopsy for leptomeningeal metastases in ALK-Rearranged NSCLC. J Thorac Oncol. 14:924–932. 2019. View Article : Google Scholar : PubMed/NCBI

51 

Wu J, Liu Z, Huang T, Wang Y, Song MM, Song T, Long G, Zhang X, Li X and Zhang L: Cerebrospinal fluid circulating tumor DNA depicts profiling of brain metastasis in NSCLC. Mol Oncol. 17:810–824. 2023. View Article : Google Scholar : PubMed/NCBI

52 

De Mattos-Arruda L, Mayor R, Ng C, Weigelt B, Martinez-Ricarte F, Torrejon D, Oliveira M, Arias A, Raventos C, Tang J, et al: Cerebrospinal fluid-derived circulating tumour DNA better represents the genomic alterations of brain tumours than plasma. Nat Commun. 6:88392015. View Article : Google Scholar : PubMed/NCBI

53 

Wang Y, Luo N, Gao Y, Wu Y, Qin X, Qi Y, Sun T, Tao R, Qi C, Liu B and Yuan S: The joint detection of CEA and ctDNA in cerebrospinal fluid: An auxiliary tool for the diagnosis of leptomeningeal metastases in cancer. J Cancer Res Clin Oncol. 149:1679–1690. 2023. View Article : Google Scholar : PubMed/NCBI

54 

Bai Y, Yu Q, Liu N, Liu J, Wang D, Liu X and Yuan S: Case report: Cerebrospinal fluid-derived circulating tumor DNA diagnoses and guides the treatment of a lung adenocarcinoma case with leptomeningeal metastasis. Front Oncol. 12:9449632022. View Article : Google Scholar : PubMed/NCBI

55 

van der Wel J, Boelens MC, Jebbink M, Smulders SA, Maas KW, Luitse M, Compter A, Boltjes R, Sol N, Monkhorst K, et al: Osimertinib-induced DNA resistance mutations in cerebrospinal fluid of EGFR mutated NSCLC patients developing leptomeningeal metastases: ORA-LM study. Neuro Oncol. Aug 7–2024.doi: 10.1093/neuonc/noae138 (Epub ahead of print). View Article : Google Scholar : PubMed/NCBI

56 

Azad TD, Nanjo S, Jin MC, Chabon JJ, Kurtz DM, Chaudhuri AA, Connolly ID, Hui AB, Liu CL, Merriott D, et al: Quantification of cerebrospinal fluid tumor DNA in lung cancer patients with suspected leptomeningeal carcinomatosis. NPJ Precis Oncol. 8:1212024. View Article : Google Scholar : PubMed/NCBI

57 

Valerius AR, Webb MJ, Hammad N, Sener U and Malani R: Cerebrospinal fluid liquid biopsies in the evaluation of adult gliomas. Curr Oncol Rep. 26:377–390. 2024. View Article : Google Scholar : PubMed/NCBI

58 

Dai L, Liu Z, Zhu Y and Ma L: Genome-wide methylation analysis of circulating tumor DNA: A new biomarker for recurrent glioblastom. Heliyon. 9:e143392023. View Article : Google Scholar : PubMed/NCBI

59 

Kojic M, Maybury MK, Waddell N, Koufariotis LT, Addala V, Millar A, Wood S, Pearson JV, Hansford JR, Hassall T, et al: Efficient detection and monitoring of pediatric brain malignancies with liquid biopsy based on patient-specific somatic mutation screening. Neuro Oncol. 25:1507–1517. 2023. View Article : Google Scholar : PubMed/NCBI

60 

Izquierdo E, Proszek P, Pericoli G, Temelso S, Clarke M, Carvalho DM, Mackay A, Marshall LV, Carceller F, Hargrave D, et al: Droplet digital PCR-based detection of circulating tumor DNA from pediatric high grade and diffuse midline glioma patients. Neurooncol Adv. 3:vdab0132021.PubMed/NCBI

61 

Li J, Zhao S, Lee M, Yin Y, Li J, Zhou Y, Ballester LY, Esquenazi Y, Dashwood RH, Davies P, et al: Reliable tumor detection by whole-genome methylation sequencing of cell-free DNA in cerebrospinal fluid of pediatric medulloblastoma. Sci Adv. 6:eabb54272020. View Article : Google Scholar : PubMed/NCBI

62 

Pages M, Rotem D, Gydush G, Reed S, Rhoades J, Ha G, Lo C, Fleharty M, Duran M, Jones R, et al: Liquid biopsy detection of genomic alterations in pediatric brain tumors from cell-free DNA in peripheral blood, CSF, and urine. Neuro Oncol. 24:1352–1363. 2022. View Article : Google Scholar : PubMed/NCBI

63 

Ding S and Song X, Geng X, Liu L, Ma H, Wang X, Wei L, Xie L and Song X: Saliva-derived cfDNA is applicable for EGFR mutation detection but not for quantitation analysis in non-small cell lung cancer. Thorac Cancer. 10:1973–1983. 2019. View Article : Google Scholar : PubMed/NCBI

64 

Wang Z, Zhang L, Li L, Li X, Xu Y, Wang M, Liang L, Jiao P, Li Y, He S, et al: Sputum Cell-Free DNA: Valued surrogate sample for detection of EGFR mutation in patients with advanced lung adenocarcinoma. J Mol Diagn. 22:934–942. 2020. View Article : Google Scholar : PubMed/NCBI

65 

Wang Z, Li X, Zhang L, Xu Y, Wang M, Liang L, Jiao P, Li Y, He S, Du J, et al: Sputum cell-free DNA: Valued surrogate sample for the detection of EGFR exon 20 p.T790M mutation in patients with advanced lung adenocarcinoma and acquired resistance to EGFR-TKIs. Cancer Med. 10:3323–3331. 2021. View Article : Google Scholar : PubMed/NCBI

66 

Ferrier ST, Tsering T, Sadeghi N, Zeitouni A and Burnier JV: Blood and saliva-derived ctDNA is a marker of residual disease after treatment and correlates with recurrence in human papillomavirus-associated head and neck cancer. Cancer Med. 12:15777–15787. 2023. View Article : Google Scholar : PubMed/NCBI

67 

Britze TE, Jakobsen KK, Gronhoj C and von Buchwald C: A systematic review on the role of biomarkers in liquid biopsies and saliva samples in the monitoring of salivary gland cancer. Acta Otolaryngol. 143:709–713. 2023. View Article : Google Scholar : PubMed/NCBI

68 

Gupta S, Singh B, Abhishek R, Gupta S and Sachan M: The emerging role of liquid biopsy in oral squamous cell carcinoma detection: Advantages and challenges. Expert Rev Mol Diagn. 24:311–331. 2024. View Article : Google Scholar : PubMed/NCBI

69 

Perrone ME, Alvarez R, Vo TT, Chung MW, Chhieng DC, Paulson VA, Colbert BG, Q Konnick E and Huang EC: Validating cell-free DNA from supernatant for molecular diagnostics on cytology specimens. Cancer Cytopathol. 129:956–965. 2021. View Article : Google Scholar : PubMed/NCBI

70 

Yang SR, Mooney KL, Libiran P, Jones CD, Joshi R, Lau HD, Stehr H, Berry GJ, Zehnder JL, Long SR, et al: Targeted deep sequencing of cell-free DNA in serous body cavity fluids with malignant, suspicious, and benign cytology. Cancer Cytopathol. 128:43–56. 2020. View Article : Google Scholar : PubMed/NCBI

71 

Leick KM, Kazarian AG, Rajput M, Tomanek-Chalkley A, Miller A, Shrader HR, Mccarthy A, Coleman KL, Kasi PM and Chan C: Peritoneal Cell-free tumor DNA as biomarker for peritoneal surface malignancies. Ann Surg Oncol. 27:5065–5071. 2020. View Article : Google Scholar : PubMed/NCBI

72 

Kinugasa H, Nouso K, Ako S, Dohi C, Matsushita H, Matsumoto K, Kato H and Okada H: Liquid biopsy of bile for the molecular diagnosis of gallbladder cancer. Cancer Biol Ther. 19:934–938. 2018. View Article : Google Scholar : PubMed/NCBI

73 

Takai E, Totoki Y, Nakamura H, Morizane C, Nara S, Hama N, Suzuki M, Furukawa E, Kato M, Hayashi H, et al: Clinical utility of circulating tumor DNA for molecular assessment in pancreatic cancer. Sci Rep. 5:184252015. View Article : Google Scholar : PubMed/NCBI

74 

Levink I, Jansen M, Azmani Z, van Ijcken W, van Marion R, Peppelenbosch MP, Cahen DL, Fuhler GM and Bruno MJ: Mutation analysis of pancreatic juice and plasma for the detection of pancreatic cancer. Int J Mol Sci. 24:131162023. View Article : Google Scholar : PubMed/NCBI

75 

Fitzgerald JM, Ramchurren N, Rieger K, Levesque P, Silverman M, Libertino JA and Summerhayes IC: Identification of H-ras mutations in urine sediments complements cytology in the detection of bladder tumors. J Natl Cancer Inst. 87:129–133. 1995. View Article : Google Scholar : PubMed/NCBI

76 

Jain S, Lin SY, Song W and Su YH: Urine-based liquid biopsy for nonurological cancers. Genet Test Mol Biomarkers. 23:277–283. 2019. View Article : Google Scholar : PubMed/NCBI

77 

Su YH, Wang M, Block TM, Landt O, Botezatu I, Serdyuk O, Lichtenstein A, Melkonyan H, Tomei LD and Umansky S: Transrenal DNA as a diagnostic tool: Important technical notes. Ann N Y Acad Sci. 1022:81–89. 2004. View Article : Google Scholar : PubMed/NCBI

78 

Su YH, Wang M, Brenner DE, Norton PA and Block TM: Detection of mutated K-ras DNA in urine, plasma, and serum of patients with colorectal carcinoma or adenomatous polyps. Ann N Y Acad Sci. 1137:197–206. 2008. View Article : Google Scholar : PubMed/NCBI

79 

Xiao Y, Ju L, Qian K, Jin W, Wang G, Zhao Y, Jiang W, Liu N, Wu K, Peng M, et al: Non-invasive diagnosis and surveillance of bladder cancer with driver and passenger DNA methylation in a prospective cohort study. Clin Transl Med. 12:e10082022. View Article : Google Scholar : PubMed/NCBI

80 

Christensen E, Nordentoft I, Birkenkamp-Demtroder K, Elbaek SK, Lindskrog SV, Taber A, Andreasen TG, Strandgaard T, Knudsen M, Lamy P, et al: Cell-Free urine and plasma DNA mutational analysis predicts neoadjuvant chemotherapy response and outcome in patients with muscle-invasive bladder cancer. Clin Cancer Res. 29:1582–1591. 2023. View Article : Google Scholar : PubMed/NCBI

81 

Tamura D, Abe M, Hiraki H, Sasaki N, Yashima-Abo A, Ikarashi D, Kato R, Kato Y, Maekawa S, Kanehira M, et al: Postoperative recurrence detection using individualized circulating tumor DNA in upper tract urothelial carcinoma. Cancer Sci. 115:529–539. 2023. View Article : Google Scholar : PubMed/NCBI

82 

Kim AK, Hamilton JP, Lin SY, Chang TT, Hann HW, Hu CT, Lou Y, Lin YJ, Gade TP, Park G, et al: Urine DNA biomarkers for hepatocellular carcinoma screening. Br J Cancer. 126:1432–1438. 2022. View Article : Google Scholar : PubMed/NCBI

83 

Adrogue HJ and Madias NE: Assessing Acid-base status: Physiologic versus physicochemical approach. Am J Kidney Dis. 68:793–802. 2016. View Article : Google Scholar : PubMed/NCBI

84 

Dermody SM, Bhambhani C, Swiecicki PL, Brenner JC and Tewari M: Trans-renal cell-free tumor DNA for Urine-based liquid biopsy of cancer. Front Genet. 13:8791082022. View Article : Google Scholar : PubMed/NCBI

85 

Alahdal M, Perera RA, Moschovas MC, Patel V and Perera RJ: Current advances of liquid biopsies in prostate cancer: Molecular biomarkers. Mol Ther Oncolytics. 30:27–38. 2023. View Article : Google Scholar : PubMed/NCBI

86 

Fonseca NM, Maurice-Dror C, Herberts C, Tu W, Fan W, Murtha AJ, Kollmannsberger C, Kwan EM, Parekh K, Schonlau E, et al: Prediction of plasma ctDNA fraction and prognostic implications of liquid biopsy in advanced prostate cancer. Nat Commun. 15:18282024. View Article : Google Scholar : PubMed/NCBI

87 

Tolmeijer SH, Boerrigter E, Van Erp NP and Mehra N: Using early on-treatment circulating tumor DNA measurements as response assessment in metastatic castration resistant prostate cancer. Oncotarget. 15:421–423. 2024. View Article : Google Scholar : PubMed/NCBI

88 

Ponti G, Maccaferri M, Manfredini M, Micali S, Torricelli F, Milandri R, Del PC, Ciarrocchi A, Ruini C, Benassi L, et al: Quick assessment of cell-free DNA in seminal fluid and fragment size for early non-invasive prostate cancer diagnosis. Clin Chim Acta. 497:76–80. 2019. View Article : Google Scholar : PubMed/NCBI

89 

Ponti G, Maccaferri M, Percesepe A, Tomasi A and Ozben T: Liquid biopsy with cell free DNA: New horizons for prostate cancer. Crit Rev Clin Lab Sci. 58:60–76. 2021. View Article : Google Scholar : PubMed/NCBI

90 

Yu B and Ma W: Biomarker discovery in hepatocellular carcinoma (HCC) for personalized treatment and enhanced prognosis. Cytokine Growth Factor Rev. Aug 24–2024.doi: 10.1016/j.cytogfr.2024.08.006 (Epub ahead of print). View Article : Google Scholar : PubMed/NCBI

91 

Zhu L, Xu R, Yang L, Shi W, Zhang Y, Liu J, Li X, Zhou J and Bing P: Minimal residual disease (MRD) detection in solid tumors using circulating tumor DNA: A systematic review. Front Genet. 14:11721082023. View Article : Google Scholar : PubMed/NCBI

92 

Li S, Li H, Li X, Zhu M, Li H and Xia F: Hybridization Chain Reaction-amplified electrochemical DNA-based sensors enable calibration-free measurements of nucleic acids directly in whole blood. Anal Chem. 93:8354–8361. 2021. View Article : Google Scholar : PubMed/NCBI

93 

Ho HY, Chung KK, Kan CM and Wong SC: Liquid biopsy in the clinical management of cancers. Int J Mol Sci. 25:85942024. View Article : Google Scholar : PubMed/NCBI

94 

Phallen J, Sausen M, Adleff V, Leal A, Hruban C, White J, Anagnostou V, Fiksel J, Cristiano S, Papp E, et al: Direct detection of early-stage cancers using circulating tumor DNA. Sci Transl Med. 9:eaan24152017. View Article : Google Scholar : PubMed/NCBI

95 

Bittla P, Kaur S, Sojitra V, Zahra A, Hutchinson J, Folawemi O and Khan S: Exploring Circulating tumor DNA (CtDNA) and its role in early detection of cancer: A systematic review. Cureus. 15:e457842023.PubMed/NCBI

96 

Yu W, Hurley J, Roberts D, Chakrabortty SK, Enderle D, Noerholm M, Breakefield XO and Skog JK: Exosome-based liquid biopsies in cancer: Opportunities and challenges. Ann Oncol. 32:466–477. 2021. View Article : Google Scholar : PubMed/NCBI

97 

Kemper M, Krekeler C, Menck K, Lenz G, Evers G, Schulze AB and Bleckmann A: Liquid Biopsies in Lung Cancer. Cancers (Basel). 15:14302023. View Article : Google Scholar : PubMed/NCBI

98 

Lin C, Liu X, Zheng B, Ke R and Tzeng CM: Liquid biopsy, ctDNA diagnosis through NGS. Life (Basel). 11:8902021.PubMed/NCBI

99 

Fernandes M, Cruz-Martins N, Souto MC, Guimaraes S, Pereira RJ, Justino A, Pina MJ, Magalhaes A, Queiroga H, Machado JC, et al: Clinical application of Next-generation sequencing of plasma Cell-free DNA for genotyping untreated advanced Non-small cell lung cancer. Cancers (Basel). 13:27072021. View Article : Google Scholar : PubMed/NCBI

100 

Roberto TM, Jorge MA, Francisco GV, Noelia T, Pilar RG and Andres C: Strategies for improving detection of circulating tumor DNA using next generation sequencing. Cancer Treat Rev. 119:1025952023. View Article : Google Scholar : PubMed/NCBI

101 

Grada A and Weinbrecht K: Next-generation sequencing: Methodology and application. J Invest Dermatol. 133:e112013. View Article : Google Scholar : PubMed/NCBI

102 

Cheng ML, Pectasides E, Hanna GJ, Parsons HA, Choudhury AD and Oxnard GR: Circulating tumor DNA in advanced solid tumors: Clinical relevance and future directions. CA Cancer J Clin. 71:176–190. 2021. View Article : Google Scholar : PubMed/NCBI

103 

Ma M, Zhu H, Zhang C, Sun X, Gao X and Chen G: ‘Liquid biopsy’-ctDNA detection with great potential and challenges. Ann Transl Med. 3:2352015.PubMed/NCBI

104 

Gale D, Lawson A, Howarth K, Madi M, Durham B, Smalley S, Calaway J, Blais S, Jones G, Clark J, et al: Development of a highly sensitive liquid biopsy platform to detect clinically-relevant cancer mutations at low allele fractions in cell-free DNA. PLoS One. 13:e01946302018. View Article : Google Scholar : PubMed/NCBI

105 

Forshew T, Murtaza M, Parkinson C, Gale D, Tsui DW, Kaper F, Dawson SJ, Piskorz AM, Jimenez-Linan M, Bentley D, et al: Noninvasive identification and monitoring of cancer mutations by targeted deep sequencing of plasma DNA. Sci Transl Med. 4:136ra682012. View Article : Google Scholar : PubMed/NCBI

106 

Cabalag CS, Yates M, Corrales MB, Yeh P, Wong SQ, Zhang BZ, Fujihara KM, Chong L, Hii MW, Dawson SJ, et al: Potential clinical utility of a targeted circulating tumor DNA Assay in esophageal adenocarcinoma. Ann Surg. 276:e120–e126. 2022. View Article : Google Scholar : PubMed/NCBI

107 

Newman AM, Bratman SV, To J, Wynne JF, Eclov NC, Modlin LA, Liu CL, Neal JW, Wakelee HA, Merritt RE, et al: An ultrasensitive method for quantitating circulating tumor DNA with broad patient coverage. Nat Med. 20:548–554. 2014. View Article : Google Scholar : PubMed/NCBI

108 

Azad TD, Chaudhuri AA, Fang P, Qiao Y, Esfahani MS, Chabon JJ, Hamilton EG, Yang YD, Lovejoy A, Newman AM, et al: Circulating tumor DNA analysis for detection of minimal residual disease after chemoradiotherapy for localized esophageal cancer. Gastroenterology. 158:494–505. 2020. View Article : Google Scholar : PubMed/NCBI

109 

Noguchi T, Sakai K, Iwahashi N, Matsuda K, Matsukawa H, Yahata T, Toujima S, Nishio K and Ino K: Changes in the gene mutation profiles of circulating tumor DNA detected using CAPP-Seq in neoadjuvant chemotherapy-treated advanced ovarian cancer. Oncol Lett. 19:2713–2720. 2020.PubMed/NCBI

110 

Jung D, Jain P, Yao Y and Wang M: Advances in the assessment of minimal residual disease in mantle cell lymphoma. J Hematol Oncol. 13:1272020. View Article : Google Scholar : PubMed/NCBI

111 

Satyal U, Srivastava A and Abbosh PH: Urine biopsy-liquid gold for molecular detection and surveillance of bladder cancer. Front Oncol. 9:12662019. View Article : Google Scholar : PubMed/NCBI

112 

Taylor K, Zou J, Magalhaes M, Oliva M, Spreafico A, Hansen AR, Mcdade SS, Coyle VM, Lawler M, Elimova E, et al: Circulating tumour DNA kinetics in recurrent/metastatic head and neck squamous cell cancer patients. Eur J Cancer. 188:29–38. 2023. View Article : Google Scholar : PubMed/NCBI

113 

Aoude LG, Brosda S, Ng J, Lonie JM, Belle CJ, Patel K, Koufariotis LT, Wood S, Atkinson V, Smithers BM, et al: Circulating tumor DNA: A promising biomarker for predicting recurrence in patients with BRAF-Negative melanoma. J Mol Diagn. 25:771–781. 2023. View Article : Google Scholar : PubMed/NCBI

114 

Grassi T, Harris FR, Smadbeck JB, Murphy SJ, Block MS, Multinu F, Schaefer KJ, Zhang P, Karagouga G, Liu MC, et al: Personalized tumor-specific DNA junctions to detect circulating tumor in patients with endometrial cancer. PLoS One. 16:e02523902021. View Article : Google Scholar : PubMed/NCBI

115 

Mansson CT, Vad-Nielsen J, Meldgaard P, Nielsen AL and Sorensen BS: EGFR transcription in non-small-cell lung cancer tumours can be revealed in ctDNA by cell-free chromatin immunoprecipitation (cfChIP). Mol Oncol. 15:2868–2876. 2021. View Article : Google Scholar : PubMed/NCBI

116 

Jafri H, Mushtaq S, Baig S, Bhatty A and Siraj S: Comparison of KRAS gene in circulating tumor DNA levels vs histological grading of colorectal cancer patients through liquid biopsy. Saudi J Gastroenterol. 29:371–375. 2023. View Article : Google Scholar : PubMed/NCBI

117 

FDA, . Summary of Safety and Effectiveness Data (SSED) P150047. Cobas EGFR Mutation Test v2®. 2016.

118 

Biglari N, Soltani-Zangbar MS, Mohammadian J, Mehdizadeh A and Abbasi K: ctDNA as a novel and promising approach for cancer diagnosis: A focus on hepatocellular carcinoma. EXCLI J. 22:752–780. 2023.PubMed/NCBI

119 

Hickman RA, Miller AM and Arcila ME: Cerebrospinal fluid: A unique source of circulating tumor DNA with broad clinical applications. Transl Oncol. 33:1016882023. View Article : Google Scholar : PubMed/NCBI

120 

Rimelen V, Ahle G, Pencreach E, Zinniger N, Debliquis A, Zalmai L, Harzallah I, Hurstel R, Alamome I, Lamy F, et al: Tumor cell-free DNA detection in CSF for primary CNS lymphoma diagnosis. Acta Neuropathol Commun. 7:432019. View Article : Google Scholar : PubMed/NCBI

121 

Venetis K, Pepe F, Pescia C, Cursano G, Criscitiello C, Frascarelli C, Mane E, Russo G, Taurelli SB, Troncone G, et al: ESR1 mutations in HR+/HER2-metastatic breast cancer: Enhancing the accuracy of ctDNA testing. Cancer Treat Rev. 121:1026422023. View Article : Google Scholar : PubMed/NCBI

122 

Teh SY, Lin R, Hung LH and Lee AP: Droplet microfluidics. Lab Chip. 8:198–220. 2008. View Article : Google Scholar : PubMed/NCBI

123 

Taniguchi K, Uchida J, Nishino K, Kumagai T, Okuyama T, Okami J, Higashiyama M, Kodama K, Imamura F and Kato K: Quantitative detection of EGFR mutations in circulating tumor DNA derived from lung adenocarcinomas. Clin Cancer Res. 17:7808–7815. 2011. View Article : Google Scholar : PubMed/NCBI

124 

Fang J, Yuan C, Luo X, He Z and Fu W: A Thermus thermophilus argonaute-coupling exponential amplification assay for ultrarapid analysis of circulating tumor DNA. Talanta. 266:1250342024. View Article : Google Scholar : PubMed/NCBI

125 

Cappello F, Angerilli V, Munari G, Ceccon C, Sabbadin M, Pagni F, Fusco N, Malapelle U and Fassan M: FFPE-Based NGS approaches into clinical practice: The limits of glory from a pathologist viewpoint. J Pers Med. 12:1026422022. View Article : Google Scholar

126 

Mantilla WA, Sanabria-Salas MC, Baldion AM, Sua LF, Gonzalez DM and Lema M: NGS in lung, breast, and unknown primary cancer in colombia: A multidisciplinary consensus on challenges and opportunities. JCO Glob Oncol. 7:1012–1023. 2021. View Article : Google Scholar : PubMed/NCBI

127 

Lin YH, Liao XJ, Chang W and Chiou CC: Ultrafast DNA amplification using microchannel Flow-through PCR device. Biosensors (Basel). 12:3032022. View Article : Google Scholar : PubMed/NCBI

128 

Xu J, Han X, Xu W, Liu J, Zhu A, Song D and Long F: Development of a hybridization chain reaction-powered lab-on-fiber device for ultrafast point-of-care testing of circulating tuor DNA in whole blood. Talanta. 259:1244752023. View Article : Google Scholar : PubMed/NCBI

129 

Wu J, Lv J, Zheng X and Wu ZS: Hybridization chain reaction and its applications in biosensing. Talanta. 234:1226372021. View Article : Google Scholar : PubMed/NCBI

130 

Kim J, Shim JS, Han BH, Kim HJ, Park J, Cho IJ, Kang SG, Kang JY, Bong KW and Choi N: Hydrogel-based hybridization chain reaction (HCR) for detection of urinary exosomal miRNAs as a diagnostic tool of prostate cancer. Biosens Bioelectron. 192:1135042021. View Article : Google Scholar : PubMed/NCBI

131 

Papakonstantinou A, Gonzalez NS, Pimentel I, Sunol A, Zamora E, Ortiz C, Espinosa-Bravo M, Peg V, Vivancos A, Saura C, et al: Prognostic value of ctDNA detection in patients with early breast cancer undergoing neoadjuvant therapy: A systematic review and meta-analysis. Cancer Treat Rev. 104:1023622022. View Article : Google Scholar : PubMed/NCBI

132 

Wang D, Zhao P, Lu T, Ren J, Zhu L, Han X, Zhang G, Dong X, Ma H, Yu M and Cai H: ctDNA as a prognostic biomarker in resectable CLM: Systematic review and meta-analysis. Open Life Sci. 18:202206152023. View Article : Google Scholar : PubMed/NCBI

133 

Wei J, Feng J, Weng Y, Xu Z, Jin Y, Wang P, Cui X, Ruan P, Luo R, Li N and Peng M: The prognostic value of ctDNA and bTMB on immune checkpoint inhibitors in human cancer. Front Oncol. 11:7069102021. View Article : Google Scholar : PubMed/NCBI

134 

Markou A, Tzanikou E and Lianidou E: The potential of liquid biopsy in the management of cancer patients. Semin Cancer Biol. 84:69–79. 2022. View Article : Google Scholar : PubMed/NCBI

135 

Bratman SV, Yang S, Iafolla M, Liu Z, Hansen AR, Bedard PL, Lheureux S, Spreafico A, Razak AA, Shchegrova S, et al: Personalized circulating tumor DNA analysis as a predictive biomarker in solid tumor patients treated with pembrolizumab. Nat Cancer. 1:873–881. 2020. View Article : Google Scholar : PubMed/NCBI

136 

Gale D, Heider K, Ruiz-Valdepenas A, Hackinger S, Perry M, Marsico G, Rundell V, Wulff J, Sharma G, Knock H, et al: Residual ctDNA after treatment predicts early relapse in patients with early-stage non-small cell lung cancer. Ann Oncol. 33:500–510. 2022. View Article : Google Scholar : PubMed/NCBI

137 

Borcoman E, Kanjanapan Y, Champiat S, Kato S, Servois V, Kurzrock R, Goel S, Bedard P and Le Tourneau C: Novel patterns of response under immunotherapy. Ann Oncol. 30:385–396. 2019. View Article : Google Scholar : PubMed/NCBI

138 

Young JS, Al-Adli N, Scotford K, Cha S and Berger MS: Pseudoprogression versus true progression in glioblastoma: What neurosurgeons need to know. J Neurosurg. 139:748–759. 2023. View Article : Google Scholar : PubMed/NCBI

139 

Zheng J, Zhou X, Fu Y and Chen Q: Advances in the study of hyperprogression of different tumors treated with PD-1/PD-L1 antibody and the mechanisms of its occurrence. Cancers (Basel). 15:13142023. View Article : Google Scholar : PubMed/NCBI

140 

Vellanki PJ, Ghosh S, Pathak A, Fusco MJ, Bloomquist EW, Tang S, Singh H, Philip R, Pazdur R and Beaver JA: Regulatory implications of ctDNA in Immuno-oncology for solid tumors. J Immunother Cancer. 11:e0053442023. View Article : Google Scholar : PubMed/NCBI

141 

Mahuron KM and Fong Y: Applications of liquid biopsy for surgical patients with cancer: A review. JAMA Surg. 159:96–103. 2024. View Article : Google Scholar : PubMed/NCBI

142 

Juarez-Avendano G, Mendez-Ramirez N, Luna-Silva NC, Gomez-Almaguer D, Pelayo R and Balandran JC: Molecular and cellular markers for measurable residual disease in acute lymphoblastic leukemia. Bol Med Hosp Infant Mex. 78:159–170. 2021.PubMed/NCBI

143 

Li Y, Solis-Ruiz J, Yang F, Long N, Tong CH, Lacbawan FL, Racke FK and Press RD: NGS-defined measurable residual disease (MRD) after initial chemotherapy as a prognostic biomarker for acute myeloid leukemia. Blood Cancer J. 13:592023. View Article : Google Scholar : PubMed/NCBI

144 

Gutman JA, Winters A, Kent A, Amaya M, Mcmahon C, Smith C, Jordan CT, Stevens B, Minhajuddin M, Pei S, et al: Higher-dose venetoclax with measurable residual disease-guided azacitidine discontinuation in newly diagnosed acute myeloid leukemia. Haematologica. 108:2616–2625. 2023. View Article : Google Scholar : PubMed/NCBI

145 

Munir T, Cairns DA, Bloor A, Allsup D, Cwynarski K, Pettitt A, Paneesha S, Fox CP, Eyre TA, Forconi F, et al: Chronic lymphocytic leukemia therapy guided by measurable residual disease. N Engl J Med. 390:326–337. 2024. View Article : Google Scholar : PubMed/NCBI

146 

Zhang JT, Liu SY, Gao W, Liu SM, Yan HH, Ji L, Chen Y, Gong Y, Lu HL, Lin JT, et al: Longitudinal undetectable molecular residual disease defines potentially cured population in localized non-small cell lung cancer. Cancer Discov. 12:1690–1701. 2022. View Article : Google Scholar : PubMed/NCBI

147 

Jung HA, Ku BM, Kim YJ, Park S, Sun JM, Lee SH, Ahn JS, Cho JH, Kim HK, Choi YS, et al: Longitudinal monitoring of circulating tumor DNA from plasma in patients with curative resected Stages I to IIIA EGFR-Mutant Non-Small cell lung cancer. J Thorac Oncol. 18:1199–1208. 2023. View Article : Google Scholar : PubMed/NCBI

148 

Costa LJ, Chhabra S, Medvedova E, Dholaria BR, Schmidt TM, Godby KN, Silbermann R, Dhakal B, Bal S, Giri S, et al: Daratumumab, Carfilzomib, Lenalidomide, and dexamethasone with minimal residual disease Response-Adapted therapy in newly diagnosed multiple myeloma. J Clin Oncol. 40:2901–2912. 2022. View Article : Google Scholar : PubMed/NCBI

149 

San-Miguel J, Avet-Loiseau H, Paiva B, Kumar S, Dimopoulos MA, Facon T, Mateos MV, Touzeau C, Jakubowiak A, Usmani SZ, et al: Sustained minimal residual disease negativity in newly diagnosed multiple myeloma and the impact of daratumumab in MAIA and ALCYONE. Blood. 139:492–501. 2022. View Article : Google Scholar : PubMed/NCBI

150 

Costa LJ, Chhabra S, Medvedova E, Dholaria BR, Schmidt TM, Godby KN, Silbermann R, Dhakal B, Bal S, Giri S, et al: Minimal residual disease response-adapted therapy in newly diagnosed multiple myeloma (MASTER): Final report of the multicentre, single-arm, phase 2 trial. Lancet Haematol. 10:e890–e901. 2023. View Article : Google Scholar : PubMed/NCBI

151 

D'Agostino M, Bertuglia G, Rota-Scalabrini D, Belotti A, More S, Corradini P, Oliva S, Ledda A, Grasso M, Pavone V, et al: Predictors of unsustained minimal residual disease negativity in multiple myeloma (MM) Patients. Blood. 1432023.doi:10.1182/blood.2023022080.

152 

Medford AJ, Moy B, Spring LM, Hurvitz SA, Turner NC and Bardia A: Molecular residual disease in breast cancer: Detection and therapeutic interception. Clin Cancer Res. 29:4540–4548. 2023. View Article : Google Scholar : PubMed/NCBI

153 

Patel RP, Somasundram PM, Smith LK, Sheppard KE and Mcarthur GA: The therapeutic potential of targeting minimal residual disease in melanoma. Clin Transl Med. 13:e11972023. View Article : Google Scholar : PubMed/NCBI

154 

Honore N, van Marcke C, Galot R, Helaers R, Ambroise J, van Maanen A, Mendola A, Dahou H, Marbaix E, Van Eeckhout P, et al: Tumor-agnostic plasma assay for circulating tumor DNA detects minimal residual disease and predicts outcome in locally advanced squamous cell carcinoma of the head and neck. Ann Oncol. 34:1175–1186. 2023. View Article : Google Scholar : PubMed/NCBI

155 

Pott C, Jurinovic V, Trotman J, Kehden B, Unterhalt M, Herold M, Jagt RV, Janssens A, Kneba M, Mayer J, et al: Minimal residual disease status predicts outcome in patients with previously untreated follicular lymphoma: A prospective analysis of the Phase III GALLIUM study. J Clin Oncol. 42:550–561. 2024. View Article : Google Scholar : PubMed/NCBI

156 

Yang K, Hu H, Wu J, Wang H, Guo Z, Yu W, Yao L, Ding F, Zhou T, Wang W, et al: Letter to the Editor: Clinical utility of urine DNA for noninvasive detection and minimal residual disease monitoring in urothelial carcinoma. Mol Cancer. 22:252023. View Article : Google Scholar : PubMed/NCBI

157 

Mo S, Ye L, Wang D, Han L, Zhou S, Wang H, Dai W, Wang Y, Luo W, Wang R, et al: Early detection of molecular residual disease and risk stratification for stage I to III colorectal cancer via circulating tumor DNA Methylation. JAMA Oncol. 9:770–778. 2023. View Article : Google Scholar : PubMed/NCBI

158 

Slater S, Bryant A, Chen HC, Begum R, Rana I, Aresu M, Peckitt C, Zhitkov O, Lazaro-Alcausi R, Borja V, et al: ctDNA guided adjuvant chemotherapy versus standard of care adjuvant chemotherapy after curative surgery in patients with high risk stage II or stage III colorectal cancer: A multi-centre, prospective, randomised control trial (TRACC Part C). BMC Cancer. 23:2572023. View Article : Google Scholar : PubMed/NCBI

159 

Armakolas A, Kotsari M and Koskinas J: Liquid biopsies, novel approaches and future directions. Cancers (Basel). 15:15792023. View Article : Google Scholar : PubMed/NCBI

160 

Xie J, Yao W, Chen L, Zhu W, Liu Q, Geng G, Fang J, Zhao Y, Xiao L, Huang Z and Zhao J: Plasma ctDNA increases tissue NGS-based detection of therapeutically targetable mutations in lung cancers. BMC Cancer. 23:2942023. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2024
Volume 28 Issue 5

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ge Q, Zhang Z, Li S, Ma J and Zhao Z: Liquid biopsy: Comprehensive overview of circulating tumor DNA (Review). Oncol Lett 28: 548, 2024.
APA
Ge, Q., Zhang, Z., Li, S., Ma, J., & Zhao, Z. (2024). Liquid biopsy: Comprehensive overview of circulating tumor DNA (Review). Oncology Letters, 28, 548. https://doi.org/10.3892/ol.2024.14681
MLA
Ge, Q., Zhang, Z., Li, S., Ma, J., Zhao, Z."Liquid biopsy: Comprehensive overview of circulating tumor DNA (Review)". Oncology Letters 28.5 (2024): 548.
Chicago
Ge, Q., Zhang, Z., Li, S., Ma, J., Zhao, Z."Liquid biopsy: Comprehensive overview of circulating tumor DNA (Review)". Oncology Letters 28, no. 5 (2024): 548. https://doi.org/10.3892/ol.2024.14681