Next Article in Journal
EWS-FLI1 and Activator Protein-1 (AP-1) Reciprocally Regulate Extracellular-Matrix Proteins in Ewing sarcoma Cells
Previous Article in Journal
Development of Antimicrobial Surfaces Using Diamond-like Carbon or Diamond-like Carbon-Based Coatings
Previous Article in Special Issue
Effects of Atorvastatin and Simvastatin on the Bioenergetic Function of Isolated Rat Brain Mitochondria
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Liquid Biopsy in the Clinical Management of Cancers

Department of Applied Biology & Chemical Technology, The Hong Kong Polytechnic University, Hong Kong SAR, China
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2024, 25(16), 8594; https://doi.org/10.3390/ijms25168594
Submission received: 14 June 2024 / Revised: 2 August 2024 / Accepted: 2 August 2024 / Published: 6 August 2024

Abstract

:
Liquid biopsy, a noninvasive diagnosis that examines circulating tumor components in body fluids, is increasingly used in cancer management. An overview of relevant literature emphasizes the current state of liquid biopsy applications in cancer care. Biomarkers in liquid biopsy, particularly circulating tumor DNA (ctDNA), circulating tumor RNAs (ctRNA), circulating tumor cells (CTCs), extracellular vesicles (EVs), and other components, offer promising opportunities for early cancer diagnosis, treatment selection, monitoring, and disease assessment. The implementation of liquid biopsy in precision medicine has shown significant potential in various cancer types, including lung cancer, colorectal cancer, breast cancer, and prostate cancer. Advances in genomic and molecular technologies such as next-generation sequencing (NGS) and digital polymerase chain reaction (dPCR) have expanded the utility of liquid biopsy, enabling the detection of somatic variants and actionable genomic alterations in tumors. Liquid biopsy has also demonstrated utility in predicting treatment responses, monitoring minimal residual disease (MRD), and assessing tumor heterogeneity. Nevertheless, standardizing liquid biopsy techniques, interpreting results, and integrating them into the clinical routine remain as challenges. Despite these challenges, liquid biopsy has significant clinical implications in cancer management, offering a dynamic and noninvasive approach to understanding tumor biology and guiding personalized treatment strategies.

1. Introduction

Cancer is the leading cause of death in the world, accounting for nearly 20 million new cancer cases and 9.7 million deaths in 2022. Cancer is also referred to as a silent killer since its symptoms are vague and thus difficult to detect early. Cancer formation consists of the following four phases: initiation, promotion, progression, and metastasis. The first phase, initiation, involves gene variants in a cell. Promotion is the phase between a premalignant lesion and the development of invasive cancer, involving the accumulation of actively proliferating preneoplastic cells. The next phase is progression where genetic and phenotypic changes and cell proliferation occur. At this phase, the tumor size increases rapidly and the cells may undergo further variations with invasive and metastatic potential [1]. The final stage is metastasis, which refers to the spread of cancer cells from a primary tumor to distant organs of the body. The metastasis process is a multistep process, named “metastatic cascade”, broadly divided into five distinct steps (Figure 1). The malignant cells invade into adjacent tissues and penetrate into lymphatic and circulatory systems. These malignant cells are now known as “CTCs”. These cells then exit from the lymphatic and circulatory systems into adjacent normal tissue or organs, which then survive and proliferate in adjacent tissue or organs, leading to colonization [2].
Liquid biopsy has emerged as a revolutionary approach in the field of oncology, offering a minimally invasive and real-time method for the detection, monitoring, and characterization of cancer. Unlike traditional tissue biopsies, which are invasive and may not always capture the heterogeneity of tumors, liquid biopsy involves the analysis of biomarkers in biofluids such as blood, urine, saliva, sputum, stool, ascites, pleural effusion, seminal plasma, or cerebrospinal fluid [3]. This approach holds great potential for transforming cancer management through the provision of clinicians with valuable insight into tumor dynamics, treatment response, and disease progression. In recent years, the field of liquid biopsy has made rapid advancements driven through technological innovations and a deeper understanding of cancer biology. The ability to detect CTCs, ctDNA, ctRNA, EVs, and metabolites in liquid biopsy samples has opened new avenues for personalized medicine and precision oncology [2] (Figure 2). With the aid of liquid biopsy, clinicians may be able to formulate tailor-made treatment strategies for individual cancers based on their individual molecular profiles, resulting in more effective and targeted therapies.
This review aims to explore the role of liquid biopsy in the clinical management of cancer, highlighting its potential applications, challenges, and future directions. By consolidating the current knowledge and research findings in this rapidly developing field, we seek to provide a comprehensive overview of the impact of liquid biopsy on cancer diagnosis, prognosis, treatment selection, and monitoring.

2. Background

Traditional methods of cancer diagnosis and monitoring have primarily relied on tissue biopsies, which involve the surgical removal of a sample of the tumor for analysis [4]. While tissue biopsies remain the gold standard for cancer diagnosis, they are limited by several factors, including their invasive nature, the possibility of tumor seeding [5], sampling errors, and a small risk of morbidity associated with tissue biopsy procedures [6]. These limitations underscore the need for noninvasive and dynamic approaches that can provide a more comprehensive picture of the disease. In addition, tissue biopsies typically obtain a sample of only a part of the tumor, thus only encompassing a part of tumor heterogeneity, limiting the information obtained on the levels of genetic and epigenetic variability of a patient’s cancer [7]. Liquid biopsy offers advantages over traditional tissue biopsies, providing a minimally invasive, dynamic, and comprehensive molecular analysis of tumors, aiding in early diagnosis, prognostication, and treatment response monitoring (Table 1). By analyzing biomolecules shed by tumors into the bloodstream, liquid biopsy provides valuable information about the genetic alterations, variational profiles, and treatment response of cancer cells. Moreover, liquid biopsy can be performed after surgical resection and when there is no detectable metastatic mass for cancer treatment monitoring. Due to its minimally invasive nature, liquid biopsy can be performed multiple times or sequentially, which allows for continuous monitoring of disease progression as well as the emergence of treatment-resistant clones, offering insights that can be used to guide clinical decision-making. With the less invasive obtaining method and the various analytes that can be sampled in liquid biopsy, it can be used as a routine method for the real-time monitoring of cancer progression, early diagnosis of cancer, and assessing treatment response [8]. Liquid biopsy may help reveal intra-tumor heterogeneity (within tumors) and inter-tumor heterogeneity (between tumors), allowing the differences between the primary tumor and metastases, and the differences exposed during disease progression to be distinguished [9]. For instance, Jin et al. successfully identified three potential prognostic ctRNA biomarkers, which may be possible to apply MRD testing involving the identification of any remnants of cancer cells after operation [10]. Through the integration of liquid biopsy into clinical practice, cancer management will be revolutionized by early detection, personalized treatment selection, and real-time monitoring of treatment outcomes. Researchers are exploring liquid biopsy’s utility in diverse cancer types and clinical settings, where the field has great potential to improve patient outcomes and expand our understanding of cancer biology.
Although liquid biopsy holds several advantages over tissue biopsy, more studies and advanced technologies are required to be conducted and invented in order for liquid biopsy to have the potential to replace tissue biopsy. The analytes in liquid biopsy are present in a very low concentration, thus the assays for the detection of analytes should be sensitive and specific enough in order to produce a valid result for the detection and monitoring of cancer. Moreover, tumor histology specification and staging cannot be determined by liquid biopsy, stating that liquid biopsy should be conducted with tissue biopsy for the treatment and diagnosis of cancer. Studies have shown that validation is still needed to increase the potential of liquid biopsy [10,11]. Therefore, implementing liquid biopsy assays in clinical practice is indeed facing significant challenges despite the promising potential of this technology.

3. Technology to Collect and Detect Liquid Biopsy

In liquid biopsy, biomarkers are identified in biofluids in order to provide valuable insights into the presence and characteristics of cancer. The main components of liquid biopsy include CTCs, ctDNA, ctRNA, EVs (exosomes), and proteins as shown in Figure 3. These biomarkers shed by tumors into the bloodstream or other body fluids can offer valuable insights into the genetic changes and heterogeneity of cancer cells. Among different types of body fluids, blood is considered the most common material to analyze, but other body fluids may hold some benefits over blood. For instance, the malignant pleural and peritoneal effusions may exhibit a higher concentration of CTCs than blood, or collecting CTCs in urine can enable the diagnosis of bladder cancer without the need for endoscopy [12].

3.1. Circulating Tumor Cells (CTCs)

Cells that shed from a tumor and enter the circulatory or lymphatic system are referred to as CTCs. They are able to travel through the bloodstream or lymphatic system to other areas of the body, which has the potential to cause distant metastases. Different molecular markers are present on different CTCs depending on the type of cancer. As most of the cancers are of an epithelial origin, a “universal” epithelial marker of cancer, an epithelial cell adhesion molecule (EpCAM) can be used as a common marker for CTCs [13]. The expression of an EpCAM varies in different cancers with breast and prostate cancer being the highest and neurogenic cancers being the lowest. Cancer-specific CTC marks such as epidermal growth factor receptor-2 (HER2), prostate-specific membrane antigen (PSMA), estrogen receptor, folate receptor, and survivin are in accordance with the specific molecular markers of the primary tumor [14]. Since CTCs are found to have a very low concentration in the blood even in patients with metastatic cancer [15], highly sensitive technologies are required to detect and isolate these cells.

3.1.1. Capture and Isolation of CTCs

Advanced Microfluidic Technologies

Microfluidic devices offer high-throughput, high-resolution isolation and analysis of CTCs from blood samples. These platforms use microscale channels to capture and isolate CTCs based on physical properties such as size, density, deformability, or electric charges [16], or based on biological properties like the expression of specific protein markers, allowing for more sensitive and specific detection of rare CTCs (Figure 4).
In terms of biological properties, EPCAM-based isolation is considered a reliable method. However, in recent findings, CTCs can reduce the expression of specific markers, which allows them to escape from detection and result in false negative results. For instance, when CTCs undergo epithelial–mesenchymal transition (EMT), EpCAM expression is down-regulated while the expression of mesenchymal markers like N-cadherin increases [17,18]. In this case, negative selection can be adopted. Negative selection refers to the depletion of nonmalignant blood cells from blood using antibodies, such as targeting cell surface antigen CD45 on white blood cells [19]. It is noted that the drawbacks of negative selection include a lower purity of CTCs relative to the positive selection approach and the risk of depleting CTCs owing to being trapped in a mass of immune cells [20,21,22].
In terms of physical properties, CTCs show a larger size than normal blood cells. Size-based exclusion methods such as the isolation by size of epithelial tumor cells (ISET) filtration system have been developed to enable EpCAM-independent isolation of CTCs. The blood passes through the pores to allow size exclusion of CTCs so that larger CTCs are trapped [23]. However, this ISET filtration system has limitations including a lower purity of CTCs and the potential loss of smaller CTCs. Density gradient centrifugation is a fast separation method but it has a lower sensitivity. Dielectrophoresis (DEP) can separate CTCs from normal blood cells based on their unique dielectric properties. The dielectric properties depend on diameter, membrane area, density, conductivity, and volume, although they can be altered during prolonged storage [12].

Examples of Microfluidic Devices

The CTC-Chip can capture CTCs by coating the micro posts with different molecular markers such as anti-EpCAM antibodies. Using gold nanoparticles on a herringbone chip (HB-Chip) can isolate and release CTCs safely by chemical ligand-exchange reaction [14]. The CTC-iChip can undergo immunomagnetic sorting of CTCs by label-free, positive, and negative selection methods based on the antigens used [24]. All in all, microfluidic devices facilitate the efficient isolation of CTCs by different mechanisms.

3.1.2. Strategies on CTCs Analysis

After the isolation of CTCs, they can be identified by immunological, molecular, or functional assays as shown in Figure 5 [25].

Immunological Technologies

In the immunological assay, CTCs are detected by antibodies against epithelial, mesenchymal, tissue-specific, or tumor-associated markers. For instance, not only EpCAM but also some of the epithelial markers including cytokeratin members and E-cadherin or mesenchymal markers like vimentin (VIM) and fibronectin can be detected [14]. Tissue-specific markers like prostate-specific antigen (PSA) show a high specificity to prostate cancer and tumor-associated markers like HER2 are overexpressed in breast cancer. It is important to note that the population of CTCs shows heterogeneity, indicating that there is a wide variety of CTC markers among various cancer types, even in the same patient [14]. Therefore, it is challenging to define the origin of the CTC population by using limited molecular markers only.

3.1.3. Molecular Technologies (RNA-Based)

Tumor cells have changed at genetic and transcriptomic levels, allowing them to escape from the primary tumor and survive as CTCs [26], hence RNA analysis of CTCs can be used for detection and quantification. CTCs are identified by quantitative reverse transcription polymerase chain reaction (RT-qPCR), RNA sequencing (RNA-seq), and RNA in situ hybridization (RNA-ISH). RT-qPCR is a frequently used technique to detect and monitor gene expression in CTCs due to its high sensitivity and cost-effectiveness when compared to other RNA-based detection methods [26]. Since the concentration of CTCs is often low in blood, droplet digital PCR (ddPCR) may also be applied. The CTCs’ RNA is separated into many partitions by lipid droplets to improve the detection of CTCs in the early stage of cancer due to the increased concentration of CTCs’ RNA and maximized amplification in a lipid droplet [27]. RNA-ISH allows the detection of localized RNA in CTCs without denaturing the cells. It can be used without any enrichment process to reduce the risk of losing CTCs during processing while obtaining a high sensitivity, namely the CTCscope method to detect eight epithelial markers and three EMT markers [28,29]. RNA-seq can study a large number of genes simultaneously in either total CTCs or a single CTC to determine the gene expression profile. Bulk RNA-seq demonstrates the average gene expression in a CTC population, while single-cell RNA-seq enables the analysis of cellular heterogeneity and identification of cellular types according to their genes in CTCs, thus illustrating the precision advantage of single-cell analysis over bulk analysis [30,31]. Although CTC transcriptomics can be used for detection and providing valuable insights about the phenotype of individual CTCs, the main challenge is the reduced stability of RNA when compared to DNA, thus a validated protocol is required to avoid any analytical errors and false results [32].

Single-Cell Analysis of CTCs Using Microfluidic Devices

A study conducted by Aceto et al. (2014) demonstrated a high level of concordance between expression patterns of CTC clusters and single CTCs, highlighting the importance of single-cell resolution RNA sequencing [33]. Single-cell analysis of CTC clusters provided insight into tumor cell migration and metastatic properties [34]. Huang et al. (2023) and Cheng et al. (2023) developed microfluidic devices for the isolation and analysis of CTCs, emphasizing the importance of efficient capture and release of CTCs in cancer diagnosis and monitoring [35,36]. Cheng et al. (2023) introduced a poly(ethylene oxide) concentration gradient-based microfluidic device for the isolation of CTCs, highlighting the importance of efficient capture and release of these cells for cancer diagnosis and management [37]. Huang et al. (2023) developed a conductive nanofibers-enhanced microfluidic device for the efficient capture and electrical stimulation-triggered rapid release of CTCs [38]. This novel approach not only enables the effective detection of CTCs but also offers a mechanism for rapid release, which is crucial for cancer diagnosis and monitoring.

Functional Assays

Functional assays, such as drug sensitivity testing on isolated CTCs or CTC-derived xenograft models, can help predict treatment response and guide personalized therapeutic strategies based on the drug sensitivity profile of CTCs. Capturing viable CTCs can allow downstream culturing of CTC cell lines that are derived from patients [37]. Some methods can maintain CTC viability and culture possibility, such as density gradient centrifugation [38] and microfluidic chips [20]. However, all the mentioned methods require extra steps to demonstrate the CTCs’ viability. Therefore, the Epithelial Immuno-SPOT (EPISPOT) assay is developed to directly assess CTC viability with fewer steps.
EPISPOT is a method to detect viable CTCs only based on the specific tumor-associated proteins that are secreted, shed, and released by CTCs using an adaption of enzyme-linked immunospot (ELISPOT) technology [39]. Cell culture is required to accumulate enough amount of released marker proteins because the immunospots are the protein fingerprint created by viable cells only and the dead cells that do not secrete a sufficient amount of marker proteins are not detected [12]. The CTCs are cultured on a nitrocellulose membrane coated with specific antibodies, which are used to capture the specific protein secreted by CTCs. After that, the fluorophore-labeled antibodies are added to bind to the secreted proteins and the signal can be visualized under a fluorescence microscope [40]. EPISPOT often combines with the leukocyte depletion negative enrichment step and it can identify CTCs without epithelial features such as EpCAM-negative CTCs [39]. EPISPOT has already been validated for prostate cancer [41], breast cancer [42], melanoma [42], head and neck squamous cell carcinoma [43], and colorectal cancer [39], thus illustrating the clinical values of the assay.

3.2. Circulating Nucleic Acids

Circulating nucleic acids, including ctDNA and ctRNA, are fragments of nucleic acids released into the bloodstream by cells, including tumor cells. The release of circulating nucleic acids into the human circulatory system can originate from six sources, including apoptosis, necrosis, NETosis, erythroblast enucleation, EV, and exogenous sources [44].

3.3. Circulating Tumor DNA (ctDNA)

ctDNA is a portion of cell-free DNA (cfDNA) that is released from tumor cells and enters the circulatory system. The presence of epigenetic or genetic alterations, including tumor-specific methylation markers, rearrangements, copy number variations, and somatic point variants, allows ctDNA to be distinguished from normal cfDNA fragments [45]. The genetic and epigenetic modifications of ctDNA molecules may reflect the genome or epigenome of the cell of origin, thus it can be used to estimate prognosis, for identification of residual disease and treatment selection, and/or indicate potential risk of relapse [46]. The half-life of ctDNA in the circulatory system varies from minutes to 2.5 h [47] and the levels of ctDNA are generally very low, which may require highly sensitive technologies for the detection of ctDNA.

3.4. Circulating Tumor RNA (ctRNA)

RNA released from cells through apoptosis, necrosis, and active secretion is referred to as cfRNA [10]. CtRNA, including microRNA (miRNA) and long non-coding RNA (lncRNA), are a portion of cfRNAs that are present in the body fluids of tumor patients. These cfRNAs can be isolated from various types of body fluids such as blood, urine, breast milk, and other fluids [48].
MiRNAs are small non-coding RNAs that are essential for the post-transcriptional regulation of gene expression. They are desirable biomarkers for noninvasive cancer diagnosis due to their stability in body fluids. Moreover, the expression patterns of miRNAs are tissue-specific, suggesting that miRNAs could be used as a biomarker for cancer diagnosis, prediction, and prognosis [49]. Numerous studies have shown that specific miRNA signatures are associated with various cancer types. miRNA-21 is an extensively studied miRNA that is found to be upregulated in a number of malignancies, including breast, lung, and colorectal cancer [50,51,52].
lncRNAs are transcripts with no protein-coding capacity. These RNAs act as regulatory factors that play an important role in different cellular processes such as cell growth, differentiation, proliferation, and apoptosis [48]. lncRNAs are bound to RNA-binding proteins and are released either inside apoptotic bodies or encapsulated in exosomes, making them resistant to RNase degradation. Thus, they can exist stably in the circulatory system and could be used as a noninvasive biomarker [53]. Studies have found that lncRNAs are expressed in a cell- and tissue-type specific pattern, allowing them to be used in the diagnosis of various cancers such as lung cancer, breast cancer, gastric cancer, liver cancer, and prostate cancer by measuring the expression level of lncRNAs [54].

3.4.1. Isolation of Circulating Cell-Free DNA

After blood taking, it proceeds to plasma processing and DNA extraction. One of the main factors that affect the sensitivity of detecting ctDNA is the unintended release of cellular genomic DNA (gDNA) from the lysed blood cells [55]. Since ctDNA has a very low concentration in the peripheral blood, which already demonstrates a significant challenge to any analytical system, the unwanted increase in gDNA can further disturb the detection. Although it is discovered that a higher concentration of ctDNA is presented in the serum, the increased level may be caused by gDNA contamination due to the clotting process, resulting in diluting the targeted ctDNA concentration [56]. Hence, the plasma sample is a more reliable source of ctDNA to minimize the contamination from gDNA. The key to reducing the release of gDNA is to process the blood sample rapidly after collection without any delay and undergo a two-step centrifugation procedure for plasma preparation, which is recommended by many experts [57]. The two-step plasma preparation protocol includes a low-speed centrifugation (800–2000× g) to remove blood cells and prevent cell lysis, followed by a high-speed centrifugation (14,000–16,000× g) to remove cellular remnant to improve the plasma quality [58]. In view of the rigorous requirements such as rapid processing and the availability of high-speed centrifugation, an alternative approach of inhibiting nuclease activity in blood and stabilizing cells in the blood collection tube (BCT) can be adopted. The most common and cost-effective BCT is the EDTA collection tube, which can inactivate DNase activity. It is noted that the best performance is maintained when the EDTA tube samples are processed within 2 h after venipuncture [59], otherwise, there is a possibility of gDNA contamination from leukocytes when the blood storage time is more than 4 h [57]. Therefore, other commercial BCTs can be used when the plasma preparation time is more than 4 h, such as Streck Cell-Free DNA BCT [60], PAXgene Blood ccfDNA Tube [61], and Roche Cell-Free DNA Collection Tubes [62]. They can extend the processing time for at least 48–72 h, irrespective of temperature and without significant compromise to ctDNA detection [63].
In DNA extraction, there are different developed methods that can be classified into the following four methods: the magnetic bead–based method, the silica column–based method, the filtration-based method, and the phenol–chloroform-based method [64]. Comparing different types of commercial DNA extraction kits, they illustrate a large variation in terms of the extraction method, plasma input, throughput, and price [65]. The magnetic bead–based method has advantages regarding cost, speed, scalability, and automation, while the silica column–based method can obtain a higher yield [66]. Therefore, the choice of DNA extraction method is also an important factor to consider.

3.4.2. Circulating Nucleic Acids Detection and Analysis

Digital PCR and Next-Generation Sequencing (NGS)

Ultrasensitive sequencing technologies are crucial for the analysis of ctDNA due to the low abundance of ctDNA in biological samples [67]. These technologies, such as ultradeep massively parallel sequencing with unique molecular identifier tagging enable the detection and quantification of minute amounts of ctDNA, which are typically released into the bloodstream by tumor cells undergoing cell death [67]. By utilizing ultradeep coverage sequencing methods, such as Illumina (San Diego, CA, USA) and Thermo Fisher (Waltham, MA, USA), researchers can achieve comparable performance to droplet digital PCR (ddPCR), like Bio-Rad (Hercules, CA, USA)’s QX200 and Thermo Fisher’s QuantStudio, for the detection and quantification of ctDNA from cancer patients [67,68]. For accurate identification of and quantification of genetic changes in ctDNA, the ultrasensitive nature of these sequencing technologies is essential, providing insight into tumor dynamics, treatment response, and disease progression based on the molecular profile of ctDNA, which ultimately guides personalized therapeutic strategies.

Methylation Profiling

Methylation analysis of ctDNA has been used in cancer diagnostics and monitoring. Numerous studies have investigated the importance of DNA methylation patterns in ctDNA for early detection, treatment assessment, and personalized medicine approaches. Tamkovich et al. (2022) discussed the potential use of aberrantly methylated DNA in liquid biopsy for patients with breast cancer, emphasizing its early involvement in cancer cell transformation [69]. A recent study by Markou and colleagues (2022) explored the role of DNA methylation analysis in early-stage non-small cell lung cancer (NSCLC) cases, demonstrating the potential for early cancer detection through DNA methylation analysis [70].

Fragmentomics Analysis

Fragmentomics analysis of cfDNA and ctDNA is essential for understanding the fragmentation properties of cfDNA, including the size distribution and biochemical characteristics of free ends. ctDNA is generally observed to have shorter fragment lengths compared to cfDNA [71]. Studies have shown that fragmentomic analysis provides comprehensive information about the entire cfDNA population, offering insights into tissue-of-origin and pathologies associated with altered fragmentomic profiles [72,73,74]. Moreover, cfDNA fragmentomics has been crucial in distinguishing cancer-derived cfDNA, determining the tissue of origin, and aiding in the early detection of cancer recurrence [75,76]. The analysis of cfDNA fragmentation patterns can also help infer the cell types contributing to the cfDNA milieu, such as cancer cells [77].
Additionally, cfDNA fragmentomic features have been shown to reliably discriminate between different disease states, highlighting the potential of liquid biopsy cfDNA fragmentomics in clinical diagnostics [78]. Furthermore, studies have emphasized the importance of understanding the origin and fragmentation mechanisms of cfDNA during physiological and pathological processes to leverage cfDNA as a molecular diagnostic tool [73]. By integrating cfDNA end motifs and fragment lengths, it is possible to identify active genes in liquid biopsies, providing insights into gene activity and underlying biological processes. Moreover, epigenetic profiling of cfDNA fragmentomes can also offer valuable information on DNA methylation patterns, aiding in predicting the CpG methylation of cfDNA without the need for bisulfite treatment [79]. Using fragmentomics to examine cfDNA fragments offers the potential for enhancing early cancer detection, monitoring treatment response, and developing personalized therapeutic strategies based on the molecular characteristics of cfDNA fragments.

3.5. Extracellular Vesicles (EVs)

EVs, including exosomes and microvesicles, ectosomes, oncosomes microparticles, and many others, are groups of membrane-closed vesicles containing DNA, RNA, proteins, and lipids enclosed in a phospholipid biolayer. They play a crucial role in intercellular communication and disease progression, including cancer [80]. In addition, EVs are secreted by all cell types and are present in various types of body fluids, allowing the collection of the biomarkers present in EVs for cancer diagnosis and management. For example, exosomes are small EVs that are emerging biomarkers for cancer diagnosis and guide treatments. Studies have found that exosomes are involved in the occurrence and development of various tumors, which act as carriers in tumor cells, allowing them to escape immune system surveillance [81]. Exosomes are also able to create a suitable microenvironment for tumor growth and guide the direction of tumor cell metastasis [82,83]. To improve the understanding of circulating EVs, researchers are integrating multi-omics approaches, implementing machine learning algorithms, and standardizing isolation and analysis protocols [84].

3.5.1. EV Isolation and Characterization Technologies

The isolation and characterization of EVs are essential for understanding their functions and potential therapeutic applications. Several techniques have been developed and refined to address the challenges associated with EV isolation and characterization. The size exclusion chromatography (SEC) method is one of the most common methods for isolating EVs from plasma in recent years due to its ability to rapidly isolate relatively pure EVs [85,86]. Nevertheless, SEC is not suitable for all downstream applications, highlighting the need to develop a variety of isolation techniques to meet the needs of different research projects. An alternative method of EV isolation is ultracentrifugation but it can be time-consuming and difficult to implement in a clinical environment [87]. There have been recent advances in EV isolation efficiency and biomarker profiling by using membrane affinity–based methods and microfluidics [88,89]. These advancements are essential for improving diagnostic capabilities and advancing precision medicine initiatives. Furthermore, the development of novel techniques such as the EVtrap [90] and the salting-out approach [91] offer alternatives for achieving high purity and efficiency in EV isolation. Characterizing EVs is equally important and a variety of methods have been used for this purpose, including nanoparticle tracking analysis (NTA), transmission electron microscopy, and Western blotting.

EV Proteomic and RNA Profiling

The integration of proteomic and RNA profiling in EV research holds great potential for advancing precision medicine and improving disease diagnosis and monitoring. The development of EV proteomic profiling has been promising for the detection and monitoring of cancer, emphasizing the importance of understanding the protein cargo of EVs for clinical applications [92,93]. Through the use of NGS, comprehensive analyses of EV RNA content have been carried out, which have shed light on the multitude of RNA species present in EVs as well as the functional implications of their presence [94].

3.6. Metabolites

Metabolomics can be utilized to identify metabolites with a molecular weight of less than 1 kDa present in body fluids [95]. There has been increasing interest in metabolomic techniques in cancer research, highlighting their potential for identifying cancer-specific metabolites in liquid biopsy samples. For instance, metabolomics has been used to study dynamic responses and drug resistance in gastric cancer cells [96], analyze the metabolic changes associated with prostate cancer using integrated RNA and metabolite profiling of urine liquid biopsies [97], and develop metabolite classifiers for NSCLC diagnosis using ultrahigh-performance liquid chromatography-mass spectrometry/mass spectrometry (UPLC-MS/MS) [98]. In addition, Wei et al. (2021) utilized UPLC-MS to analyze serum metabolic features of gastric cardia adenocarcinoma (GCA) patients and healthy controls in order to identify a novel metabolic biomarker for early detection and prognosis in GCA patients [99]. Moreover, a study has demonstrated that benzoic acid might have the potential to be a diagnostic marker of colorectal cancer [100] and significant differences in metabolite levels can be found in different stages of the same cancer type [101]. The findings of these studies highlight the potential of metabolomics for advancing precision medicine and improving patient outcomes in cancer diagnosis, biomarker discovery, and treatment strategies.

4. Applications in Cancer Management

In cancer management, liquid biopsy plays a crucial role in several aspects. Liquid biopsy has demonstrated significant promise in early detection, diagnosis, monitoring cancer progression, treatment selection, and personalized medicine, as well as in detecting MRD and predicting relapse across different stages of cancer [102].

4.1. Early Detection and Diagnosis

Regarding early detection, liquid biopsy has transformed cancer screening by facilitating the identification of biomarkers in body fluids, enabling the detection of cancer at its early stages [103,104,105,106]. Due to the low amounts of ctDNA released by early tumors, research shows that variants in ctDNA alone have limited sensitivity for early-stage cancer detection. Consequently, different strategies were developed to improve sensitivity. For instance, a strategy called CancerSEEK, which involves the evaluation of ctDNA and circulating proteins such as carcinoembryonic antigen (CEA), cancer antigen 125 (CA-125), and cancer antigen 19-9 (CA19-9), has been proposed to increase the sensitivity for early detection [104]. Studies have demonstrated varying sensitivities across different cancer types, with the highest sensitivity for ovarian cancer and the lowest for breast cancer [8,50,107,108,109,110,111]. Therefore, combining ctDNA analysis with the assessment of circulating proteins can enhance the overall sensitivity for early detection.
In addition, ctRNA has shown promise in providing noninvasive information about gene expression without the need for biopsies through the use of circulating RNA sequencing [112]. Shen et al. (2023) emphasizes the importance of ctRNA in the early detection of cancer, emphasizing the clinical application of liquid biopsy in endometrial carcinoma [113]. In addition, miRNA is the most sufficient cfRNA molecule in body fluids so it also has the potential for enabling early diagnosis. For instance, various types of miRNA in sputum such as miR-145, miR-126, and miR-7 have been shown to obtain a high sensitivity and specificity to detect NSCLC [114]. However, there is a lack of consensus on miRNA panels to detect cancer development so validation studies of the discovered miRNA [115] and new studies of undiscovered miRNAs based on the genetic background of each population, type, and stage of cancer development is recommended [116].
Moreover, recent advances in liquid biopsy techniques, including the detection of exosome microRNAs and protein biomarkers, have made them more useful for diagnosing and monitoring cancer, providing real-time feedback and noninvasive disease assessment [103,117]. Recent advances in nanotechnology-enabled biosensors have allowed the detection of exosomal RNAs and proteins for cancer screening, diagnosis, and prognosis [118]. There has been evidence that short-length exosomal DNA or RNA derived from tumor cells can be detected more readily in liquid biopsy, demonstrating its high sensitivity [119]. Nanotechnology and antibody-conjugated signals in nanocavities have also been proposed to detect cancer markers in liquid biopsy [120].

4.2. Prognostication and Predictive Biomarkers

Recent studies have underscored the significant role of liquid biopsy in identifying prognostic and predictive biomarkers across various types of cancer, including esophageal cancer [121], prostate cancer [122], bladder cancer [123], colorectal cancer [10], and NSCLC [124]. Liquid biopsy has been shown to have both prognostic and predictive value in cancer management, offering molecular and phenotypic information about cancers [125]. The analysis of nucleic acids in liquid biopsies has demonstrated prognostic potential and predictive value for guiding treatment decisions in colorectal cancer and hepatocellular carcinoma [126,127].

4.3. Detection of Minimal Residual Disease (MRD)

Liquid biopsy has demonstrated significant promise in the detection of minimal residual disease (MRD) in various types of cancer. For instance, highly sensitive liquid biopsy assays have been successfully used to detect and characterize MRD in colorectal cancer patients. MRD occurs when persistent cancer cells spread from the primary lesion to distant organs or into the bloodstream when no radiological relapse has occurred after resection of the tumor [128]. In lung cancer, Sardarabadi et al. (2021) emphasized the use of liquid biopsy–based biosensors for detecting and assessing early stages of disease, including resistance to treatment and tumor mutational burden [129]. Furthermore, liquid biopsy is being used as a unique minimally invasive tool for breast cancer, diagnosing its MRD, studying its metastasis biology, and exploring its drug resistance mechanisms [130]. Moreover, it has been shown that ctDNA is useful in identifying MRD in locoregional gastric cancer, predicting recurrence, and guiding treatment regimens in the presence of MRD [131].

4.4. Treatment Selection and Personalized Medicine

The use of liquid biopsy to identify specific biomarkers in body fluids has significantly influenced treatment selection and personalized medicine. Several recent studies have highlighted the importance of liquid biopsy when selecting targeted therapies and evaluating treatment responses. Personalized medicine has benefited tremendously from liquid biopsy, enabling the identification of tumor-specific biomarkers that are used to optimize treatment regimens and monitor therapeutic outcomes [132,133]. The use of liquid biopsy has enabled accurate prediction, monitoring, and rational selection of appropriate therapies for individual patients by analyzing ctDNA and CTCs as well [134,135]. Aside from this, liquid biopsy has also been demonstrated to have promise in guiding therapeutic decisions for many types of cancer by providing real-time molecular information that is useful in guiding treatment choices and evaluating response to treatment. Lung cancer, breast cancer, colorectal cancer, and prostate cancer are introduced in this review because they were the global top four cancer types, respectively, in both sexes in 2022 in terms of incidence and mortality [136].

4.5. Lung Cancer

Lung cancer is one of the most common cancers with the highest mortality rate but with a low rate of early detection so most patients are diagnosed at the advanced stage, which leads to a lower survival rate. A prescreening program is thus an important tool to enable early detection and liquid biopsy can detect lung cancer in a safer and earlier way. Although tissue biopsy is regarded as the current gold-standard method, it may not be the best choice for detection or monitoring lung cancer. Firstly, a tissue biopsy may not be completely feasible because of the tumor location or performance status [137]. Secondly, tumor heterogeneity can result in noncomprehensive genomic profiles by tissue biopsy obtained in one site only [138]. Lung biopsy shows a high incidence of major and minor complications and tissue biopsy may yield insufficient DNA for genetic analysis [139]. Therefore, with the properties of noninvasiveness, simple accessibility, and great repeatability, liquid biopsy is considered a promising alternative to traditional tissue biopsy.
Lung cancer can be primarily classified into two types—NSCLC and small cell lung cancer (SCLC)—in which NSCLC accounts for about 85% of all cancer types [138]. A targeted therapy is developed for NSCLC patients to increase overall survival, namely the EGFR tyrosine kinase inhibitors (TKI) for patients with the EGFR variant. Kolesar et al. explored the use of liquid biopsy in guiding the selection of TKI therapies at various stages of lung cancer treatment to enhance the precision and efficacy of therapy for lung cancer patients with EGFR variants and resistance to standard treatments [140]. EGFR T790M is an acquired drug-resistant EGFR-dependent variant. Although SensiScreen EGFR Liquid assays are developed to detect EGFR variants at high sensitivity and specificity by using liquid biopsies as samples [141], EGFR T790M does not necessarily have to be screened for because of the development of Osimertinib, an approved treatment for EGFR-positive patients exhibiting a T790M resistance variant [142]. For other EGFR-independent variants that result in drug resistance, including Kirsten rat sarcoma (KRAS), phosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA), HER2, RAF murine sarcoma viral oncogene homolog B1 (BRAF), and mesenchymal–epithelial transition (MET), they can be detected by liquid biopsy [143]. Hence, by integrating liquid biopsy with pharmacogenomics testing, lung cancer precision therapy can be enhanced by detecting tumors, monitoring variants and acquired resistance, and assisting in the selection of precise drugs through serial molecular profiling based on blood samples [140].

4.6. Breast Cancer

In breast cancer, there is an increase in both ctDNA and CTCs that correlate with cancer development. The analysis of these biomarkers is beneficial to manage advanced or early breast cancer patients. It is found that a high concentration of ctDNA is related to a more aggressive or relapsing disease [144]. CTCs are also useful in characterizing phenotypes and determining the possibility of metastatic spread and prognosis [145]. Hence, liquid biopsy shows its potential applications in diagnosis, treatment, and management.
Breast cancer can be classified into four subtypes based on molecular information, including luminal A, luminal B, HER2-positive, and triple-negative subtypes [146]. This molecular classification can categorize the patients who can benefit from the targeted therapy. However, when breast cancer becomes metastatic, it is difficult to be controlled. After confirming the status of hormone receptors and HER2 by a biopsy of metastatic breast cancer lesions, the determination of metastatic phenotype can be performed by repeated sampling, which can provide useful information on molecular changes and treatment customization [147]. For example, the CD47 and PD-L1 markers in CTCs correlate with disease progression [148]. Moreover, the high rates of pSTAT3+ and TLR4+ in CTCs are detected in early-stage and metastatic patients, respectively [149]. The metabolic classification of CTCs has the potential to help recognize aggressive CTC subpopulations and discover new targeted therapies.
The detection of the PIK3CA variant is important for breast cancer because the PIK3CA variant can result in resistance to HER2-targeted therapies [150]. It is noted that there is a high incidence of discrepancy in the PIK3CA variant between primary breast cancer tumors and metastases, hence the detection of metastatic lesions is essential [151]. However, sufficient tumor tissue samples may not always be available or accessible for detection, or the tissue biopsy obtained at the time of surgical resection may not be able to reflect the current tumor molecular characteristics [151]. In view of that, liquid biopsy provides a real-time examination of genetic alterations in a minimally invasive way. In the SOLAR-1 trial, the assessment of PIK3CA mutational status was compared between formalin-fixed paraffin-embedded (FFPE) samples from the primary or metastatic sites, and ctDNA from the plasma in the same patient [152]. The result showed that PIK3CA variant testing on liquid biopsy achieves a higher specificity (97%) but lower sensitivity (55%) when compared to tissue biopsy. Similarly, a study by Nakai et al. also demonstrated that the PIK3CA variant is detected with a lower sensitivity in liquid biopsy, although heterogeneity related to the PIK3CA variant can be detected [153]. According to the results, the Therascreen PIK3CA RGQ PCR Kit (QIAGEN GmbH, QIAGEN Strasse 1, 40,724 Hilden, Germany) is approved by the U.S. Food and Drug Administration (FDA) to conduct the PIK3CA mutational analysis on both tissue biopsy by using gDNA from FFPE samples, and liquid biopsy by using ctDNA extracted from plasma samples [151]. The kit can detect 11 variants in PIK3CA, including exon 7 (C420R), exon 9 (E542K, E545A, E545D, E545G, E545K, Q546E, and Q546R), and exon 20 (H1047L, H1047R, and H1047Y) [154]. Therefore, liquid biopsy can provide valuable information for treatment selection and monitoring in breast cancer patients.
The estrogen receptor 1 (ESR1) variant plays a significant role in endocrine therapy (ET) resistance for metastatic breast cancer, and it can be detected by liquid biopsy. ddPCR and NGS are two commonly used methods for identifying ESR1 variants in ctDNA, in which ddPCR provides higher sensitivity and accuracy, while NGS can evaluate larger regions like gene panels without prior knowledge of the variants [155]. Moreover, a study by Urso et al. assessed the concordance of the ESR1 variant in tumor tissue samples from a metastatic lesion and ctDNA from plasma. The result demonstrates a high concordance rate between the tumor tissue and ctDNA (91%), indicating the reliability and feasibility of liquid biopsy for the ESR1 variant detection [156]. Compared to tissue sequencing, ctDNA may be superior because it can accurately reveal the molecular profile of metastatic cancer [152]. Therefore, integrating liquid biopsies into the diagnostic workflow in breast cancer patients can help highlight the resistance to ET and guide treatment decisions.

4.7. Colorectal Cancer (CRC)

In CRC, liquid biopsy has shown promise in various applications, such as early-stage detection, guiding and monitoring treatments, predicting relapses and metastases, revealing tumor heterogeneity, and detecting MRD [157].
In the early diagnosis and screening of CRC, hemoccult test and fecal immunochemical test (FIT) are two noninvasive detection methods that are mostly studied, but they demonstrate some limitations such as having a low sensitivity [158,159]. Hence, liquid biopsy was evaluated as an alternative and has shown satisfactory performance. CTCs have the potential to become a screening tool for early-stage CRC. Tsai et al. indicated a high sensitivity and adequate specificity of the CellMax (CMx) blood-based CTC test [160]. In addition, a combined analysis of the CTC count with an immunochemical fecal occult blood test (iFOBT) or serum Carcinoembryonic antigen (CEA) assay was conducted by Tsai et al., showing that the implementation of CTC testing can reduce the false positive rate of iFOBT and increase the disease detection rate of a serum CEA assay [161].
Apart from CTCs, ctDNA is also useful for CRC screening. Epigenetic changes including DNA methylation and histone modifications are some of the early events of carcinogenesis, which can be reflected in ctDNA to aid early diagnosis [162]. The FDA-approved Epi proColon test uses the real-time PCR assay to detect the methylation of septin9 (SEPT9) DNA in CRC patients [163]. However, it shows a limitation of having low sensitivity (68.2%) and specificity (79.1%) across all stages of CRC detection [164,165]. Another test, namely the ColoSure test, is a single-marker test to identify the methylation of VIM in CRC from stool DNA samples with a sensitivity ranging from 38% to 88% [166]. Recently, the ColoSure test has been replaced by the more efficient FDA-approved Cologuard test. It is a multitarget stool DNA test that assesses KRAS variants, methylation of bone morphogenetic protein 3 (BMP3) and Neuregulin 4 (NRG4), and hemoglobin concentration [167]. The sensitivity and specificity are reported to be 92.3% and 86.6%, respectively, showing the improved diagnostic accuracy of CRC [168].
Liquid biopsy can also provide insights for treatment monitoring and efficacy evaluation [169]. Boysen et al. investigated post-treatment ctDNA analysis in ddPCR and mass spectrometric–based multiplexed platform, MassARRAY (MA), showing that ctDNA can be used as a marker for MRD in the post-treatment of CRC [170]. Another study by Zhou et al. demonstrated the potential applications of ctDNA in locally advanced rectal cancer (LARC), such as reflecting real-time tumor bulk, predicting tumor response to neoadjuvant chemoradiotherapy (nCRT), and predicting the prognosis [171].

4.8. Prostate Cancer

The treatment of prostate cancer involves androgen deprivation therapy (ADT), which is regarded as the standard treatment and has shown its effectiveness in the initial management of advanced or metastatic prostate cancer. However, some patients develop resistance to the therapy, resulting in castration-resistant prostate cancer (CRPC) [172]. Androgen receptor signaling inhibitor (ARSI) is a drug blocking the androgen receptor at various levels that can prolong the survival of CRPC patients [173], but some patients also develop resistance to ARSI due to alterations in increasing AR transcriptional activity and target gene expression [174]. It is noted that the PSA marker, bone scintigraphy, computed tomography (CT), and positron emission tomography (PET) scans do not provide information on disease drivers or resistance mechanisms [175]. Other methods are required to provide a more comprehensive disease characterization, thus liquid biopsy assays are developed to provide insights into drug resistance mechanisms.
The androgen receptor splice variant 7 (AR-V7) is the most commonly detected variant associated with ARSI resistance and it can be used as a predictive variant to ARSIs such as enzalutamide and abiraterone acetate [176]. It can evade the action of anti-androgen therapies that target the ligand-binding domain (LBD) by retaining its DNA-binding domain but lacking the LBD [177], making it a negative treatment response. AR-V7 detection can be achieved in CTCs and ctRNA, such as using the EPIC Sciences CTC AR-V7 nuclear localization assay [178] or the AdnaTest ProstrateCancerDetect AR-V7 quantitative polymerase chain reaction (qPCR) assay by QIAGEN [179]. According to a study by Nimir et al., CTC-based detection obtains a higher sensitivity and specificity for AR-V7 when compared to ctRNA, indicating that CTC testing may be more reliable [180].
Homologous recombination repair (HRR) is responsible for repairing the double-stranded DNA breaks or interstrand cross-links. Defective DNA repair (homologous recombination deficiency, HRD) is regarded as a hallmark of prostate cancer [181]. BRCA (BRCA1 and BRCA2) is a tumor suppressor gene that is critical for HRR by recruiting proteins such as RAD51 [182]. BRCA variants are consistently associated with an HRD phenotype in multiple cancers including prostate cancer, while other HRR pathway genes such as ATM, PALB2, or RAD51 are also associated with an HRD phenotype [183]. It is observed that HRD can cause tumors to be more sensitive to poly(ADP-ribose) polymerase inhibitors (PARPi) treatment [183] such as olaparib or rucaparib. FoundationOne Liquid CDx test is an FDA-approved next-generation sequencing–based test to identify variants in BRCA1, BRCA2, and ATM in cfDNA to guide the treatment of olaparib or rucaparib [184]. Other tests, including BRACAnalysis CDx, Illumina TruSight Oncology 500 (TSO500), and Guardant360 CDx, are also developed by using plasma cfDNA as samples [185,186,187]. All in all, liquid biopsy offers a minimally invasive approach for detecting treatment resistance and variants to improve the guidance of treatment to patients.
CTC enumeration, such as the CellSearch assay, which is approved by the FDA [188], can be used for monitoring treatment response and prognosis. A study by de Kruijff et al. demonstrated that CTC count is independently associated with poor progression–free survival and overall survival of metastatic CRPC patients treated with cabazitaxel [189]. Apart from the CTC count, several markers in CTCs such as PSMA [190] or EMT markers [191] are also found to be associated with the prognosis of metastatic prostate cancer patients. Therefore, liquid biopsy may explore a better way for the assessment of prognosis.

5. Example of Clinical Trials Evaluating Liquid Biopsy for Cancer

There have been numerous clinical trials on liquid biopsy, including the detection of actionable variants, evaluating treatment response, and predicting prognosis. For instance, an ongoing clinical trial at the Jewish General Hospital in Montreal, Canada, called “TRICIA” (TRIPLE Negative Breast Cancer Markers In Liquid Biopsies Using Artificial Intelligence), is developing a test or score based on ctDNA expression within a cohort of patients with triple-negative breast cancer, which demonstrates the potential of liquid biopsy to guide treatment decisions based on specific breast cancer subtypes. [192]. Another prospective phase II clinical trial in CRC utilized liquid biopsies to detect spatial and temporal heterogeneity of resistance to anti-EGFR monoclonal antibodies by combining sequential profiling of ctDNA with mathematical modeling and imaging of tumor progression. This clinical trial showed that liquid biopsy can be used to monitor the progression of cancer and the response to treatment [193]. Furthermore, EGFR variant status is being evaluated in NSCLC patients using the ddPCR method in ongoing clinical trials, suggesting that liquid biopsy can play a major role in guiding treatment decisions for NSCLC patients [194]. As a result of these trials, it is evident that liquid biopsy can be useful for identifying specific variants in lung cancer patients and guiding targeted therapies.

6. Liquid Biopsy Regulatory Considerations and Challenges

Integrating liquid biopsy into clinical practice presents several regulatory challenges and considerations. Regulatory issues, standardization of methods, diagnostic performance, and further research are key factors affecting the integration of liquid biopsy in the clinic [195]. Challenges such as a lack of consensus in preanalytical and analytical processes, clinical validation, regulatory approval, and low amounts of CTCs, ctRNA, and ctDNA hinder the successful implementation of liquid biopsy in clinical settings. The success of regulatory pathways for liquid biopsy diagnostics has been attributed in part to the incremental value of FDA approval for tests developed under the Clinical Laboratory Improvement Amendment (CLIA), as well as their complexity, which can hinder their widespread replication in CLIA laboratories [196]. In addition to its clinical utility, liquid biopsy is used to noninvasively monitor tumor genomes and to ensure appropriate treatment for patients. However, liquid biopsy remains a challenge in effective cancer management due to the lack of sensitive and specific biomarkers, the limitations of sampling single primary tumors, and the difficulty of monitoring cancer progression over time [197].
Other the challenges faced in integrating liquid biopsy into clinical practice is the need to standardize techniques, validate assays, and interpret results [198]. Considering the complexity of liquid biopsy approaches, robust validation processes are required to ensure accurate and reliable results [199]. Moreover, tumor heterogeneity, clonality, and the dynamic nature of tumors pose challenges when interpreting liquid biopsy results for effective cancer management [200]. The lack of standardization leads to different issues such as low reproducibility of results, technical and biological variability, and difficulty in interpreting data, resulting in problems of integrating liquid biopsies into clinical practice [198].
To enhance the acceptance of liquid biopsy procedures in routine clinical practice, several strategies can be implemented based on the available literature. Firstly, the development and adherence to standardized protocols and guidelines are necessary. The implementation of liquid biopsy in clinical practice relies greatly on international standards, such as from the International Organization for Standardization (ISO). It is essential for different laboratories and settings to follow standardized protocols for sample collection, processing, and analysis. In cancer management, following international standards helps harmonize practices, improve data quality, and facilitate information exchange [201]. For example, the workflow and quality evaluation of sequencing data from massively parallel sequencing should be based on ISO20397-1:2021 [198] and ISO20397-2:2021 [198]. To be used in clinical laboratories, assays should follow ISO15189 [198]. A standardized liquid biopsy method and workflow could further enhance the clinical usefulness of this approach in cancer management. Healthcare providers can ensure accurate, reproducible, and clinically relevant liquid biopsy results by following established guidelines and protocols [162]. Additionally, standardizing approaches facilitate the sharing of data, collaboration, and integration of liquid biopsy into routine clinical practice [198,202]. Secondly, the validation of the assay according to the international standards is very important. For instance, the implementation of NGS-based genomic profiling assays using blood samples for analyzing cfDNA can overcome practical challenges with sample integrity and failure, enabling liquid biopsy in advanced solid tumor patients to be used clinically [203]. Thirdly, addressing preanalytical considerations such as standardizing collection methods, optimizing storage conditions, and ensuring proper handling of samples are essential to mitigate variability and improve the diagnostic accuracy of liquid biopsy procedures, which can assist in overcoming challenges related to low circulating tumor content and interpreting sequencing data, making liquid biopsy more feasible in routine clinical practice [204].
To ensure the accessibility and affordability of liquid biopsy for all patients, establishing standardized reimbursement policies and guidelines is crucial to help address cost barriers and ensure that all patients have equal opportunities to benefit from liquid biopsy technologies, ultimately improving cancer care and patient outcomes. In addition, the field of liquid biopsy can progress to provide more cost-effective cancer diagnosis and monitoring by creating user-friendly tools and promoting research and technological advancements. Moreover, healthcare systems can facilitate collaborations among researchers, industry partners, and regulatory bodies to drive innovation and cost reduction [205].
In the future, technological advances could significantly enhance the possibilities and applications of liquid biopsy in cancer diagnosis and treatment. Advances in technologies such as dPCR and NGS will revolutionize liquid biopsy analysis, making it possible to detect ctDNA/ctRNA and other biomarkers more precisely and sensitively [145]. Furthermore, the integration of microfluidics and artificial intelligence are expected to further improve the accuracy and efficiency of liquid biopsy, enabling real-time monitoring of disease progression and treatment responses [206]. As a result of these technological innovations, the utility of liquid biopsy is poised to increase in precision oncology, providing noninvasive and dynamic insights into tumor biology, treatment response, and disease monitoring [201]. The development of liquid biopsy will play an increasingly important role in personalized medicine, transforming cancer care and improving patient outcomes.

7. Conclusions

Liquid biopsy has emerged as an important tool in cancer management, providing noninvasive and real-time insights into tumor biology and guiding personalized treatment plans. Through the dynamic insight provided by liquid biopsy into tumor characteristics and treatment responses, liquid biopsy has profound clinical implications, revolutionizing personalized treatments and cancer diagnostics. While liquid biopsy has its challenges, such as standardization and cost-effectiveness, it holds enormous promise for improving cancer care and patient outcomes.

Author Contributions

Writing—Original Draft Preparation, H.-Y.H. and K.-S.C.; Review and Editing, H.-Y.H., K.-S.C. and C.-M.K.; Conceptualization, Methodology and Supervision C.-M.K. and S.-C.W. All authors have read and agreed to the published version of the manuscript.

Funding

This research is supported by research impact fund P0034588 and internal dept funding P00432219.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Delmonico, L.; Alves, G.; Bines, J. Cell free DNA biology and its involvement in breast carcinogenesis. Adv. Clin. Chem. 2020, 97, 171–223. [Google Scholar] [CrossRef] [PubMed]
  2. He, J.; Xi, N.; Han, Z.; Luo, W.; Shen, J.; Wang, S.; Li, J.; Guo, Z.; Cheng, H. The Role of Liquid Biopsy Analytes in Diagnosis, Treatment and Prognosis of Colorectal Cancer. Front. Endocrinol. 2022, 13, 875442. [Google Scholar] [CrossRef] [PubMed]
  3. Ponti, G.; Manfredini, M.; Tomasi, A. Non-blood sources of cell-free DNA for cancer molecular profiling in clinical pathology and oncology. Crit. Rev. Oncol. Hematol. 2019, 141, 36–42. [Google Scholar] [CrossRef] [PubMed]
  4. Zhu, S.; Fang, Y.; Guo, K.; Zhang, N.; Xiang, N. Next-generation Liquid Biopsy Instruments: Challenges and Opportunities. Electrophoresis 2023, 44, 775–783. [Google Scholar] [CrossRef] [PubMed]
  5. Gao, W.; Chen, Y.; Yang, J.; Zhuo, C.; Zhang, H.; Shi, Y. Clinical Perspectives on Liquid Biopsy in Metastatic Colorectal Cancer. Front. Genet. 2021, 12, 634642. [Google Scholar] [CrossRef] [PubMed]
  6. Rastogi, A. Changing Role of Histopathology in the Diagnosis and Management of Hepatocellular Carcinoma. World J. Gastroenterol. 2018, 24, 4000–4013. [Google Scholar] [CrossRef] [PubMed]
  7. Hirahata, T.; Ul Quraish, R.; Quraish, A.U.; Ul Quraish, S.; Naz, M.; Razzaq, M.A. Liquid Biopsy: A Distinctive Approach to the Diagnosis and Prognosis of Cancer. Cancer Inform. 2022, 21, 11769351221076062. [Google Scholar] [CrossRef] [PubMed]
  8. Adhit, K.K.; Wanjari, A.; Menon, S.; Siddhaarth, K. Liquid Biopsy: An Evolving Paradigm for Non-invasive Disease Diagnosis and Monitoring in Medicine. Cureus 2023, 15, e50176. [Google Scholar] [CrossRef] [PubMed]
  9. 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]
  10. Jin, N.; Kan, C.-M.; Pei, X.M.; Cheung, W.L.; Ng, S.S.M.; Wong, H.T.; Cheng, H.Y.-L.; Leung, W.W.; Wong, Y.N.; Tsang, H.F.; et al. Cell-free circulating tumor RNAs in plasma as the potential prognostic biomarkers in colorectal cancer. Front. Oncol. 2023, 13, 1134445. [Google Scholar] [CrossRef]
  11. Ilie, M.; Hofman, P. Pros: Can tissue biopsy be replaced by liquid biopsy? Transl. Lung Cancer Res. 2016, 5, 420–423. [Google Scholar] [CrossRef] [PubMed]
  12. Vidlarova, M.; Rehulkova, A.; Stejskal, P.; Prokopova, A.; Slavik, H.; Hajduch, M.; Srovnal, J. Recent Advances in Methods for Circulating Tumor Cell Detection. Int. J. Mol. Sci. 2023, 24, 3902. [Google Scholar] [CrossRef] [PubMed]
  13. Liu, Y.; Wang, Y.; Sun, S.; Chen, Z.; Xiang, S.; Ding, Z.; Huang, Z.; Zhang, B. Understanding the versatile roles and applications of EpCAM in cancers: From bench to bedside. Exp. Hematol. Oncol. 2022, 11, 97. [Google Scholar] [CrossRef]
  14. Lin, D.; Shen, L.; Luo, M.; Zhang, K.; Li, J.; Yang, Q.; Zhu, F.; Zhou, D.; Zheng, S.; Chen, Y.; et al. Circulating tumor cells: Biology and clinical significance. Signal Transduct. Target. Ther. 2021, 6, 404. [Google Scholar] [CrossRef]
  15. Wu, T.M.; Liu, J.B.; Liu, Y.; Shi, Y.; Li, W.; Wang, G.R.; Ma, Y.S.; Fu, D. Power and Promise of Next-Generation Sequencing in Liquid Biopsies and Cancer Control. Cancer Control 2020, 27, 1073274820934805. [Google Scholar] [CrossRef] [PubMed]
  16. Descamps, L.; Le Roy, D.; Deman, A.-L. Microfluidic-Based Technologies for CTC Isolation: A Review of 10 Years of Intense Efforts towards Liquid Biopsy. Int. J. Mol. Sci. 2022, 23, 1981. [Google Scholar] [CrossRef]
  17. Yadav, A.; Kumar, A.; Siddiqui, M.H. Detection of circulating tumour cells in colorectal cancer: Emerging techniques and clinical implications. World J. Clin. Oncol. 2021, 12, 1169–1181. [Google Scholar] [CrossRef]
  18. Wiegmans, A.P.; Ivanova, E.; Naei, V.Y.; Monkman, J.; Fletcher, J.; Mullally, W.; Warkiani, M.E.; O’Byrne, K.; Kulasinghe, A. Poor patient outcome correlates with active engulfment of cytokeratin positive CTCs within cancer-associated monocyte population in lung cancer. Clin. Exp. Metastasis 2024, 41, 219–228. [Google Scholar] [CrossRef] [PubMed]
  19. Armakolas, A.; Kotsari, M.; Koskinas, J. Liquid Biopsies, Novel Approaches and Future Directions. Cancers 2023, 15, 1579. [Google Scholar] [CrossRef] [PubMed]
  20. Habli, Z.; AlChamaa, W.; Saab, R.; Kadara, H.; Khraiche, M.L. Circulating Tumor Cell Detection Technologies and Clinical Utility: Challenges and Opportunities. Cancers 2020, 12, 1930. [Google Scholar] [CrossRef]
  21. Lemma, S.; Perrone, A.M.; De Iaco, P.; Gasparre, G.; Kurelac, I. Current methodologies to detect circulating tumor cells: A focus on ovarian cancer. Am. J. Cancer Res. 2021, 11, 4111–4126. [Google Scholar] [PubMed]
  22. Chu, P.Y.; Hsieh, C.H.; Wu, M.H. The Combination of Immunomagnetic Bead-Based Cell Isolation and Optically Induced Dielectrophoresis (ODEP)-Based Microfluidic Device for the Negative Selection-Based Isolation of Circulating Tumor Cells (CTCs). Front. Bioeng. Biotechnol. 2020, 8, 921. [Google Scholar] [CrossRef] [PubMed]
  23. Eslami-S, Z.; Cortés-Hernández, L.E.; Alix-Panabières, C. Epithelial Cell Adhesion Molecule: An Anchor to Isolate Clinically Relevant Circulating Tumor Cells. Cells 2020, 9, 1836. [Google Scholar] [CrossRef] [PubMed]
  24. Fridrichova, I.; Kalinkova, L.; Ciernikova, S. Clinical Relevancy of Circulating Tumor Cells in Breast Cancer: Epithelial or Mesenchymal Characteristics, Single Cells or Clusters? Int. J. Mol. Sci. 2022, 23, 12141. [Google Scholar] [CrossRef] [PubMed]
  25. Guan, Y.; Xu, F.; Tian, J.; Chen, H.; Yang, C.; Huang, S.; Gao, K.; Wan, Z.; Li, M.; He, M.; et al. Pathology of circulating tumor cells and the available capture tools (Review). Oncol. Rep. 2020, 43, 1355–1364. [Google Scholar] [CrossRef] [PubMed]
  26. Hassan, S.; Blick, T.; Williams, E.D.; Thompson, E.W. Applications of RNA characterisation in circulating tumour cells. Front. Biosci.-Landmark 2020, 25, 874–892. [Google Scholar] [CrossRef] [PubMed]
  27. Kalinich, M.; Kwan, T.T.; Toner, M.; Haber, D.A.; Maheswaran, S. Quantitative Analysis of Circulating Tumor Cells Using RNA-Based Digital Scoring. In Tumor Liquid Biopsies; Schaffner, F., Merlin, J.-L., von Bubnoff, N., Eds.; Springer: Cham, Switzerland, 2020; pp. 77–88. [Google Scholar]
  28. Payne, R.E.; Wang, F.; Su, N.; Krell, J.; Zebrowski, A.; Yagüe, E.; Ma, X.J.; Luo, Y.; Coombes, R.C. Viable circulating tumour cell detection using multiplex RNA in situ hybridisation predicts progression-free survival in metastatic breast cancer patients. Br. J. Cancer 2012, 106, 1790–1797. [Google Scholar] [CrossRef] [PubMed]
  29. Bhatia, S.; Wang, P.; Toh, A.; Thompson, E.W. New Insights Into the Role of Phenotypic Plasticity and EMT in Driving Cancer Progression. Front. Mol. Biosci. 2020, 7, 71. [Google Scholar] [CrossRef] [PubMed]
  30. Cheng, Y.-H.; Chen, Y.-C.; Lin, E.; Brien, R.; Jung, S.; Chen, Y.-T.; Lee, W.; Hao, Z.; Sahoo, S.; Min Kang, H.; et al. Hydro-Seq enables contamination-free high-throughput single-cell RNA-sequencing for circulating tumor cells. Nat. Commun. 2019, 10, 2163. [Google Scholar] [CrossRef]
  31. Mihalcioiu, C.; Li, J.; Badescu, D.; Camirand, A.; Kremer, N.; Bertos, N.; Omeroglu, A.; Sebag, M.; Di Battista, J.; Park, M.; et al. Improved platform for breast cancer circulating tumor cell enrichment and characterization with next-generation sequencing technology. Am. J. Cancer Res. 2023, 13, 25–44. [Google Scholar]
  32. Chauhan, A.; Kaur, R.; Ghoshal, S.; Pal, A. Exploration of Circulating Tumour Cell (CTC) Biology: A Paradigm Shift in Liquid Biopsy. Indian J. Clin. Biochem. 2021, 36, 131–142. [Google Scholar] [CrossRef] [PubMed]
  33. Aceto, N.; Bardia, A.; Miyamoto, D.T.; Donaldson, M.C.; Wittner, B.S.; Spencer, J.A.; Yu, M.; Pely, A.; Engstrom, A.; Zhu, H.; et al. Circulating Tumor Cell Clusters Are Oligoclonal Precursors of Breast Cancer Metastasis. Cell 2014, 158, 1110–1122. [Google Scholar] [CrossRef] [PubMed]
  34. Amintas, S.; Bedel, A.; Moreau-Gaudry, F.; Boutin, J.; Buscail, L.; Merlio, J.-P.; Vendrely, V.; Dabernat, S.; Buscail, É. Circulating Tumor Cell Clusters: United We Stand Divided We Fall. Int. J. Mol. Sci. 2020, 21, 2653. [Google Scholar] [CrossRef] [PubMed]
  35. Cheng, Y.; Zhang, S.; Qin, L.; Zhou, J.; Shi, H.; Yang, Y.; Sun, J.; Tian, F.; Liu, C. Poly(ethylene Oxide) Concentration Gradient-Based Microfluidic Isolation of Circulating Tumor Cells. Anal. Chem. 2023, 95, 3468–3475. [Google Scholar] [CrossRef] [PubMed]
  36. Huang, Y.; Li, X.; Hou, J.; Luo, Z.; Yang, G.; Zhou, S. Conductive Nanofibers-Enhanced Microfluidic Device for the Efficient Capture and Electrical Stimulation-Triggered Rapid Release of Circulating Tumor Cells. Biosensors 2023, 13, 497. [Google Scholar] [CrossRef]
  37. Burr, R.; Edd, J.F.; Chirn, B.; Mishra, A.; Haber, D.A.; Toner, M.; Maheswaran, S. Negative-Selection Enrichment of Circulating Tumor Cells from Peripheral Blood Using the Microfluidic CTC-iChip. In Mammary Stem Cells: Methods and Protocols; Vivanco, M.D., Ed.; Springer: New York, NY, USA, 2022; pp. 309–321. [Google Scholar]
  38. Eslami-S, Z.; Cortés-Hernández, L.E.; Thomas, F.; Pantel, K.; Alix-Panabières, C. Functional analysis of circulating tumour cells: The KEY to understand the biology of the metastatic cascade. Br. J. Cancer 2022, 127, 800–810. [Google Scholar] [CrossRef]
  39. Mazard, T.; Cayrefourcq, L.; Perriard, F.; Senellart, H.; Linot, B.; de la Fouchardière, C.; Terrebonne, E.; François, E.; Obled, S.; Guimbaud, R.; et al. Clinical Relevance of Viable Circulating Tumor Cells in Patients with Metastatic Colorectal Cancer: The COLOSPOT Prospective Study. Cancers 2021, 13, 2966. [Google Scholar] [CrossRef]
  40. Zhang, X.; Xie, P.; Zhang, K.; Zhang, W. Circulating tumour cell isolation, analysis and clinical application. Cell. Oncol. 2023, 46, 533–544. [Google Scholar] [CrossRef]
  41. Budna-Tukan, J.; Świerczewska, M.; Mazel, M.; Cieślikowski, W.A.; Ida, A.; Jankowiak, A.; Antczak, A.; Nowicki, M.; Pantel, K.; Azria, D.; et al. Analysis of Circulating Tumor Cells in Patients with Non-Metastatic High-Risk Prostate Cancer before and after Radiotherapy Using Three Different Enumeration Assays. Cancers 2019, 11, 802. [Google Scholar] [CrossRef]
  42. Cayrefourcq, L.; De Roeck, A.; Garcia, C.; Stoebner, P.-E.; Fichel, F.; Garima, F.; Perriard, F.; Daures, J.-P.; Meunier, L.; Alix-Panabières, C. S100-EPISPOT: A New Tool to Detect Viable Circulating Melanoma Cells. Cells 2019, 8, 755. [Google Scholar] [CrossRef]
  43. Garrel, R.; Mazel, M.; Perriard, F.; Vinches, M.; Cayrefourcq, L.; Guigay, J.; Digue, L.; Aubry, K.; Alfonsi, M.; Delord, J.-P.; et al. Circulating Tumor Cells as a Prognostic Factor in Recurrent or Metastatic Head and Neck Squamous Cell Carcinoma: The CIRCUTEC Prospective Study. Clin. Chem. 2019, 65, 1267–1275. [Google Scholar] [CrossRef] [PubMed]
  44. de Miranda, F.S.; Barauna, V.G.; Dos Santos, L.; Costa, G.; Vassallo, P.F.; Campos, L.C.G. Properties and Application of Cell-Free DNA as a Clinical Biomarker. Int. J. Mol. Sci. 2021, 22, 9110. [Google Scholar] [CrossRef]
  45. Ramirez-Garrastacho, M.; Bajo-Santos, C.; Line, A.; Martens-Uzunova, E.S.; de la Fuente, J.M.; Moros, M.; Soekmadji, C.; Tasken, K.A.; Llorente, A. Extracellular vesicles as a source of prostate cancer biomarkers in liquid biopsies: A decade of research. Br. J. Cancer 2022, 126, 331–350. [Google Scholar] [CrossRef]
  46. Connal, S.; Cameron, J.M.; Sala, A.; Brennan, P.M.; Palmer, D.S.; Palmer, J.D.; Perlow, H.; Baker, M.J. Liquid biopsies: The future of cancer early detection. J. Transl. Med. 2023, 21, 118. [Google Scholar] [CrossRef] [PubMed]
  47. Wan, J.C.M.; Massie, C.; Garcia-Corbacho, J.; Mouliere, F.; Brenton, J.D.; Caldas, C.; Pacey, S.; Baird, R.; Rosenfeld, N. Liquid biopsies come of age: Towards implementation of circulating tumour DNA. Nat. Rev. Cancer 2017, 17, 223–238. [Google Scholar] [CrossRef]
  48. Kolenda, T.; Guglas, K.; Baranowski, D.; Sobocinska, J.; Kopczynska, M.; Teresiak, A.; Blizniak, R.; Lamperska, K. cfRNAs as biomarkers in oncology-still experimental or applied tool for personalized medicine already? Rep. Pract. Oncol. Radiother. 2020, 25, 783–792. [Google Scholar] [CrossRef] [PubMed]
  49. Zhang, C.; Sun, C.; Zhao, Y.; Wang, Q.; Guo, J.; Ye, B.; Yu, G. Overview of MicroRNAs as Diagnostic and Prognostic Biomarkers for High-Incidence Cancers in 2021. Int. J. Mol. Sci. 2022, 23, 11389. [Google Scholar] [CrossRef]
  50. Bautista-Sánchez, D.; Arriaga-Canon, C.; Pedroza-Torres, A.; De La Rosa-Velázquez, I.A.; González-Barrios, R.; Contreras-Espinosa, L.; Montiel-Manríquez, R.; Castro-Hernández, C.; Fragoso-Ontiveros, V.; Álvarez-Gómez, R.M.; et al. The Promising Role of miR-21 as a Cancer Biomarker and Its Importance in RNA-Based Therapeutics. Mol. Ther. Nucleic Acids 2020, 20, 409–420. [Google Scholar] [CrossRef]
  51. Wang, W.; Li, X.; Liu, C.; Zhang, X.; Wu, Y.; Diao, M.; Tan, S.; Huang, S.; Cheng, Y.; You, T. MicroRNA-21 as a diagnostic and prognostic biomarker of lung cancer: A systematic review and meta-analysis. Biosci. Rep. 2022, 42, BSR20211653. [Google Scholar] [CrossRef]
  52. Nguyen, H.T.; Kacimi, S.E.O.; Nguyen, T.L.; Suman, K.H.; Lemus-Martin, R.; Saleem, H.; Do, D.N. MiR-21 in the Cancers of the Digestive System and Its Potential Role as a Diagnostic, Predictive, and Therapeutic Biomarker. Biology 2021, 10, 417. [Google Scholar] [CrossRef]
  53. Ratti, M.; Lampis, A.; Ghidini, M.; Salati, M.; Mirchev, M.B.; Valeri, N.; Hahne, J.C. MicroRNAs (miRNAs) and Long Non-Coding RNAs (lncRNAs) as New Tools for Cancer Therapy: First Steps from Bench to Bedside. Target. Oncol. 2020, 15, 261–278. [Google Scholar] [CrossRef] [PubMed]
  54. Beylerli, O.; Gareev, I.; Sufianov, A.; Ilyasova, T.; Guang, Y. Long noncoding RNAs as promising biomarkers in cancer. Noncoding RNA Res. 2022, 7, 66–70. [Google Scholar] [CrossRef] [PubMed]
  55. Alcaide, M.; Cheung, M.; Hillman, J.; Rassekh, S.R.; Deyell, R.J.; Batist, G.; Karsan, A.; Wyatt, A.W.; Johnson, N.; Scott, D.W.; et al. Evaluating the quantity, quality and size distribution of cell-free DNA by multiplex droplet digital PCR. Sci. Rep. 2020, 10, 12564. [Google Scholar] [CrossRef] [PubMed]
  56. Ungerer, V.; Bronkhorst, A.J.; Holdenrieder, S. Preanalytical variables that affect the outcome of cell-free DNA measurements. Crit. Rev. Clin. Lab. Sci. 2020, 57, 484–507. [Google Scholar] [CrossRef]
  57. Meddeb, R.; Pisareva, E.; Thierry, A.R. Guidelines for the Preanalytical Conditions for Analyzing Circulating Cell-Free DNA. Clin. Chem. 2019, 65, 623–633. [Google Scholar] [CrossRef]
  58. Hu, X.; Zhang, H.; Wang, Y.; Lin, Y.; Li, Q.; Li, L.; Zeng, G.; Ou, R.; Cheng, X.; Zhang, Y.; et al. Effects of blood-processing protocols on cell-free DNA fragmentomics in plasma: Comparisons of one- and two-step centrifugations. Clin. Chim. Acta 2024, 560, 119729. [Google Scholar] [CrossRef] [PubMed]
  59. Martignano, F. Cell-Free DNA: An Overview of Sample Types and Isolation Procedures. In Cell-Free DNA as Diagnostic Markers: Methods and Protocols; Casadio, V., Salvi, S., Eds.; Springer: New York, NY, USA, 2019; pp. 13–27. [Google Scholar]
  60. Diaz, I.M.; Nocon, A.; Held, S.A.E.; Kobilay, M.; Skowasch, D.; Bronkhorst, A.J.; Ungerer, V.; Fredebohm, J.; Diehl, F.; Holdenrieder, S.; et al. Pre-Analytical Evaluation of Streck Cell-Free DNA Blood Collection Tubes for Liquid Profiling in Oncology. Diagnostics 2023, 13, 1288. [Google Scholar] [CrossRef]
  61. Ullius, A.; Provencher, E.; Voss, T. Abstract 1965: Multimodal analysis of circulating cell-free RNA (ccfRNA), cell-free DNA (ccfDNA) and genomic DNA (gDNA) from blood samples collected in PAXgene Blood ccfDNA Tubes. Cancer Res. 2020, 80, 1965. [Google Scholar] [CrossRef]
  62. Parackal, S.; Zou, D.; Day, R.; Black, M.; Guilford, P. Comparison of Roche Cell-Free DNA collection Tubes® to Streck Cell-Free DNA BCT®s for sample stability using healthy volunteers. Pract. Lab. Med. 2019, 16, e00125. [Google Scholar] [CrossRef]
  63. Sorber, L.; Zwaenepoel, K.; Jacobs, J.; De Winne, K.; Van Casteren, K.; Augustus, E.; Lardon, F.; Prenen, H.; Peeters, M.; Van Meerbeeck, J. Specialized blood collection tubes for liquid biopsy: Improving the pre-analytical conditions. Mol. Diagn. Ther. 2020, 24, 113–124. [Google Scholar] [CrossRef]
  64. Lee, H.; Park, C.; Na, W.; Park, K.H.; Shin, S. Precision cell-free DNA extraction for liquid biopsy by integrated microfluidics. NPJ Precis. Oncol. 2020, 4, 3. [Google Scholar] [CrossRef] [PubMed]
  65. van der Leest, P.; Boonstra, P.A.; ter Elst, A.; van Kempen, L.C.; Tibbesma, M.; Koopmans, J.; Miedema, A.; Tamminga, M.; Groen, H.J.M.; Reyners, A.K.L.; et al. Comparison of Circulating Cell-Free DNA Extraction Methods for Downstream Analysis in Cancer Patients. Cancers 2020, 12, 1222. [Google Scholar] [CrossRef] [PubMed]
  66. Terp, S.K.; Pedersen, I.S.; Stoico, M.P. Extraction of Cell-Free DNA: Evaluation of Efficiency, Quantity, and Quality. J. Mol. Diagn. 2024, 26, 310–319. [Google Scholar] [CrossRef] [PubMed]
  67. Tran, L.S.; Pham, H.-A.T.; Tran, V.T.T.; Tran, T.T.T.; Dang, A.-T.H.; Le, D.-T.V.; Nguyen, S.-L.; Nguyen, N.-V.; Nguyen, T.; Vo, B.T.; et al. Ultra-Deep Massively Parallel Sequencing with Unique Molecular Identifier Tagging Achieves Comparable Performance to Droplet Digital PCR for Detection and Quantification of Circulating Tumor DNA from Lung Cancer Patients. PLoS ONE 2019, 14, e0226193. [Google Scholar] [CrossRef] [PubMed]
  68. Saha, S.; Araf, Y.; Promon, S.K. Circulating tumor DNA in cancer diagnosis, monitoring, and prognosis. J. Egypt. Natl. Cancer Inst. 2022, 34, 8. [Google Scholar] [CrossRef] [PubMed]
  69. Tamkovich, S.; Tupikin, A.; Kozyakov, A.; Laktionov, P. Size and Methylation Index of Cell-Free and Cell-Surface-Bound DNA in Blood of Breast Cancer Patients in the Contest of Liquid Biopsy. Int. J. Mol. Sci. 2022, 23, 8919. [Google Scholar] [CrossRef]
  70. Markou, A. DNA Methylation Analysis of Tumor Suppressor Genes in Liquid Biopsy Components of Early Stage NSCLC: A Promising Tool for Early Detection. Clin. Epigenet. 2022, 14, 61. [Google Scholar] [CrossRef]
  71. Liu, X.; Lang, J.; Li, S.; Wang, Y.; Peng, L.; Wang, W.; Han, Y.; Qi, C.; Song, L.; Yang, S.; et al. Fragment Enrichment of Circulating Tumor DNA with Low-Frequency Mutations. Front. Genet. 2020, 11, 147. [Google Scholar] [CrossRef]
  72. Kim, J.; Hong, S.-P.; Lee, S.; Lee, W.; Lee, D.; Kim, R.; Park, Y.J.; Moon, S.; Park, K.; Cha, B.; et al. Multidimensional Fragmentomic Profiling of Cell-Free DNA Released from Patient-Derived Organoids. Human Genom. 2023, 17, 96. [Google Scholar] [CrossRef]
  73. Qi, T.; Pan, M.; Shi, H.; Wang, L.; Bai, Y.M.; Ge, Q. Cell-Free DNA Fragmentomics: The Novel Promising Biomarker. Int. J. Mol. Sci. 2023, 24, 1503. [Google Scholar] [CrossRef]
  74. Shi, X.; Guo, S.; Duan, Q.; Zhang, W.; Gao, S.; Jing, W.; Jiang, G.; Kong, X.; Li, P.; Li, Y.; et al. Detection and Characterization of Pancreatic and Biliary Tract Cancers Using Cell-Free DNA Fragmentomics. J. Exp. Clin. Cancer Res. 2024, 43, 145. [Google Scholar] [CrossRef] [PubMed]
  75. Park, S.; Kang, J.-K.; Lee, N.; Lee, S.-H.; Kim, H.-P.; Kim, S.Y.; Kim, T.-Y.; Kim, H.; Jung, H.A.; Sun, J.-M.; et al. Predicting Disease Recurrence in Limited Disease Small Cell Lung Cancer Using Cell-Free DNA-based Mutation and Fragmentome Analyses. Transl. Lung Cancer Res. 2024, 13, 280–291. [Google Scholar] [CrossRef] [PubMed]
  76. Wong, D.F.; Luo, P.; Znassi, N.; Arteaga, D.P.; Gray, D.; Danesh, A.; Han, M.; Zhao, E.Y.; Pedersen, S.; Prokopec, S.D.; et al. Integrated, Longitudinal Analysis of Cell-Free DNA in Uveal Melanoma. Cancer Res. Commun. 2023, 3, 267–280. [Google Scholar] [CrossRef] [PubMed]
  77. Sundby, R.T.; Szymanski, J.J.; Pan, A.; Jones, P.A.; Mahmood, S.Z.; Reid, O.H.; Srihari, D.; Armstrong, A.E.; Chamberlain, S.; Burgic, S.; et al. Early Detection of Malignant and Pre-Malignant Peripheral Nerve Tumors Using Cell-Free DNA Fragmentomics. medRxiv 2024. [Google Scholar] [CrossRef] [PubMed]
  78. Maansson, C.T.; Thomsen, L.S.; Meldgaard, P.; Nielsen, A.L.; Sorensen, B.S. Integration of Cell-Free DNA End Motifs and Fragment Lengths Can Identify Active Genes in Liquid Biopsies. Int. J. Mol. Sci. 2024, 25, 1243. [Google Scholar] [CrossRef]
  79. Zhou, Q.; Kang, G.; Jiang, P.; Qiao, R.; Lam, W.K.; Yu, S.C.Y.; Mary-Jane, L.; Lu, J.; Cheng, S.H.; Gai, W.; et al. Epigenetic Analysis of Cell-Free DNA by Fragmentomic Profiling. Proc. Natl. Acad. Sci. USA 2022, 119, e2209852119. [Google Scholar] [CrossRef]
  80. Stevic, I.; Buescher, G.; Ricklefs, F.L. Monitoring Therapy Efficiency in Cancer through Extracellular Vesicles. Cells 2020, 9, 130. [Google Scholar] [CrossRef]
  81. Liu, J.; Ren, L.; Li, S.; Li, W.; Zheng, X.; Yang, Y.; Fu, W.; Yi, J.; Wang, J.; Du, G. The biology, function, and applications of exosomes in cancer. Acta Pharm. Sin. B 2021, 11, 2783–2797. [Google Scholar] [CrossRef] [PubMed]
  82. Oliva, S.; D’Agostino, M.; Boccadoro, M.; Larocca, A. Clinical Applications and Future Directions of Minimal Residual Disease Testing in Multiple Myeloma. Front. Oncol. 2020, 10, 1. [Google Scholar] [CrossRef]
  83. Guo, W.; Li, Y.; Pang, W.; Shen, H. Exosomes: A Potential Therapeutic Tool Targeting Communications between Tumor Cells and Macrophages. Mol. Ther. 2020, 28, 1953–1964. [Google Scholar] [CrossRef]
  84. Li, J.; Guan, X.; Fan, Z.; Li, C.; Li, Y.; Wang, X.; Cao, W.; Liu, D.X. Non-Invasive Biomarkers for Early Detection of Breast Cancer. Cancers 2020, 12, 2767. [Google Scholar] [CrossRef] [PubMed]
  85. Deun, J.V.; Jo, A.; Li, Y.; Lin, H.K.; Weissleder, R.; Im, H.; Lee, H. Integrated Dual-Mode Chromatography to Enrich Extracellular Vesicles from Plasma. Adv. Biosyst. 2020, 4, 1900310. [Google Scholar] [CrossRef]
  86. Vestad, B.; Nyman, T.A.; Hove-Skovsgaard, M.; Stensland, M.; Hoel, H.; Trøseid, A.-M.S.; Aspelin, T.; Aass, H.C.D.; Puhka, M.; Hov, J.R.; et al. Potential Role of Plasma Extracellular Vesicles in Microbial Translocation and Cardiovascular Risk in People Living with HIV and Type 2 Diabetes. arXiv 2021. [Google Scholar] [CrossRef]
  87. Benecke, L.; Chiang, D.; Ebnoether, E.; Pfaffl, M.W.; Müller, L. Isolation and Analysis of Tumor-derived Extracellular Vesicles from Head and Neck Squamous Cell Carcinoma Plasma by Galectin-based Glycan Recognition Particles. Int. J. Oncol. 2022, 61, 133. [Google Scholar] [CrossRef]
  88. Havers, M.; Broman, A.; Lenshof, A.; Laurell, T. Advancement and Obstacles in Microfluidics-Based Isolation of Extracellular Vesicles. Anal. Bioanal. Chem. 2022, 415, 1265–1285. [Google Scholar] [CrossRef] [PubMed]
  89. Antounians, L.; Tzanetakis, A.; Pellerito, O.; Catania, V.D.; Sulistyo, A.; Montalva, L.; McVey, M.J.; Zani, A. The Regenerative Potential of Amniotic Fluid Stem Cell Extracellular Vesicles: Lessons Learned by Comparing Different Isolation Techniques. Sci. Rep. 2019, 9, 1837. [Google Scholar] [CrossRef]
  90. Charrier, H.; Beseme, O.; Michel, J.B.; Mulder, P.; Amouyel, P.; Pinet, F.; Turkieh, A. Lim Domain Binding 3 (Ldb3) Identified as a Potential Marker of Cardiac Extracellular Vesicles. Int. J. Mol. Sci. 2022, 23, 7374. [Google Scholar] [CrossRef]
  91. Serratì, S.; Palazzo, A.J.; Lapenna, A.; Mateos, H.; Mallardi, A.; Marsano, R.M.; Quarta, A.; Rosso, M.D.; Azzariti, A. Salting-Out Approach Is Worthy of Comparison with Ultracentrifugation for Extracellular Vesicle Isolation from Tumor and Healthy Models. Biomolecules 2021, 11, 1857. [Google Scholar] [CrossRef] [PubMed]
  92. Xue, V.W.; Yang, C.; Wong, S.C.C.; Cho, W.C. Proteomic Profiling in Extracellular Vesicles for Cancer Detection and Monitoring. Proteomics 2021, 21, 2000094. [Google Scholar] [CrossRef]
  93. Bryl-Górecka, P.; Olde, B.; Gidlöf, O.; Törngren, K.; Erlinge, D. Increased Expression of miR-224-5p in Circulating Extracellular Vesicles of Patients with Reduced Coronary Flow Reserve. BMC Cardiovasc. Disord. 2022, 22, 321. [Google Scholar] [CrossRef]
  94. Tesovnik, T.; Bizjan, B.J.; Šket, R.; Debeljak, M.; Battelino, T.; Kovač, J. Technological Approaches in the Analysis of Extracellular Vesicle Nucleotide Sequences. Front. Bioeng. Biotechnol. 2021, 9, 787551. [Google Scholar] [CrossRef] [PubMed]
  95. Qiu, S.; Cai, Y.; Yao, H.; Lin, C.; Xie, Y.; Tang, S.; Zhang, A. Small molecule metabolites: Discovery of biomarkers and therapeutic targets. Signal Transduct. Target. Ther. 2023, 8, 132. [Google Scholar] [CrossRef]
  96. 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] [PubMed]
  97. Lee, B.; Mahmud, I.; Marchica, J.; Dereziński, P.; Qi, F.; Wang, F.; Joshi, P.; Valerio, F.; Rivera, I.; Patel, V.; et al. Integrated RNA and Metabolite Profiling of Urine Liquid Biopsies for Prostate Cancer Biomarker Discovery. Sci. Rep. 2020, 10, 3716. [Google Scholar] [CrossRef] [PubMed]
  98. Liu, Y.; Wu, J.; Zhang, K.; Yang, Q.; Yang, J.; Cao, R.; Gu, F.; Liang, J.; Liu, Y.; Hu, Y.; et al. Application of Metabolomics by UHPLC-MS/MS in Diagnostics and Biomarker Discovery of Non-Small Cell Lung Cancer. Transl. Cancer Res. 2019, 8, 2371–2379. [Google Scholar] [CrossRef]
  99. Wei, M.; Zhao, X.; Wang, P.; Song, X.; Hu, J.; Zhong, K.; Lei, L.-L.; Xu, R.; Han, W.; Yang, M.; et al. Novel Metabolic Biomarker for Early Detection and Prognosis to the Patients with Gastric Cardia Adnocarcinoma. arXiv 2021. [Google Scholar] [CrossRef]
  100. Uchiyama, K.; Yagi, N.; Mizushima, K.; Higashimura, Y.; Hirai, Y.; Okayama, T.; Yoshida, N.; Katada, K.; Kamada, K.; Handa, O.; et al. Serum metabolomics analysis for early detection of colorectal cancer. J. Gastroenterol. 2017, 52, 677–694. [Google Scholar] [CrossRef]
  101. Geijsen, A.; van Roekel, E.H.; van Duijnhoven, F.J.B.; Achaintre, D.; Bachleitner-Hofmann, T.; Baierl, A.; Bergmann, M.M.; Boehm, J.; Bours, M.J.L.; Brenner, H.; et al. Plasma metabolites associated with colorectal cancer stage: Findings from an international consortium. Int. J. Cancer 2020, 146, 3256–3266. [Google Scholar] [CrossRef]
  102. Ren, F.; Fei, Q.; Qiu, K.; Zhang, Y.; Zhang, H.; Sun, L. Liquid Biopsy Techniques and Lung Cancer: Diagnosis, Monitoring and Evaluation. J. Exp. Clin. Cancer Res. 2024, 43, 96. [Google Scholar] [CrossRef]
  103. Lin, B.; Jiang, J.; Jia, J.M.; Zhou, X. Recent Advances in Exosomal miRNA Biosensing for Liquid Biopsy. Molecules 2022, 27, 7145. [Google Scholar] [CrossRef]
  104. Khachfe, H.H. Use of Liquid Biopsies in Gastrointestinal Cancers. World J. Gastrointest. Oncol. 2021, 13, 1210–1212. [Google Scholar] [CrossRef] [PubMed]
  105. Ding, J.; Zhao, W. The Application of Liquid Biopsy Techniques in High-Risk Population for Hepatocellular Carcinoma. Cancer Manag. Res. 2022, 14, 2735–2748. [Google Scholar] [CrossRef] [PubMed]
  106. Yan, T.; Huang, J.; Huang, S.; Ahir, B.K.; Li, L.-M.; Zhong, J.H. Advances in the Detection of Pancreatic Cancer Through Liquid Biopsy. Front. Oncol. 2021, 11, 801173. [Google Scholar] [CrossRef]
  107. Du, S.; Zhao, Y.; Lv, C.; Wei, M.; Gao, Z.; Meng, X. Applying Serum Proteins and MicroRNA as Novel Biomarkers for Early-Stage Cervical Cancer Detection. Sci. Rep. 2020, 10, 9033. [Google Scholar] [CrossRef] [PubMed]
  108. Gawel, S.H.; Jackson, L.; Jeanblanc, N.; Davis, G. Current and Future Opportunities for Liquid Biopsy of Circulating Biomarkers to Aid in Early Cancer Detection. J. Cancer Metast. Treat. 2022, 8, 26. [Google Scholar] [CrossRef]
  109. Jeffrey, G.P.; Gordon, L.G.; Hill, M.M.; Ramm, G.A. Liquid Biopsies for Hepatocellular Cancer and Their Potential in Clinical Practice. Hepatology 2020, 71, 2160–2162. [Google Scholar] [CrossRef] [PubMed]
  110. Wade, R.; Nevitt, S.; Liu, Y.; Harden, M.; Khouja, C.; Raine, G.; Churchill, R.; Dias, S. Multi-Cancer Early Detection Tests for General Population Screening: A Systematic Literature Review. medRxiv 2024. [Google Scholar] [CrossRef]
  111. Qvick, A.; Bratulic, S.; Carlsson, J.; Stenmark, B.; Karlsson, C.; Nielsen, J.; Gatto, F.; Helenius, G. Plasma Glycosaminoglycans and Cell-Free DNA to Discriminate Benign and Malignant Lung Diseases. medRxiv 2024. [Google Scholar] [CrossRef]
  112. Kan, C.M.; Pei, X.M.; Yeung, M.H.Y.; Jin, N.; Ng, S.S.M.; Tsang, H.F.; Cho, W.C.S.; Yim, A.K.; Yu, A.C.; Wong, S.C.C. Exploring the Role of Circulating Cell-Free RNA in the Development of Colorectal Cancer. Int. J. Mol. Sci. 2023, 24, 11026. [Google Scholar] [CrossRef]
  113. Shen, Y.; Shi, R.; Zhao, R.; Wang, H. Clinical Application of Liquid Biopsy in Endometrial Carcinoma. Med. Oncol. 2023, 40, 92. [Google Scholar] [CrossRef]
  114. Bagheri, A.; Khorshid, H.R.K.; Tavallaie, M.; Mowla, S.J.; Sherafatian, M.; Rashidi, M.; Zargari, M.; Boroujeni, M.E.; Hosseini, S.M. A panel of noncoding RNAs in non-small-cell lung cancer. J. Cell. Biochem. 2019, 120, 8280–8290. [Google Scholar] [CrossRef] [PubMed]
  115. Marcuello, M.; Vymetalkova, V.; Neves, R.P.L.; Duran-Sanchon, S.; Vedeld, H.M.; Tham, E.; van Dalum, G.; Flügen, G.; Garcia-Barberan, V.; Fijneman, R.J.A.; et al. Circulating biomarkers for early detection and clinical management of colorectal cancer. Mol. Asp. Med. 2019, 69, 107–122. [Google Scholar] [CrossRef] [PubMed]
  116. Galvão-Lima, L.J.; Morais, A.H.F.; Valentim, R.A.M.; Barreto, E.J.S.S. miRNAs as biomarkers for early cancer detection and their application in the development of new diagnostic tools. BioMed. Eng. Online 2021, 20, 21. [Google Scholar] [CrossRef]
  117. Preethi, K.A.; Selvakumar, S.C.; Ross, K.; Jayaraman, S.; Tusubira, D.; Sekar, D. Liquid Biopsy: Exosomal microRNAs as Novel Diagnostic and Prognostic Biomarkers in Cancer. Mol. Cancer 2022, 21, 54. [Google Scholar] [CrossRef] [PubMed]
  118. Hsu, C.-C.; Wu, Y. Recent Advances in Nanotechnology-Enabled Biosensors for Detection of Exosomes as New Cancer Liquid Biopsy. Exp. Biol. Med. 2022, 247, 2152–2172. [Google Scholar] [CrossRef]
  119. Li, S.; Yi, M.; Dong, B.; Tan, X.; Luo, S.; Wu, K. The Role of Exosomes in Liquid Biopsy for Cancer Diagnosis and Prognosis Prediction. Int. J. Cancer 2020, 148, 2640–2651. [Google Scholar] [CrossRef]
  120. Takeuchi, T.; Mori, K.; Sunayama, H.; Takano, E.; Kitayama, Y.; Shimizu, T.; Hirose, Y.; Inubushi, S.; Sasaki, R.; Tanino, H. Antibody-Conjugated Signaling Nanocavities Fabricated by Dynamic Molding for Detecting Cancers Using Small Extracellular Vesicle Markers from Tears. J. Am. Chem. Soc. 2020, 142, 6617–6624. [Google Scholar] [CrossRef] [PubMed]
  121. Matsushita, D.; Arigami, T.; Okubo, K.; Sasaki, K.; Noda, M.; Kita, Y.; Mori, S.; Uenosono, Y.; Ohtsuka, T.; Natsugoe, S. The Diagnostic and Prognostic Value of a Liquid Biopsy for Esophageal Cancer: A Systematic Review and Meta-Analysis. Cancers 2020, 12, 3070. [Google Scholar] [CrossRef] [PubMed]
  122. Lu, Y.T.; Delijani, K.; Mecum, A.; Goldkorn, A. Current Status of Liquid Biopsies for the Detection and Management of Prostate Cancer. Cancer Manag. Res. 2019, 11, 5271–5291. [Google Scholar] [CrossRef] [PubMed]
  123. Rink, M.; Schwarzenbach, H.; Vetterlein, M.W.; Soave, A. The Current Role of Circulating Biomarkers in Non-Muscle Invasive Bladder Cancer. Transl. Androl. Urol. 2019, 8, 61–75. [Google Scholar] [CrossRef] [PubMed]
  124. Lim, M.; Kim, C.-J.; Sunkara, V.; Kim, M.H.; Cho, Y.K. Liquid Biopsy in Lung Cancer: Clinical Applications of Circulating Biomarkers (CTCs and ctDNA). Micromachines 2018, 9, 100. [Google Scholar] [CrossRef] [PubMed]
  125. Martins, I.; Ribeiro, I.P.; Jorge, J.; Gonçalves, A.C.; Sarmento-Ribeiro, A.B.; Melo, J.B.; Carreira, I.M. Liquid Biopsies: Applications for Cancer Diagnosis and Monitoring. Genes 2021, 12, 349. [Google Scholar] [CrossRef] [PubMed]
  126. Lampis, A.; Ghidini, M.; Ratti, M.; Mirchev, M.; Okuducu, A.F.; Valeri, N.; Hahne, J.C. Circulating Tumour DNAs and Non-Coding RNAs as Liquid Biopsies for the Management of Colorectal Cancer Patients. Gastrointest. Disord. 2020, 2, 212–235. [Google Scholar] [CrossRef]
  127. Agashe, R.; Kurzrock, R. Circulating Tumor Cells: From the Laboratory to the Cancer Clinic. Cancers 2020, 12, 2361. [Google Scholar] [CrossRef] [PubMed]
  128. Nagayama, S.; Low, S.K.; Kiyotani, K. Precision Medicine for Colorectal Cancer with Liquid Biopsy and Immunotherapy. Cancers 2021, 13, 4803. [Google Scholar] [CrossRef] [PubMed]
  129. Sardarabadi, P.; Kojabad, A.A.; Jafari, D.; Liu, C.-H. Liquid Biopsy-Based Biosensors for MRD Detection and Treatment Monitoring in Non-Small Cell Lung Cancer (NSCLC). Biosensors 2021, 11, 394. [Google Scholar] [CrossRef] [PubMed]
  130. Stergiopoulou, D.; Markou, A.; Strati, A.; Zavridou, M.; Tzanikou, E.; Mastoraki, S.; Kallergi, G.; Georgoulias, V.; Lianidou, E. Comprehensive Liquid Biopsy Analysis as a Tool for the Early Detection of Minimal Residual Disease in Breast Cancer. Sci. Rep. 2023, 13, 1258. [Google Scholar] [CrossRef] [PubMed]
  131. Paschold, L.; Binder, M. Circulating Tumor DNA in Gastric and Gastroesophageal Junction Cancer. Curr. Oncol. 2022, 29, 1430–1441. [Google Scholar] [CrossRef]
  132. Pardini, B.; Sabo, A.A.; Birolo, G.; Calin, G.A. Noncoding RNAs in Extracellular Fluids as Cancer Biomarkers: The New Frontier of Liquid Biopsies. Cancers 2019, 11, 1170. [Google Scholar] [CrossRef]
  133. Eigėlienė, N.; Saarenheimo, J.; Jekunen, A. Potential of Liquid Biopsies for Breast Cancer Screening, Diagnosis, and Response to Treatment. Oncology 2019, 96, 115–124. [Google Scholar] [CrossRef]
  134. Yang, J.; Hu, J.; Li, Y.; Luo, W.; Liu, J.; Ye, D. Clinical Applications of Liquid Biopsy in Hepatocellular Carcinoma. Front. Oncol. 2022, 12, 781820. [Google Scholar] [CrossRef] [PubMed]
  135. Ye, S.; You, Q.; Song, S.; Wang, H.; Wang, C.; Zhu, L.; Yang, Y. Nanostructures and Nanotechnologies for the Detection of Extracellular Vesicle. Adv. Biol. 2022, 7, 2200201. [Google Scholar] [CrossRef] [PubMed]
  136. Bray, F.; Laversanne, M.; Sung, H.; Ferlay, J.; Siegel, R.L.; Soerjomataram, I.; Jemal, A. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2024, 74, 229–263. [Google Scholar] [CrossRef] [PubMed]
  137. Di Capua, D.; Bracken-Clarke, D.; Ronan, K.; Baird, A.-M.; Finn, S. The Liquid Biopsy for Lung Cancer: State of the Art, Limitations and Future Developments. Cancers 2021, 13, 3923. [Google Scholar] [CrossRef] [PubMed]
  138. Li, W.; Liu, J.B.; Hou, L.K.; Yu, F.; Zhang, J.; Wu, W.; Tang, X.M.; Sun, F.; Lu, H.M.; Deng, J.; et al. Liquid biopsy in lung cancer: Significance in diagnostics, prediction, and treatment monitoring. Mol. Cancer 2022, 21, 25. [Google Scholar] [CrossRef]
  139. Esagian, S.M.; Grigoriadou, G.; 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] [PubMed]
  140. Kolesar, J.; Peh, S.; Thomas, L.; Baburaj, G.; Mukherjee, N.; Kantamneni, R.; Lewis, S.; Pai, A.; Udupa, K.S.; Kumar An, N.; et al. Integration of liquid biopsy and pharmacogenomics for precision therapy of EGFR mutant and resistant lung cancers. Mol. Cancer 2022, 21, 61. [Google Scholar] [CrossRef]
  141. Jensen, S.G.; Epistolio, S.; Madsen, C.L.; Kyneb, M.H.; Riva, A.; Paganotti, A.; Barizzi, J.; Petersen, R.K.; Børgesen, M.; Molinari, F.; et al. A new sensitive and fast assay for the detection of EGFR mutations in liquid biopsies. PLoS ONE 2021, 16, e0253687. [Google Scholar] [CrossRef]
  142. Schmid, S.; Li, J.J.N.; Leighl, N.B. Mechanisms of osimertinib resistance and emerging treatment options. Lung Cancer 2020, 147, 123–129. [Google Scholar] [CrossRef]
  143. Del Re, M.; Crucitta, S.; Gianfilippo, G.; Passaro, A.; Petrini, I.; Restante, G.; Michelucci, A.; Fogli, S.; de Marinis, F.; Porta, C.; et al. Understanding the Mechanisms of Resistance in EGFR-Positive NSCLC: From Tissue to Liquid Biopsy to Guide Treatment Strategy. Int. J. Mol. Sci. 2019, 20, 3951. [Google Scholar] [CrossRef]
  144. Mazzitelli, C.; Santini, D.; Corradini, A.G.; Zamagni, C.; Trerè, D.; Montanaro, L.; Taffurelli, M. Liquid Biopsy in the Management of Breast Cancer Patients: Where Are We Now and Where Are We Going. Diagnostics 2023, 13, 1241. [Google Scholar] [CrossRef] [PubMed]
  145. Shegekar, T.; Vodithala, S.; Juganavar, A. The Emerging Role of Liquid Biopsies in Revolutionising Cancer Diagnosis and Therapy. Cureus 2023, 15, e43650. [Google Scholar] [CrossRef]
  146. Orrantia-Borunda, E.; Anchondo-Nuñez, P.; Acuña-Aguilar, L.E.; Gómez-Valles, F.O.; Ramírez-Valdespino, C.A. Subtypes of Breast Cancer. In Breast Cancer [Internet]; Exon Publications: Brisbane, Australia, 2022. [Google Scholar]
  147. Sanches, S.M.; Braun, A.C.; Calsavara, V.F.; Barbosa, P.N.V.P.; Chinen, L.T.D. Comparison of hormonal receptor expression and HER2 status between circulating tumor cells and breast cancer metastases. Clinics 2021, 76, e2971. [Google Scholar] [CrossRef]
  148. Papadaki, M.A.; Koutsopoulos, A.V.; Tsoulfas, P.G.; Lagoudaki, E.; Aggouraki, D.; Monastirioti, A.; Koutoulaki, C.; Apostolopoulou, C.A.; Merodoulaki, A.C.; Papadaki, C. Clinical relevance of immune checkpoints on circulating tumor cells in breast cancer. Cancers 2020, 12, 376. [Google Scholar] [CrossRef] [PubMed]
  149. Freitas, A.J.A.d.; Causin, R.L.; Varuzza, M.B.; Calfa, S.; Hidalgo Filho, C.M.T.; Komoto, T.T.; Souza, C.d.P.; Marques, M.M.C. Liquid Biopsy as a Tool for the Diagnosis, Treatment, and Monitoring of Breast Cancer. Int. J. Mol. Sci. 2022, 23, 9952. [Google Scholar] [CrossRef] [PubMed]
  150. Kim, J.W.; Lim, A.R.; You, J.Y.; Lee, J.H.; Song, S.E.; Lee, N.K.; Jung, S.P.; Cho, K.R.; Kim, C.Y.; Park, K.H. PIK3CA Mutation is Associated with Poor Response to HER2-Targeted Therapy in Breast Cancer Patients. Cancer Res. Treat. 2023, 55, 531–541. [Google Scholar] [CrossRef] [PubMed]
  151. Nakai, M.; Yamada, T.; Sekiya, K.; Sato, A.; Hankyo, M.; Kuriyama, S.; Takahashi, G.; Kurita, T.; Yanagihara, K.; Yoshida, H.; et al. PIK3CA mutation detected by liquid biopsy in patients with metastatic breast cancer. J. Nippon Med. Sch. 2021, 89, 66–71. [Google Scholar] [CrossRef]
  152. Venetis, K.; CursanSo, G.; Pescia, C.; D’Ercole, M.; Porta, F.M.; Blanco, M.C.; Frascarelli, C.; Ivanova, M.; Guerini Rocco, E.; Fusco, N. Liquid biopsy: Cell-free DNA based analysis in breast cancer. J. Liq. Biopsy 2023, 1, 100002. [Google Scholar] [CrossRef]
  153. André, 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]
  154. Alsmadi, O.; Abdel-Razeq, H.; Talab, Y.; Abdulelah, H.; Shaheen, Z.; Tbakhi, A. PIK3CA Mutational Profiling in a Patient Cohort with HR+/HER2-Advanced Metastatic Breast Cancer at a Tertiary Cancer Center. Fortune J. Health Sci. 2024, 7, 158–163. [Google Scholar] [CrossRef]
  155. Liao, H.; Huang, W.; Pei, W.; Li, H. Detection of ESR1 mutations based on liquid biopsy in estrogen receptor-positive metastatic breast cancer: Clinical impacts and prospects. Front. Oncol. 2020, 10, 587671. [Google Scholar] [CrossRef]
  156. Urso, L.; Vernaci, G.; Carlet, J.; Lo Mele, M.; Fassan, M.; Zulato, E.; Faggioni, G.; Menichetti, A.; Di Liso, E.; Griguolo, G. ESR1 gene mutation in hormone receptor-positive HER2-negative metastatic breast cancer patients: Concordance between tumor tissue and circulating tumor DNA analysis. Front. Oncol. 2021, 11, 625636. [Google Scholar] [CrossRef]
  157. Mazouji, O.; Ouhajjou, A.; Incitti, R.; Mansour, H. Updates on clinical use of liquid biopsy in colorectal cancer screening, diagnosis, follow-up, and treatment guidance. Front. Cell Dev. Biol. 2021, 9, 660924. [Google Scholar] [CrossRef]
  158. Lou, S.; Shaukat, A. Noninvasive strategies for colorectal cancer screening: Opportunities and limitations. Curr. Opin. Gastroenterol. 2021, 37, 44–51. [Google Scholar] [CrossRef]
  159. Hanna, M.; Dey, N.; Grady, W.M. Emerging tests for noninvasive colorectal cancer screening. Clin. Gastroenterol. Hepatol. 2023, 21, 604–616. [Google Scholar] [CrossRef]
  160. Tsai, W.-S.; You, J.-F.; Hung, H.-Y.; Hsieh, P.-S.; Hsieh, B.; Lenz, H.-J.; Idos, G.; Friedland, S.; Yi-Jiun Pan, J.; Shao, H.-J.; et al. Novel Circulating Tumor Cell Assay for Detection of Colorectal Adenomas and Cancer. Clin. Transl. Gastroenterol. 2019, 10, e00088. [Google Scholar] [CrossRef]
  161. Tsai, W.-S.; Hung, W.-S.; Wang, T.-M.; Liu, H.; Yang, C.-Y.; Wu, S.-M.; Hsu, H.-L.; Hsiao, Y.-C.; Tsai, H.-J.; Tseng, C.-P. Circulating tumor cell enumeration for improved screening and disease detection of patients with colorectal cancer. Biomed. J. 2021, 44, S190–S200. [Google Scholar] [CrossRef]
  162. Ding, Y.; Li, W.; Wang, K.; Xu, C.; Hao, M.; Ding, L. Perspectives of the application of liquid biopsy in colorectal cancer. BioMed Res. Int. 2020, 2020, 6843180. [Google Scholar] [CrossRef] [PubMed]
  163. Shirley, M. Epi proColon® for colorectal cancer screening: A profile of its use in the USA. Mol. Diagn. Ther. 2020, 24, 497–503. [Google Scholar] [CrossRef] [PubMed]
  164. Kamel, F.; Eltarhoni, K.; Nisar, P.; Soloviev, M. Colorectal Cancer Diagnosis: The Obstacles We Face in Determining a Non-Invasive Test and Current Advances in Biomarker Detection. Cancers 2022, 14, 1889. [Google Scholar] [CrossRef]
  165. van den Puttelaar, R.; Nascimento de Lima, P.; Knudsen, A.B.; Rutter, C.M.; Kuntz, K.M.; de Jonge, L.; Escudero, F.A.; Lieberman, D.; Zauber, A.G.; Hahn, A.I.; et al. Effectiveness and Cost-Effectiveness of Colorectal Cancer Screening with a Blood Test That Meets the Centers for Medicare & Medicaid Services Coverage Decision. Gastroenterology 2024, 167, 368–377. [Google Scholar] [CrossRef]
  166. Laugsand, E.A.; Brenne, S.S.; Skorpen, F. DNA methylation markers detected in blood, stool, urine, and tissue in colorectal cancer: A systematic review of paired samples. Int. J. Colorectal Dis. 2021, 36, 239–251. [Google Scholar] [CrossRef]
  167. Toth, J.F., III; Trivedi, M.; Gupta, S. Screening for Colorectal Cancer: The Role of Clinical Laboratories. Clin. Chem. 2024, 70, 150–164. [Google Scholar] [CrossRef]
  168. Fatemi, N.; Tierling, S.; Es, H.A.; Varkiani, M.; Mojarad, E.N.; Aghdaei, H.A.; Walter, J.; Totonchi, M. DNA methylation biomarkers in colorectal cancer: Clinical applications for precision medicine. Int. J. Cancer 2022, 151, 2068–2081. [Google Scholar] [CrossRef] [PubMed]
  169. Tao, X.-Y.; Li, Q.-Q.; Zeng, Y. Clinical application of liquid biopsy in colorectal cancer: Detection, prediction, and treatment monitoring. Mol. Cancer 2024, 23, 145. [Google Scholar] [CrossRef]
  170. Boysen, A.K.; Pallisgaard, N.; Andersen, C.S.A.; Spindler, K.-L.G. Circulating tumor DNA as a marker of minimal residual disease following local treatment of metastases from colorectal cancer. Acta Oncol. 2020, 59, 1424–1429. [Google Scholar] [CrossRef]
  171. Zhou, J.; Wang, C.; Lin, G.; Xiao, Y.; Jia, W.; Xiao, G.; Liu, Q.; Wu, B.; Wu, A.; Qiu, H.; et al. Serial Circulating Tumor DNA in Predicting and Monitoring the Effect of Neoadjuvant Chemoradiotherapy in Patients with Rectal Cancer: A Prospective Multicenter Study. Clin. Cancer Res. 2021, 27, 301–310. [Google Scholar] [CrossRef] [PubMed]
  172. Movassaghi, M.; Chung, R.; Anderson, C.B.; Stein, M.; Saenger, Y.; Faiena, I. Overcoming Immune Resistance in Prostate Cancer: Challenges and Advances. Cancers 2021, 13, 4757. [Google Scholar] [CrossRef]
  173. Gasperoni, L.; Giunta, E.F.; Montanari, D.; Masini, C.; De Giorgi, U. New-generation androgen receptor signaling inhibitors (ARSIs) in metastatic hormone-sensitive prostate cancer (mHSPC): Pharmacokinetics, drug-drug interactions (DDIs), and clinical impact. Expert Opin. Drug Metab. Toxicol. 2024, 20, 491–502. [Google Scholar] [CrossRef] [PubMed]
  174. Sperger, J.M.; Emamekhoo, H.; McKay, R.R.; Stahlfeld, C.N.; Singh, A.; Chen, X.E.; Kwak, L.; Gilsdorf, C.S.; Wolfe, S.K.; Wei, X.X.; et al. Prospective Evaluation of Clinical Outcomes Using a Multiplex Liquid Biopsy Targeting Diverse Resistance Mechanisms in Metastatic Prostate Cancer. J. Clin. Oncol. 2021, 39, 2926–2937. [Google Scholar] [CrossRef] [PubMed]
  175. Tulpule, V.; Morrison, G.J.; Falcone, M.; Quinn, D.I.; Goldkorn, A. Integration of Liquid Biopsies in Clinical Management of Metastatic Prostate Cancer. Curr. Oncol. Rep. 2022, 24, 1287–1298. [Google Scholar] [CrossRef]
  176. Ionescu, F.; Zhang, J.; Wang, L. Clinical Applications of Liquid Biopsy in Prostate Cancer: From Screening to Predictive Biomarker. Cancers 2022, 14, 1728. [Google Scholar] [CrossRef]
  177. Melnyk, J.E.; Steri, V.; Nguyen, H.G.; Hwang, Y.C.; Gordan, J.D.; Hann, B.; Feng, F.Y.; Shokat, K.M. Targeting a splicing-mediated drug resistance mechanism in prostate cancer by inhibiting transcriptional regulation by PKCβ1. Oncogene 2022, 41, 1536–1549. [Google Scholar] [CrossRef]
  178. Lu, D.; Krupa, R.; Harvey, M.; Graf, R.P.; Schreiber, N.; Barnett, E.; Carbone, E.; Jendrisak, A.; Gill, A.; Orr, S.; et al. Development of an immunofluorescent AR-V7 circulating tumor cell assay-A blood-based test for men with metastatic prostate cancer. J. Circ. Biomark. 2020, 9, 13–19. [Google Scholar] [CrossRef]
  179. Liu, H.E.; Vuppalapaty, M.; Hoerner, C.R.; Bergstrom, C.P.; Chiu, M.; Lemaire, C.; Che, J.; Kaur, A.; Dimmick, A.; Liu, S. Detecting androgen receptor (AR), AR variant 7 (AR-V7), prostate-specific membrane antigen (PSMA), and prostate-specific antigen (PSA) gene expression in CTCs and plasma exosome-derived cfRNA in patients with metastatic castration-resistant prostate cancer (mCRPC) by integrating the VTX-1 CTC isolation system with the QIAGEN AdnaTest. BMC Cancer 2024, 24, 482. [Google Scholar]
  180. Nimir, M.; Ma, Y.; Jeffreys, S.A.; Opperman, T.; Young, F.; Khan, T.; Ding, P.; Chua, W.; Balakrishnar, B.; Cooper, A.; et al. Detection of AR-V7 in Liquid Biopsies of Castrate Resistant Prostate Cancer Patients: A Comparison of AR-V7 Analysis in Circulating Tumor Cells, Circulating Tumor RNA and Exosomes. Cells 2019, 8, 688. [Google Scholar] [CrossRef]
  181. Abida, W.; Antonarakis, E.; Sartor, A.O. Management of Advanced Prostate Cancer with Germline or Somatic Homologous Recombination Repair Deficiency; UpToDate: Wellesley, MA, USA, 2023. [Google Scholar]
  182. von Werdt, A.; Brandt, L.; Schärer, O.D.; Rubin, M.A. PARP Inhibition in Prostate Cancer with Homologous Recombination Repair Alterations. JCO Precis. Oncol. 2021, 5, 1639–1649. [Google Scholar] [CrossRef]
  183. Stewart, M.D.; Merino Vega, D.; Arend, R.C.; Baden, J.F.; Barbash, O.; Beaubier, N.; Collins, G.; French, T.; Ghahramani, N.; Hinson, P.; et al. Homologous Recombination Deficiency: Concepts, Definitions, and Assays. Oncologist 2022, 27, 167–174. [Google Scholar] [CrossRef]
  184. Woodhouse, R.; Li, M.; Hughes, J.; Delfosse, D.; Skoletsky, J.; Ma, P.; Meng, W.; Dewal, N.; Milbury, C.; Clark, T.; et al. Clinical and analytical validation of FoundationOne Liquid CDx, a novel 324-Gene cfDNA-based comprehensive genomic profiling assay for cancers of solid tumor origin. PLoS ONE 2020, 15, e0237802. [Google Scholar] [CrossRef]
  185. Mekonnen, N.; Yang, H.; Shin, Y.K. Homologous recombination deficiency in ovarian, breast, colorectal, pancreatic, non-small cell lung and prostate cancers, and the mechanisms of resistance to PARP inhibitors. Front. Oncol. 2022, 12, 880643. [Google Scholar] [CrossRef]
  186. Armstrong, A.J. Predictive biomarkers in advanced prostate cancer. Clin. Adv. Hematol. Oncol. HO 2023, 6, 290–293. [Google Scholar]
  187. Vandekerkhove, G.; Giri, V.N.; Halabi, S.; McNair, C.; Hamade, K.; Bitting, R.L.; Wyatt, A.W. Toward Informed Selection and Interpretation of Clinical Genomic Tests in Prostate Cancer. JCO Precis. Oncol. 2024, 8, e2300654. [Google Scholar] [CrossRef] [PubMed]
  188. Kim, C.-J.; Dong, L.; Amend, S.R.; Cho, Y.-K.; Pienta, K.J. The role of liquid biopsies in prostate cancer management. Lab Chip 2021, 21, 3263–3288. [Google Scholar] [CrossRef] [PubMed]
  189. de Kruijff, I.E.; Sieuwerts, A.M.; Onstenk, W.; Kraan, J.; Smid, M.; Van, M.N.; van der Vlugt-Daane, M.; Oomen-de Hoop, E.; Mathijssen, R.H.J.; Lolkema, M.P.; et al. Circulating Tumor Cell Enumeration and Characterization in Metastatic Castration-Resistant Prostate Cancer Patients Treated with Cabazitaxel. Cancers 2019, 11, 1212. [Google Scholar] [CrossRef]
  190. Nagaya, N.; Nagata, M.; Lu, Y.; Kanayama, M.; Hou, Q.; Hotta, Z.-u.; China, T.; Kitamura, K.; Matsushita, K.; Isotani, S. Prostate-specific membrane antigen in circulating tumor cells is a new poor prognostic marker for castration-resistant prostate cancer. PLoS ONE 2020, 15, e0226219. [Google Scholar] [CrossRef]
  191. Yang, Y.J.; Kong, Y.Y.; Li, G.X.; Wang, Y.; Ye, D.W.; Dai, B. Phenotypes of circulating tumour cells predict time to castration resistance in metastatic castration-sensitive prostate cancer. BJU Int. 2019, 124, 258–267. [Google Scholar] [CrossRef]
  192. Shoukry, M.; Broccard, S.; Kaplan, J.; Gabriel, E. The Emerging Role of Circulating Tumor DNA in the Management of Breast Cancer. Cancers 2021, 13, 3813. [Google Scholar] [CrossRef] [PubMed]
  193. Vacante, M.; Ciuni, R.; Burzotta, F.; Biondi, A. The Liquid Biopsy in the Management of Colorectal Cancer: An Overview. Biomedicines 2020, 8, 308. [Google Scholar] [CrossRef]
  194. Palacín-Aliana, I.; García-Romero, N.; Asensi-Puig, A.; Carrión-Navarro, J.; González-Rumayor, V.; Ayuso-Sacido, Á. Clinical Utility of Liquid Biopsy-Based Actionable Mutations Detected via ddPCR. Biomedicines 2021, 9, 906. [Google Scholar] [CrossRef]
  195. Nordgård, O.; Forthun, R.B.; Lapin, M.; Grønberg, B.H.; Kalland, K.H.; Kopperud, R.K.; Thomsen, L.C.V.; Tjensvoll, K.; Gilje, B.; Gjertsen, B.T.; et al. Liquid Biopsies in Solid Cancers: Implementation in a Nordic Healthcare System. Cancers 2021, 13, 1861. [Google Scholar] [CrossRef]
  196. Goodsaid, F. The Labyrinth of Product Development and Regulatory Approvals in Liquid Biopsy Diagnostics. Clin. Transl. Sci. 2019, 12, 431–439. [Google Scholar] [CrossRef]
  197. Paracchini, L.; D’Incalci, M.; Marchini, S. Liquid Biopsy in the Clinical Management of High-Grade Serous Epithelial Ovarian Cancer—Current Use and Future Opportunities. Cancers 2021, 13, 2386. [Google Scholar] [CrossRef]
  198. Kan, C.M.; Tsang, H.F.; Pei, X.M.; Ng, S.S.M.; Yim, A.K.; Yu, A.C.; Wong, S.C.C. Enhancing Clinical Utility: Utilization of International Standards and Guidelines for Metagenomic Sequencing in Infectious Disease Diagnosis. Int. J. Mol. Sci. 2024, 25, 3333. [Google Scholar] [CrossRef]
  199. Temilola, D.O.; Wium, M.; Coulidiati, T.H.; Adeola, H.A.; Carbone, G.M.; Catapano, C.V.; Zerbini, L.F. The Prospect and Challenges to the Flow of Liquid Biopsy in Africa. Cells 2019, 8, 862. [Google Scholar] [CrossRef] [PubMed]
  200. Liebs, S.; Nonnenmacher, A.; Klauschen, F.; Keilholz, U.; Vecchione, L. Liquid Biopsy Assessment of Synchronous Malignancies: A Case Report and Review of the Literature. Esmo Open 2019, 4, e000528. [Google Scholar] [CrossRef]
  201. Wang, H.; Zhang, Y.; Zhang, H.; Cao, H.; Mao, J.; Chen, X.; Wang, L.; Zhang, N.; Luo, P.; Xue, J.; et al. Liquid Biopsy for Human Cancer: Cancer Screening, Monitoring, and Treatment. Medcomm 2024, 5, e564. [Google Scholar] [CrossRef]
  202. Mikilps-Mikgelbs, R.; Pupola, D.; Antone, E.; Kirshners, A.; Luguzis, A.; Salna, E.; Krams, A.; Ērglis, A. Liquid Biopsy—A Novel Diagnostic Tool for Management of Early-Stage Peripheral Lung Cancer. Proc. Latv. Acad. Sci. Sect. B Nat. Exact. Appl. Sci. 2022, 76, 325–332. [Google Scholar] [CrossRef]
  203. Bekaii-Saab, T.; Bridgewater, J.; Normanno, N. Practical Considerations in Screening for Genetic Alterations in Cholangiocarcinoma. Ann. Oncol. 2021, 32, 1111–1126. [Google Scholar] [CrossRef] [PubMed]
  204. Trujillo, B.; Wu, A.; Wetterskog, D.; Attard, G. Blood-Based Liquid Biopsies for Prostate Cancer: Clinical Opportunities and Challenges. Br. J. Cancer 2022, 127, 1394–1402. [Google Scholar] [CrossRef]
  205. Bao, Y.; Zhang, D.; Guo, H.; Ma, W. Beyond Blood: Advancing the Frontiers of Liquid Biopsy in Oncology and Personalized Medicine. Cancer Sci. 2024, 115, 1060–1072. [Google Scholar] [CrossRef]
  206. Bao, H.; Min, L.; Bu, F.; Wang, S.; Meng, J. Recent Advances of Liquid Biopsy: Interdisciplinary Strategies Toward Clinical Decision-making. Interdiscip. Med. 2023, 1, e20230021. [Google Scholar] [CrossRef]
Figure 1. Metastasis cascade. This figure was created with BioRender (https://biorender.com) (accessed on 10 June 2024).
Figure 1. Metastasis cascade. This figure was created with BioRender (https://biorender.com) (accessed on 10 June 2024).
Ijms 25 08594 g001
Figure 2. Biomarkers present in liquid biopsy. This figure was created with BioRender (https://biorender.com) (accessed on 10 June 2024).
Figure 2. Biomarkers present in liquid biopsy. This figure was created with BioRender (https://biorender.com) (accessed on 10 June 2024).
Ijms 25 08594 g002
Figure 3. Biomarkers in blood. This figure was created with BioRender (https://biorender.com) (accessed on 10 June 2024).
Figure 3. Biomarkers in blood. This figure was created with BioRender (https://biorender.com) (accessed on 10 June 2024).
Ijms 25 08594 g003
Figure 4. The isolation methods of CTCs. This figure was created with BioRender (https://biorender.com) (accessed on 9 June 2024).
Figure 4. The isolation methods of CTCs. This figure was created with BioRender (https://biorender.com) (accessed on 9 June 2024).
Ijms 25 08594 g004
Figure 5. The detection methods of CTCs. This figure was created with BioRender (https://biorender.com) (accessed on 9 June 2024).
Figure 5. The detection methods of CTCs. This figure was created with BioRender (https://biorender.com) (accessed on 9 June 2024).
Ijms 25 08594 g005
Table 1. The comparison between liquid biopsy and tissue biopsy.
Table 1. The comparison between liquid biopsy and tissue biopsy.
Liquid BiopsyTissue Biopsy
Samples derived from body fluid Samples derived from needle biopsy or surgical biopsy
Minimally invasiveInvasive
Preserved by preservation reagentsPreserved by paraffin embedding or cryopreservation
Can be performed after surgical resection and when there is no detectable metastatic massImpossible to be performed after surgical resection and when there is no detectable metastatic mass
Does not allow tumor histotype specification and stagingAllows histological diagnosis and staging
Easily performed at multiple or sequential time points during treatmentNot suitable for frequent monitoring
Allows real-time monitoring of diseaseSingle information point over time and space
Potential to reflect tumor heterogeneityNot suitable to reflect tumor heterogeneity
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ho, H.-Y.; Chung, K.-S.; Kan, C.-M.; Wong, S.-C. Liquid Biopsy in the Clinical Management of Cancers. Int. J. Mol. Sci. 2024, 25, 8594. https://doi.org/10.3390/ijms25168594

AMA Style

Ho H-Y, Chung K-S, Kan C-M, Wong S-C. Liquid Biopsy in the Clinical Management of Cancers. International Journal of Molecular Sciences. 2024; 25(16):8594. https://doi.org/10.3390/ijms25168594

Chicago/Turabian Style

Ho, Ho-Yin, Kei-See (Kasey) Chung, Chau-Ming Kan, and Sze-Chuen (Cesar) Wong. 2024. "Liquid Biopsy in the Clinical Management of Cancers" International Journal of Molecular Sciences 25, no. 16: 8594. https://doi.org/10.3390/ijms25168594

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