Next Article in Journal
Impact of Intratracheal Administration of Polyethylene Glycol-Coated Silver Nanoparticles on the Heart of Normotensive and Hypertensive Mice
Next Article in Special Issue
Transcriptome Profiling of Circulating Tumor Cells to Predict Clinical Outcomes in Metastatic Castration-Resistant Prostate Cancer
Previous Article in Journal
Accumulation of Dystrophin-Positive Muscle Fibers and Improvement of Neuromuscular Junctions in mdx Mouse Muscles after Bone Marrow Transplantation under Different Conditions
Previous Article in Special Issue
Clinical Validation of a Size-Based Microfluidic Device for Circulating Tumor Cell Isolation and Analysis in Renal Cell Carcinoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Liquid Biopsy in Lung Cancer: Biomarkers for the Management of Recurrence and Metastasis

1
British Columbia Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
2
Molecular Oncology Laboratory, Experimental Research Unit, School of Medicine, São Paulo State University (UNESP), Botucatu, SP 18618-687, Brazil
3
British Columbia Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
4
Department of Pathology and Laboratory Medicine, IWK Health Centre, Halifax, NS B3K 6R8, Canada
5
Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, NS B3K 6R8, Canada
6
Beatrice Hunter Cancer Research Institute, Halifax, NS B3H 4R2, Canada
7
Department of Surgery and Orthopedics, Faculty of Medicine, São Paulo State University (UNESP), Botucatu, SP 18618-687, Brazil
8
Division of Otolaryngology, Department of Surgery, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2023, 24(10), 8894; https://doi.org/10.3390/ijms24108894
Submission received: 1 May 2023 / Revised: 11 May 2023 / Accepted: 16 May 2023 / Published: 17 May 2023
(This article belongs to the Special Issue Liquid Biopsy in Cancers)

Abstract

:
Liquid biopsies have emerged as a promising tool for the detection of metastases as well as local and regional recurrence in lung cancer. Liquid biopsy tests involve analyzing a patient’s blood, urine, or other body fluids for the detection of biomarkers, including circulating tumor cells or tumor-derived DNA/RNA that have been shed into the bloodstream. Studies have shown that liquid biopsies can detect lung cancer metastases with high accuracy and sensitivity, even before they are visible on imaging scans. Such tests are valuable for early intervention and personalized treatment, aiming to improve patient outcomes. Liquid biopsies are also minimally invasive compared to traditional tissue biopsies, which require the removal of a sample of the tumor for further analysis. This makes liquid biopsies a more convenient and less risky option for patients, particularly those who are not good candidates for invasive procedures due to other medical conditions. While liquid biopsies for lung cancer metastases and relapse are still being developed and validated, they hold great promise for improving the detection and treatment of this deadly disease. Herein, we summarize available and novel approaches to liquid biopsy tests for lung cancer metastases and recurrence detection and describe their applications in clinical practice.

Graphical Abstract

1. Introduction

Lung cancer is the leading cause of cancer-related death worldwide, with an estimated 1.8 million deaths in 2020 [1]. Despite significant progress in understanding and treating lung cancer, the 5-year relative survival rate remains relatively low (25.4% from 2013 to 2019), as reported by the Surveillance, Epidemiology, and End Results (SEER) Program of the National Cancer Institute [2]. Lung cancer has a high mortality rate, mainly due to diagnosis occurring at later stages, as the time from onset to symptom presentation is largely variable, attributed to the slow-growing rate of lung tumors [3]. As a result, many lung cancer patients are diagnosed at advanced stages, when the disease has become locally invasive and/or has spread to distant organs, making it difficult to administer treatment, resulting in a poor prognosis [4].
Metastasis is a complex process by which cancer cells spread from the primary tumor to distant sites in the body, such as the bones, liver, or brain [5,6,7]. Cancer-related deaths are overwhelmingly the result of this disease progression (>90%), where metastasis leads to the impairment of vital organ functions [8,9]. The likelihood of metastasis depends on the histological type of cancer and the patient’s overall health. Approximately 30–40% of non-small-cell lung cancer (NSCLC) patients have metastatic disease at the time of diagnosis [10,11]. For small-cell lung cancer (SCLC), the incidence of metastasis at diagnosis is higher (~60%) due to the aggressive nature of this cancer type [12].
Recurrence occurs when cancer cells reappear after remission [13]. Recurrence can be especially challenging to manage, as the cancer may have become increasingly resistant to treatment over time. Several traits influence the risk of recurrence, including the type and stage of cancer and the effectiveness of the primary treatment [14,15,16]. Approximately 30–55% of patients with NSCLC develop recurrence, which proves fatal, despite multiple curative resections [14,17,18].
Treatment options for advanced lung cancer are limited, and most patients are not candidates for curative treatment. In these cases, molecular testing plays a crucial role in identifying actionable alterations that can guide the selection of targeted therapies and immunotherapies; the use of targeted therapies, such as EGFR and ALK inhibitors, has proven beneficial for improved treatment outcomes and reduced toxicity when compared to traditional chemotherapy [19,20,21,22,23]. Similarly, immunotherapies, such as PD-1 and PD-L1 inhibitors, have demonstrated significant clinical benefits in advanced lung cancer patients [24,25,26]. However, the challenge lies in obtaining sufficient tumor tissue for molecular testing in patients with advanced disease stages, as they may not be eligible for repeated biopsies, depending on the patient performance status, tumor location, or extent of disease [27]. Furthermore, repeat biopsies carry their own risk of complications [28]. In these cases, fine needle biopsies or cytology specimens are the only potential sources of tissue for molecular testing, but the limited amount of tumor material obtained from these can sometimes be insufficient for accurate testing.
Liquid biopsy is a minimally invasive procedure that involves the analysis of tumor-derived materials such as circulating tumor cells (CTCs), cell-free DNA (cfDNA), and extracellular vesicles (EVs) that can be processed from various body fluids including blood, urine, and pleural effusion [29]. Liquid biopsy is more globally applicable to aid in tumor screening, tumor staging, the characterization of intratumoral heterogeneity, the monitoring of tumor progression, and the therapy response for individualized treatment [30]. The application of liquid biopsies has proven to be a significant asset in lung cancer’s early detection and treatment [31]. In addition, liquid biopsies have the potential to significantly improve the management of patients with advanced lung cancer, by providing non-invasive and real-time monitoring of disease progression, the treatment response, and recurrence. A recent meta-analysis showed that ctDNA detection can be more sensitive to predicted tumor recurrence in lung cancer patients than CTCs, although both have potential results [32]. It has also been indicated that liquid biopsies are useful in guiding adjuvant and/or neoadjuvant treatment, predicting prognosis, and detecting molecular changes related to tumor resistance [29,33,34,35]. Furthermore, liquid biopsies show promise as an alternative by enabling a broad range of cytological and molecular evaluation methods through minimally invasive procedures [36,37].
As technology continues to evolve, liquid biopsies are slowly becoming indispensable for the clinical management of advanced lung cancer. Although previous studies have investigated liquid biopsies in lung cancer [38,39,40,41,42], our review offers novel insights by specifically highlighting their potential applications for the management of metastasis and recurrence in NSCLC. Here, we comprehensively review relevant data, contributing to the literature by thoroughly elucidating the role of liquid biopsies with a focus on clinical applications for the better management of metastatic and recurrent NSCLC, which are the most serious and poorly managed clinical complications of lung cancer leading to treatment failure and patient death.

2. Current Liquid Biopsy Samples and Biomarkers for Late-Stage Lung Cancer

Liquid biopsy for advanced-stage lung cancer typically involves the analysis of samples such as peripheral blood, urine, and cerebrospinal fluid (CSF), which contain various biomarkers, including ctDNA, CTCs, EVs, and microRNAs. Here, we provide an overview of the current state of knowledge on the use of liquid biopsies in detecting recurrence and metastasis in lung cancer, including a description of the various biomarkers and technologies used in liquid biopsies.

2.1. Sample Subtypes of Liquid Biopsies Used in Advanced-Stage Lung Cancer

The examination of body fluids helps to further determine the characteristics of lung cancer by identifying the presence of cancer cells or substrates secreted by them [43]. The levels of these molecules can vary according to different conditions, such as stress factors, tumor location, status, metastasis extent, and genome instability [44]. Therefore, these indicators serve as biomarkers, allowing detection, assessment, and diagnosis.
In the advanced stages of NSCLC, the initial clinical application for liquid biopsy was the detection of sensitizing EGFR mutations, which may be associated with an enhanced tumor response. In addition, the evaluation of acquired resistance in samples such as peripheral blood is a supplementary tool to tissue analysis used in clinical examination [33]. Currently, with the implementation of broad-based platforms such as next-generation sequencing (NGS), the additional genotyping of oncogene drivers supports the utility of plasma ctDNA, as reviewed elsewhere [45,46,47,48]; in fact, the absence of detectable alterations in plasma has been associated with a lower tumor burden [49]. However, the use of plasma is not sufficient for patients with metastasis exclusive to the brain or leptomeninges, and, for these cases, the use of CSF arises as an alternative source of ctDNA [50].
Table 1 lists examples of biomarkers that can be detected in body fluids, such as plasma, serum, sputum, bronchoalveolar lavage (BAL), pleural effusion, urine, and cerebrospinal fluid (CSF), and their applications in cancer diagnosis, prognosis, and treatment. Examples of biomarkers include ctDNA, miRNA, gene mutations, and protein signatures. Such biomarkers can help to determine resistance mechanisms, predict clinical outcomes, and monitor treatment efficacy.

2.2. Biomarker Subtypes of Liquid Biopsies Used in Advanced-Stage Lung Cancer

Several types of biomarkers have been reported in the context of liquid biopsy for advanced-stage lung cancer. These include CTCs, ctDNA, cell-free RNA (cfRNA), EVs, DNA methylation markers, and other circulating elements.

2.2.1. Extracellular Vesicles (EVs)

EVs are nano-sized lipid-bound particles that are secreted by cells into the extracellular space [73,74,75]. There are three main subtypes of EVs: microvesicles (MVs), exosomes, and apoptotic bodies, which can be differentiated based on their biogenesis, release pathways, size, content, and function [73]. EVs have emerged as promising biomarkers for liquid biopsy in lung cancer due to their ability to carry various types of molecular cargo, including proteins, lipids, DNA, RNA, and other important cellular components [76,77,78,79]. EVs are released by tumor cells and can be detected in blood, saliva, urine, and other body fluids. They have been shown to play a role in cell-to-cell communication, promoting tumor growth, angiogenesis, immune evasion, and metastasis in lung and other cancers [77,80,81]. Several studies have demonstrated the potential of EVs as liquid biopsy biomarkers for lung cancer. For example, EV-associated mutations in oncogenic driver genes, such as EGFR and ALK, have been detected in EV samples from lung cancer patients, and their presence has been correlated with the tumor mutational status and treatment response [69,82,82,83,84,85]. EV-based liquid biopsy has also been shown to be able to detect minimal residual disease (MRD) and monitor disease recurrence in lung cancer patients after surgery or other treatments, providing valuable information for personalized treatment decisions and surveillance [86,87].
In addition, studies have demonstrated that cancer cells release high levels of EVs, which can facilitate multiple steps in the metastatic cascade. EVs can promote tumor cell proliferation, migration, and invasion, and can also modulate the tumor microenvironment, promoting angiogenesis and immune evasion [88,89,90,91]. Moreover, EVs released by cancer cells can promote the formation of a pre-metastatic niche in distant organs, preparing the microenvironment for the arrival of cancer cells and facilitating their growth and colonization [90,92,93]. EVs can also facilitate the spread of cancer by supporting the survival and growth of CTCs in the bloodstream. By carrying bioactive molecules, EVs can protect CTCs from immune surveillance and apoptosis, and can also promote the formation of CTC clusters or aggregates with platelets and leukocytes, which can facilitate their survival in the bloodstream [94,95].

2.2.2. Circulating Tumor DNA (ctDNA)

ctDNA is a subset of cfDNA (circulating free DNA) that originates from tumor cells [44,96]. cfDNA refers to the fragmented DNA that is found in the bloodstream of an individual. The levels of total cfDNA are often higher in individuals with cancer compared to those without cancer, and this is thought to be due in part to the presence of ctDNA [97]. The amount of ctDNA released into the bloodstream can vary between different types of cancer, as well as between individual patients with the same type of cancer [98].
ctDNA levels are generally measured from plasma, as serum has a high likelihood of being contaminated by the release of genomic DNA from white blood cells during clotting [99]. ctDNA levels in plasma are generally low, approximately 5–10 ng/mL [100], and so the method of detection used is incredibly important. Droplet digital polymerase chain reaction (ddPCR) and NGS have greatly improved ctDNA’s detection sensitivity in recent years [101,102], and there are a large number of metrics that can be used for ctDNA analysis, including ctDNA concentration/levels, the quantitation of tumor-specific mutations in the ctDNA, and differing methylation patterns indicating epigenetic changes [103].
ctDNA-based biomarkers have been approved for clinical use in NSCLC, including EGFR mutation detection and NGS screening of a mutation panel, which includes ALK, EGFR, and KRAS [104]. However, this does not reflect the variety of recent advances in the field. ctDNA has many applications in advanced/metastatic lung cancer [105], including predicting the treatment response and survival for patients under treatment with chemotherapy regimens [106], targeted therapies [107,108], immune checkpoint inhibitors [109], and chemo-immunotherapy [110]. A recent study showed that longitudinal dynamic changes in ctDNA metrics using a machine learning model are associated with patient survival in metastatic NSCLC, with the potential utility of ctDNA for predicting treatment outcomes early in clinical trials, as ctDNA outperformed radiographic endpoints [105]. Similarly, serial ctDNA analysis has been shown to identify recurrence and distant metastasis earlier than conventional radiologic imaging [111]. Furthermore, the detection of ctDNA after the treatment of NSCLC could predict early recurrence after treatment [112], and clonal ctDNA mutations were shown to be a marker for MRD in NSCLC after surgical resection [113]. An increased number of variants have been linked to prognosis in advanced NSCLC and may predict lymph node metastasis even at the preoperative stage [114]. Therefore, ctDNA analysis shows great promise for liquid biopsy applications in lung cancer, but the challenge remains in translating findings into routine clinical practice.

2.2.3. Cell-Free RNAs (cfRNAs)

cfRNAs are detected in the bloodstream and other body fluids. Similarly to ctDNAs, a subset of these may be released from tumor cells [115,116]. There are several classes of RNA that can be found in circulation, including messenger RNAs (mRNAs) and non-coding RNAs (ncRNAs). Quantification is usually undertaken using PCR-based assays such as reverse-transcription quantitative PCR (RT-qPCR) and ddPCR, and advancements in NGS methods have been invaluable for biomarker discovery [117,118,119,120].
Within mRNAs, PD-L1 mRNA levels have been useful in predicting sensitivity to anti-PD-L1 therapy [121]. Beyond the therapy response, measuring the levels of mRNAs from saliva in combination with the presence of CTCs has been shown to discern NSCLC patients from healthy controls [122]. However, due to the presence of ribonucleases in the blood, mRNAs are generally fragmented in circulation and thus give low-quality reads when analyzed by sequencing [116].
ncRNA species have garnered significant interest due to their association with exosomes and protein complexes, which can protect them from degradation [115,116,118,123]. In particular, microRNAs (miRNAs) are of great interest as biomarkers and are the most abundant cfRNA species [118]. There have been several examples of miRNAs that show an association with metastases in NSCLC, such as miR-422-a with lymph node metastasis [124], and miR-483-5p and miR-342-5p with leptomeningeal metastasis [125]. The expression of miRNAs can, however, be highly variable between patients and requires validation [126,127].
Beyond miRNAs, ncRNAs such as long non-coding RNAs (lncRNAs), tRNA-derived fragments (tRFs), circular RNAs (circRNAs), and PIWI-interacting RNAs (piRNAs) can also be detected in liquid biopsy samples [118,123]. In lung cancer, the lncRNA metastasis-associated lung adenocarcinoma 1 (MALAT-1) is often differentially expressed in tumor samples compared to normal ones and may promote brain metastasis, which has led to multiple studies looking into its utility as a biomarker for NSCLC [128,129,130]. Although lncRNAs are one of the lowest-abundance cfRNAs in circulation, MALAT-1 is detectable in serum and may complement biomarker panels to improve NSCLC patient diagnosis and prognosis [130].
While much more is still to be learned about the utility of cfRNAs in advanced-stage disease, there has been some success with the development of the blood-based neuroendocrine neoplasms test (NETest), which has been used in the monitoring of gastrointestinal neuroendocrine tumors by profiling the gene expression of 51 mRNAs from peripheral blood [131,132]. However, there have not been any clinical tests for lung cancer cfRNAs developed yet.

2.2.4. DNA Methylation Markers

DNA methylation, which includes the dynamic addition of a methyl group to a cytosine nucleotide, converting the cytosine into a 5-methylcytosine (5mC), can greatly influence gene and downstream protein expression. Aberrant hypermethylation is a common finding within tumor tissue when compared to generally normal and healthy tissue. Global blood-based methylation biomarkers obtained from liquid biopsy material (cfDNA, ctDNA) have been difficult to determine; however, methylation profiles for lung cancer subtypes have shown robust differences between diseased and control states [133,134,135,136,137], and differences can even be detected between circulating tumor material and direct tumor tissue [138]. These profiles show remarkable differences even in early cancer stages, with certain methylation markers specific to particular cancers, whereas some are detectable pan-cancer [139,140]. Some markers have notable specificity to lung cancer metastasis [141]; however, the detection of these metastasis markers, and furthermore of recurrence, is a relatively novel and ongoing area of research [141,142,143].
The quality of methylation profiling is significantly influenced by the method of detection and analysis. While bisulfite conversion is still considered the gold standard, its complex processing can result in higher error rates. The Infinium HumanMethylation450 BeadChip array and the Human MethylationEPIC BeadChip are currently the most widely used methylation profiling platforms.
Methylation signatures are powerful cancer screening tools, where methylation scores have been shown to not only accurately classify cancer-derived samples from non-cancerous, but also predict cancer subtypes with high accuracy [144]. Especially in instances of a high cancer predisposition and the subsequent prediction of cancer onset, methylation profiling can provide an inexpensive yet efficient method for cancer management.

3. Monitoring Minimal Residual Disease (MRD)

After curative treatment, clinical follow-up must include the evaluation of MRD, which refers to the small number of cancer cells that may remain in the patient’s body after treatment, even if they cannot be detected by standard imaging tests [145]. These residual cells have the potential to grow and cause recurrence, making MRD an important tool for monitoring treatment responses and predicting outcomes [112].
While genetic profiling has greatly increased therapy success, fatal disease recurs in 30–70% of resected cases [32,146]. Lung cancer patients generally respond well to initial treatment; however, they develop resistance approximately two years later due to acquired or de novo molecular alterations, which might have been present in lower frequencies in the cells before the treatment [147,148,149,150]. Once the treatment kills the sensitive target cells, pre-existing resistant subclones are released to proliferate, inducing the emergence of a tumor population resistant to the applied therapy, i.e., MRD [147].
MRD detection is challenging, particularly in the context of lung cancer, because these tumors tend to develop chromosomal instability later in their natural history [151]. Clonal evolution and tumor heterogeneity are important factors to consider in monitoring MRD, as they can affect the accuracy of detection and the risk of recurrence. Throughout the natural history of the disease, the genomes of cancer cells accumulate changes, and they mutate and evolve in response to treatment and other environmental factors. This process, called clonal evolution, can lead to the emergence of new cancer cell subclones, leading to an increase in tumor heterogeneity, which can contribute towards treatment resistance and recurrence [147,151,152].

3.1. Clonal Evolution and Tumor Heterogeneity within MRD

As cancer grows by clonal evolution, dynamic molecular changes are acquired, generating subclonal cell variation, which can be defined as intratumoral heterogeneity (ITH). These changes include genetic, epigenetic, and gene expression alterations and may impact the antitumoral immune response. The advances in NGS and single-cell sequencing have enabled better knowledge of such alterations and their clinical implications, predicting the risk of metastasis, treatment response, and resistance [147,151]. Unfortunately, treatment resistance is common and can arise through various mechanisms, including point mutations, gene fusions, and cellular plasticity, giving rise to unpredictable phenotypes. As a result, employing multiple treatment modalities can enhance the efficacy, but combination therapies may also escalate treatment-related toxicity [151,152]. Mechanisms that cause lung adenocarcinomas with mutant EGFR to become resistant to osimertinib have been proposed. Tumors can show two or more resistance mechanisms, such as focal copy-number amplifications in MET, KRAS, and PD-L1, gene fusions of ALK, MKRN1-BRA, and ESR1-AKAP12, as well as neuroendocrine differentiation [153]. Therefore, combination therapy strategies have been strongly indicated in these cases [154].
In the context of ITH, liquid biopsies offer a range of potential applications, including early detection, the detection of MRD, the guidance of treatment decisions, the investigation of treatment efficacy and resistance, and real-time disease monitoring [40,155].

3.2. Minimal Residual Disease (MRD) and the Role of Liquid Biopsy in Detecting Lung Cancer Recurrence

Currently, the genetic profiling of solid tumors and detection of MRD in patients with lung cancer who have undergone curative treatment are primarily achieved using surgical or biopsy specimens. However, these methods have several limitations, such as invasiveness, making routine implementation challenging. Additionally, they provide only a temporally limited snapshot of the tumor and may fail to capture disease heterogeneity [156]. In contrast, liquid biopsy is a non-invasive method for longitudinally assessing MRD at any time point post-surgery [157].
Studies have shown that liquid biopsies are able to detect MRD well before the detection of recurrence by standard clinical methods [157,158,159]. A systematic review of 13 studies using ctDNA to detect MRD in NSCLC patients found that detection was achieved an average of 5.5 months prior to detection by radiography or other clinical methods [158]. One of these studies investigated perioperative ctDNA detection as a marker of MRD in 330 patients with stage I-III NSCLC with a custom panel of 769 genes. Negative perioperative ctDNA detection was more relevant in predicting recurrence-free survival than any other factor considered, including TNM staging of the extent of cancer progression [160]. A recent ten-year study of 13 breast cancer patients that used multiple biomarker detection methods derived from both CTCs and ctDNA in liquid biopsy samples found that these combined tests could uncover MRD at least four years prior to the emergence of clinically detectable metastases [161]. Another study evaluated the neoadjuvant immunotherapy efficacy (NAT) in resected NSCLC and assessed the risks for recurrence using ctDNA. During immunotherapy, ctDNA detection was highly concordant with the pathologic response, with overall accuracy of 91.67%. After three months of surgery, ctDNA was able to predict the patient’s recurrence with 83% sensitivity and 90% specificity. The recurrence prediction using ctDNA was capable of anticipating the radiographic recurrence, with a median time of 6.83 months [162].
Liquid biopsy tests for MRD can be divided into tumor-informed (requires knowledge of the tumor’s genetic profile beforehand by examining a sample of the tumor or analyzing preoperative cell-free DNA) and tumor-uninformed (intended to identify the existence of MRD without any prior knowledge of the specific molecular alterations that may be present in the tumor of an individual patient) assays [163]. Assays that use the tumor-informed approach, such as RaDaR [164], Signatera [165], and the recently tested HIFI platform [47], are personalized using the baseline genotype of the patient’s primary tumor or preoperative cfDNA and require longitudinal follow-up to probe for emergent mutations. This strategy has higher sensitivity and specificity than the tumor-uninformed approach but can have a delayed turnaround time. The tumor-uninformed approach includes assays such as DELFI [166]. With this approach, the same assay is used for each patient, which leads to a shorter turnaround but diminished specificity and sensitivity. Trials of liquid biopsy assays for MRD detection in NSCLC patients are currently underway [163].

4. Metastasis

Metastasis involves the successful completion of the metastatic cascade (Figure 1). During the metastatic cascade, biomarkers released directly from cancer cells or associated with cancer cells can be used as potential biomarkers in liquid biopsy. These biomarkers, including CTCs, ctDNA, exosomes, and specific proteins, hold potential as indicators of cancer metastasis and can be detected and analyzed in liquid biopsy samples. They offer a non-invasive and dynamic approach to assessing the presence and characteristics of metastatic tumors.

4.1. The Metastatic Cascade and Associated Biomarkers

Metastasis is a complex process by which cancer cells spread from the primary tumor to distant sites in the body [167]. In lung cancer, metastasis commonly occurs through the bloodstream or lymphatic system, leading to the development of secondary tumors in other organs, such as the brain, liver, and bones [5]. Chambers et al. [168] proposed a model called the “metastatic cascade” to describe the process of cancer dissemination. The metastatic cascade involves several steps, including the detachment of cells from the primary tumor, invasion of surrounding tissues (intravasation), spread throughout the circulatory system or lymphatic vessels (circulation), extravasation into distant tissues, formation of micrometastases, and establishment of a secondary tumor (colonization) [7,167,169,170,171].
During the metastatic cascade, a series of complex events result in the release of biomarkers into the bloodstream [172]. These biomarkers play a significant role in the biological processes involved in metastasis. For example, EVs released by cancer cells can have a multifaceted influence on endothelial cells (ECs). The interaction between tumor-derived EVs and ECs can promote tumor angiogenesis, trigger endothelial–mesenchymal transition (EMT), and disrupt the endothelial vascular barrier [91,173,174,175]. This disruption allows CTCs to cross the endothelial barrier and enter the bloodstream, facilitating the process of metastasis. Furthermore, EVs can indirectly induce tumor angiogenesis by promoting the phenotype switching of different cell types into cancer-associated fibroblasts, activating tumor-associated ECs and platelets, and remodeling the extracellular matrix [91,173]. Additionally, tumor-derived EVs released by tumor-educated platelets (TEPs) may enhance the permeability of endothelial cells by secreting adenosine triphosphate (ATP), which can modulate the dynamics of endothelial cell junctions and facilitate CTCs’ extravasation [176].
Other examples of biomarkers include platelets and neutrophils, which help CTCs to evade the immune system and survive in the bloodstream by forming clusters or aggregates with CTCs [177,178,179]. EVs contain integrins, such as αVβ5 for liver metastasis or α6β4 and α6β1 for lung metastasis, that are responsible for guiding cancer cells to specific organs, contributing to the formation of the pre-metastatic niche and influencing the site of metastatic colonization [180,181]. Proteins such as matrix metalloproteinases (MMPs) [182,183,184,185] and vascular endothelial growth factor (VEGF) facilitate angiogenesis, tumor cell invasion, immune surveillance escape, and the preparation of the pre-metastatic niche [186,187,188,189].

4.2. Applications of Liquid Biopsy in Metastatic Lung Cancer

4.2.1. Liquid Biopsy for the Early Detection of Lung Cancer Metastasis

One major role for liquid biopsy in managing lung cancer metastasis is in the context of leptomeningeal metastasis (LM), which occurs in approximately 3–5% of NSCLC and is more common in EGFR-mutant NSCLC [190,191]. Because this metastatic site is not accessible for tissue biopsy, the current gold standard for diagnosis is the presence of neoplastic cells in the CSF [192]. Several studies have reported that the CSF is a more suitable sample for liquid biopsy than serum or plasma in the context of LM and other central nervous system metastases. ctDNA in the CSF of NSCLC patients with central nervous system metastasis has a more complete genomic alteration profile than ctDNA in matched plasma samples [193]. ctDNA in the CSF of NSCLC with LM was shown to be more comprehensive for profiling driver mutations than plasma, regardless of the presence or absence of an additional extracranial metastatic site [194]. Targeted NGS of cancer-relevant genes in the CSF of NSCLC patients with central nervous system metastasis revealed that CSF had higher sensitivity for identifying actionable driver mutations than plasma [195], and CSF was reported to be a more representative liquid biopsy sample type for LM in the context of EGFR-mutant NSCLC [191]. Another study reported that cfDNA in the CSF of NSCLC with LM showed a higher variant allele frequency than matched plasma, hypothesizing that the enrichment of ctDNA in the CSF may explain the high sensitivity of CSF compared to plasma in this context, and showed that a high level of genomic instability in cfDNA in the CSF was associated with shorter overall survival. Although CSF is the most commonly used liquid biopsy medium for NSCLC with LM, obtaining CSF is invasive as it requires the use of a lumbar puncture. A recent study reported that miR-483-5p and miR-342-5p, encapsulated in exosomes in the serum, may be involved in the LM of lung cancer and could offer a non-invasive alternative to the use of CSF for predicting LM in NSCLC. Additionally, miR-330-3p levels have been shown to be higher in NSCLC patients with brain metastasis [196].
Beyond LM and other central nervous system metastases, blood is generally used for liquid biopsy in lung cancer. For example, miR-422a in the plasma demonstrated diagnostic value for lymphatic metastasis [125]; exosomal miR-210-3p, miR-193a-3p, and miR-5100 in the plasma could discriminate between those with metastasis and those without [197]; and the downregulation of exosomal miR-574-5p and upregulation of exosomal miR-328-3p and miR-423-3p were observed in the plasma of lung cancer patients with bone metastasis versus those without [198]. The concentration of circulating free DNA in plasma from a lung cancer cohort was also associated with the stage of the lung cancer and the number of metastatic sites present [199], and the presence of CTCs in advanced NSCLC was associated with poor prognosis and the existence of distant metastatic sites [200]. A bioinformatic tool called ECLIPSE was recently developed and used to analyze the subclonal architecture in ctDNA from plasma samples from the TRACERx cohort, demonstrating that subclonal expansion in the primary tumor can be measured using ctDNA and is associated with metastatic potential [201]. ECLIPSE was developed specifically to handle the low ctDNA levels common in early-stage NSCLC so that liquid biopsy can be used to identify patients at higher risk for future metastases.

4.2.2. Liquid Biopsy to Monitor Treatment Response and Identify Actionable Mutations in the Clinic

Liquid biopsies have also been shown to be effective in monitoring treatment responses in lung cancer patients, and the genetic profiles obtained from liquid biopsies tend to be consistent with those obtained from tissue biopsies. A study evaluated the concordance between liquid biopsies and tissue biopsies in detecting driver mutations in advanced non-small-cell lung cancer patients and reported a concordance rate of 85.7% [202]. Another study has reported a concordance rate of 70% to 80% between liquid biopsies and tissue biopsies in detecting EGFR mutations in non-small-cell lung cancer patients [203]. Changes in the levels of ctDNA during systemic treatments such as chemotherapy [106,204], targeted therapies [107,108], and immune checkpoint inhibitors [106,109] have been linked to treatment responses and survival times in patients with metastatic cancer. Recently, Assaf et al. showed that the use of ctDNA metrics over multiple time points can be beneficial for risk stratification and survival prediction in patients with metastatic NSCLC who receive chemo-immunotherapy combinations [105]. Such studies demonstrate the clinical applications of liquid biopsy tests for the prediction of the immunotherapy response in patients with advanced NSCLC. More recently, the surveillance of circulating biomarkers, mainly ctDNA and CTCs, has been suggested as a beneficial resource for clinicians when making therapeutic decisions during the immunotherapy of patients with advanced NSCLC [205]. A concrete example of using serial liquid biopsy to guide individualized treatment for a patient with advanced NSCLC that later progressed to bone metastasis has been reported in a recent case study. The patient was initially started on an ALK inhibitor after an ALK rearrangement was noted by fluorescence in situ hybridization (FISH) and switched over to a newly approved KRAS-p.G12C inhibitor after ctDNA analysis subsequent to disease progression revealed no ALK rearrangement but a KRAS-p.G12C mutation [206].

5. Nucleic-Acid-Based Methods for Detecting Lung Cancer in Liquid Biopsies

The most common approaches for nucleic-acid-based detection in liquid biopsy samples include multiplex-hybridization-based methods, PCR-based methods, and NGS. These approaches vary in sensitivity and in the variants that they are capable of detecting, as reviewed in our previous paper [45]. Table 2 displays the technical approaches based on their sensitivity, multiplexing capability, turnaround time (TAT), and limit of detection (LOD). Long-read sequencing can be used to analyze fragmentomic signatures of circulating DNA, and single-cell sequencing can be used to study cancer metastasis, evolution, and progression.

5.1. Multiplex-Hybridization-Based and PCR-Based Methods

Multiplex-hybridization-based and PCR-based assays are typically used to screen for variants in a predetermined gene panel. The nCounter technology is a multiplex-hybridization-based assay that has been evaluated for liquid biopsy applications in NSCLC, as this platform is capable of generating high-quality gene expression data from low-quality genomic material [201]. This platform has been shown to have similar performance to NGS [202]. A comparison between two PCR-based systems (ddPCR-BioRad and Qiagen QIAcuity Digital PCR) for the liquid biopsy analysis of cfDNA in a clinical study of NSCLC patients showed that solid digital PCR was generally more sensitive, and KRAS and EGFR mutations were detected with differing sensitivities [207].

5.2. NGS Methods

Given the large number of possible oncogene targets in advanced cancers, the wider genomic range of NGS is more applicable to novel biomarker discovery and can be comparable to hotspot mutation panels in sensitivity and cost [50,208]. Some NGS platforms for ctDNA analysis have been FDA-approved for use in clinical settings: the Guardant360 [209] and FoundationOne Liquid CDx [210] platforms can both be used to detect clinically actionable mutations in ctDNA for late-stage NSCLC cancer cases and have been recommended in lieu of smaller panels such as EGFR mutation PCR tests [50]. Targeted NGS panels such as these sequence a large but limited set of cancer-associated genes, and they have high sensitivity since they sequence genes of interest with high depth, but rare variants are not within the scope of analysis. Whole-genome or whole-exome sequencing can screen for all variants, but the tradeoff is lower sensitivity in comparison with targeted NGS due to the decreased sequencing depth, which can be prohibitive when there are low concentrations of ctDNA available for analysis [211].
Table 2. Capabilities of selected sequence-based detection methods that have been used to analyze liquid biopsy samples in the context of lung cancer. The limit of detection (LOD) for a genetic test is generally defined as the lowest mutant allele fraction (MAF) that can be detected with a given probability (usually 95%) and depends on the sample type, the sample concentration, and the variant being detected.
Table 2. Capabilities of selected sequence-based detection methods that have been used to analyze liquid biopsy samples in the context of lung cancer. The limit of detection (LOD) for a genetic test is generally defined as the lowest mutant allele fraction (MAF) that can be detected with a given probability (usually 95%) and depends on the sample type, the sample concentration, and the variant being detected.
Detection MethodMultiplexing
(Number of Markers)
Turnaround Time (TAT)Sensitivity/Limit of Detection (LOD)
Multiplex-hybridization-based methodsnanoString nCounter: 800+ target genesDependent on panel [212]Sensitivity of 95% and specificity of 82% [213]
0.02–2% MAF [214]
PCR-based methodsSmall, predetermined gene panels ~2–3 days depending on panel [72]qPCR: >10% MAF [215]
dPCR: ~0.01% MAF [216]
NGS-based methodsWES: entire exome. WGS: entire genome. Targeted panels: large number of genes (e.g., TSO500 uses 500-gene panel [217])~13 days depending on panel [72,218]<1% MAF, can be <0.1% with specialized methods [211]
Abbreviations: dPCR: digital polymerase chain reaction; qPCR: quantitative polymerase chain reaction; MAF: mutant allele fraction; WES: whole-exome sequencing; WGS: whole-genome sequencing.

5.3. Fragmentomics and Long-Read Sequencing

Fragmentomics is the analysis of the non-random fragmentation patterns of cfDNA [219], and these fragments have been observed to be promising biomarkers for detecting tissue of origin in liquid biopsy. Conventional NGS methods can have limited utility in fragmentomic analyses, as NGS tends to output reads shorter than 500 base pairs [220,221]. Some recent approaches for obtaining fragmentomic signatures using NGS include linked-read sequencing used for obtaining long-read information from short reads [222], and the DNA evaluation of fragments for early interception (DELFI) approach [223,224]. Another form of NGS, called hybrid capture sequencing, has been used to measure the ctDNA fragment length and detect minimal residual disease in stage II-IIIA NSCLC patients [225].
Long-read sequencing technologies, including Oxford Nanopore Technologies (ONT) and PacBio devices, can directly read long nucleic acid fragments and their methylation patterns [226,227,228]. This makes them well suited to fragmentomics studies and the analysis of repetitive regions of the genome. Recently, a customized workflow using ONT was developed to sequence ctDNA. This successfully detected copy number alterations in lung cancer patients [229]. PacBio technology has recently been used to assess the length and methylation scores of plasma DNA in hepatocellular carcinoma (HCC) patients. An HCC methylation score was developed to differentiate liver-derived ctDNA from other tissue DNA. Using longer DNA fragments improved the diagnostic capability of the HCC score [230].

5.4. Single-Cell Sequencing

The motivation to perform single-cell sequencing of CTCs is to obtain an up-to-date molecular profile of metastatic cancer cells, to study mechanisms of metastasis, and to characterize the cells’ progression and evolution. The low volume of DNA or RNA derived from a CTC necessitates whole-genome sequencing (WGS) or whole-transcriptome amplification (WTA) to meet the required limit of detection for sequencing [231]. A recent comparison of four WTA methods for use in the single-cell sequencing of CTCs found that multiple annealing and looping-based amplification cycles (MALBAC) followed by low-pass WGS outperformed the other three methods in coverage breadth, reproducibility, and uniformity, though none of the methods met the sensitivity or specificity required for clinical use [232]. A study using MALBAC for the single-cell sequencing of CTCs isolated from lung cancer patients found that while most genomic features were heterogeneous among all CTCs, copy number variations were consistent for CTCs derived from the same patient and could be used to discriminate between lung adenocarcinoma and SCLC [233]. Another study that performed single-cell sequencing of CTCs derived from SCLC patients observed that copy number alterations were predictive of the response to chemotherapy and that the evolutionary history of cells could be deduced from copy number alterations detected at different times over the course of treatment [234]. Facilitated by WGS and WTA methods, the single-cell sequencing of CTCs is a promising approach to monitoring the progression of metastatic lung cancer.

6. Emerging Approaches for Liquid Biopsy of Lung Cancer

Nucleic-acid-based methods to detect ctDNA and CTC or EV-derived biomarkers in liquid biopsy are well studied and used with increasing frequency in clinical settings. Here, we highlight two less common approaches to biomarker detection that show promise for NSCLC monitoring: the lung microbiome and tumor-educated platelets (TEPs).

6.1. Microbiome

The interest in understanding the link between the microbiome and the development and progression of lung cancer has increased in recent years. It is known that the microbiome plays an important role in human health and disease by modulating a host’s innate and adaptive immune system, immune responses, and metabolism, and by protecting from invading pathogens [235,236,237]. The tumor microenvironment (TME) influences cancer progression and therapy responses [238] and many recent studies have demonstrated associations between the TME and microbiota composition and responses to immune checkpoint inhibitors in cancer patients, including NSCLC patients, suggesting that modulation of the microbiota through diet, probiotics, and fecal microbiota transplantations (FMT) could improve treatment efficacy [238,239,240,241,242,243,244]. There is also a possibility that the lung microbiome change could be used as a biomarker for detecting clinical phenotypes [245]. Some studies have focused on the analysis of the tumor tissue microbiome and showed evidence of its link with metastasis [246,247], while others have focused on liquid biopsies and the analysis of the microbiome as potential markers. Epithelial brushing samples collected via bronchoscopy from patients with lung cancer, incident cancer, and those who did not develop cancer after a 10-year follow-up were analyzed through 16S ribosomal RNA gene (rDNA) sequencing. Results suggested that a shift in Veillonella, Streptococcus, Prevotella, and Paenibacillus may occur in the airways of patients with incident lung cancer months before the bronchoscopy date [245]. Emerging evidence has demonstrated that alterations of circulating microbiome DNA in blood could serve as promising non-invasive biomarkers for cancer detection, including lung cancer [236]. Microbe-derived plasma cfRNAs were consistently detected by different computational methods across lung cancer and four other cancers, and the highest bacterial abundance was found for Proteobacteria, followed by Firmicutes and Actinobacteria [248]. NGS analysis of bronchoalveolar lavage fluid showed that the bacterial diversity was lower in lung cancer than in benign lung nodules, where four species of Porphyromonas somerae, Corynebacterium accolens, Burkholderia cenocepacia, and Streptococcus mitis were enriched in lung cancer compared with benign lung nodules [249]. The same study also concluded that the abundance of the lung microbiota is closely related to the development of infiltrating adenocarcinoma. Moreover, 16S rRNA gene sequencing on malignant pleural effusion (MPE) samples and controls has shown that there are compositional differences among pleural effusions related to non-malignant, para-malignant, and malignant disease [250]. The pleural fluid of MPE-Lung and Mesothelioma was associated with Rickettsiella, Ruminococcus, Enterococcus, and Lactobacillales. Mortality in MPE-Lung was associated with enrichment in Methylobacterium, Blattabacterium, and Deinococcus.
These findings suggest the potential use of lung microbiome profiling using liquid biopsies as a promising method of diagnosis. However, more research is needed with robust cohorts of patients to understand the relationship between the microbiome and lung cancer to validate findings.

6.2. Tumor-Educated Platelets (TEPs)

Besides CTCs, other types of circulating cells, such as platelets and circulating immune cells, can have altered abundances or phenotypes due to cancer, and can also be investigated as biomarkers [251,252]. Platelets and macrophages are known to be exploited by cancer cells to support the immune evasion of tumor cells and angiogenesis. Platelets in particular are abundant in circulation, easy to isolate, and known to be altered in cancerous patients, so they present an attractive target for liquid biopsy. TEPs are defined as platelets derived from cancer patients that have distinctively altered RNA and protein content [253]. Potential mechanisms underlying the “education” of platelets include the absorption of factors derived from tumors, cell-signal-driven changes in platelets’ RNA processing, and modified platelet manufacturing by megakaryocytes. Tumor-educated platelets may influence the overexpression of additional cancer-promoting signals, including genes associated with EMT, such as TGFβ, and invasion-promoting genes such as matrix MMP-9 [254]. Metastatic NSCLC has been shown to be associated with the significant downregulation of several genes in platelets [255], and mRNA signatures have been used to detect NSCLC using TEPs [256]. A diagnostic sequencing pipeline called ThromboSeq has been able to identify characteristic changes in RNA splicing in TEPs and has been used to distinguish both early- and late-stage NSCLC patients from healthy controls [257,258]. Given the relatively high accuracy of TEP RNA-seq panels, TEPs are a promising emerging biomarker for lung cancer.

7. Conclusions

Liquid biopsy is a minimally invasive technique that has shown promise in detecting tumor biomarkers in the body fluids of patients with lung cancer. Due to its advantages, such as test repeatability and suitability for patients with poor clinical conditions, it offers an option for patients who are not suitable candidates for traditional solid tissue biopsies. Monitoring and identifying minimal residual disease in patients undergoing treatment is of great importance in clinical practice as it enables early therapeutic intervention with the potential of reducing disease recurrence.
Furthermore, significant advancements have been observed in the ability of liquid biopsies to identify specific mutations associated with lung cancer in advanced disease stages, which can facilitate individualized treatment strategies and lead to enhanced treatment responses and survival rates. However, continued research is required to improve the sensitivity and specificity of liquid biopsy tests, with the ultimate goal of enabling the detection of serious clinical complications in lung cancer, such as recurrence and distant metastases, before they become clinically evident. With ongoing technological advancements, liquid biopsy tests will become a routine part of clinical practice for lung cancer patients, providing personalized therapies and improving patient outcomes.

Author Contributions

V.G.P.S., A.F. and L.J.B. contributed equally to this work. V.G.P.S., L.J.B., P.P.R. and W.L.L. conceptualized and designed the article. All authors contributed to data collection, curation, analysis, interpretation, and writing. V.G.P.S., L.J.B., N.T., G.L.S., E.N.H., C.G., W.L.L., V.D.M. and P.P.R. were responsible for editing. V.G.P.S., L.J.B., M.E.P., N.T., J.T., K.B., A.L.S., I.W.M., M.E., R.P.d.A. and G.L.S. developed the display items. E.N.H., C.G., W.L.L., V.D.M. and P.P.R. were the principal investigators. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants from the Canadian Institutes for Health Research (FRN-143345 and FRN-183775), Coordenação de Aperfeiçoamento de Pessoal de Nível Superior/Coordination for the Improvement of Higher Education Personnel (CAPES), and the BC Cancer Foundation. A.F., M.E.P., and G.L.S. were supported by the CIHR Frederick Banting and Charles Best Canada Graduate Scholarships. V.G.P.S. and R.P.A. were supported by CAPES (V.G.S. Capes-Print Brazil, Finance code: 001; R.P.A. fellowship grant # 88887.691765/2022-00). N.T. was supported by fellowships from the University of British Columbia.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

Figures were generated using Biorender.com.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

5mC: 5-methylcytosine; ALK: anaplastic lymphoma kinase; ATP: adenosine triphosphate; BAL: bronchoalveolar fluid; cfDNA: cell-free DNA; cfRNA: cell-free RNA; circRNA: circularRNA; CSF: cerebrospinal fluid; CTC: circulating tumor cell; ctDNA: circulating tumor DNA; ddPCR: droplet digital PCR; DELFI: DNA evaluation of fragments for early interception; DNA: deoxyribonucleic acid; EC: endothelial cell; ECM: extracellular matrix; EGFR: epidermal growth factor receptor; EMT: epithelial–mesenchymal transition; EV: extracellular vesicle; FDA: Food and Drug Administration; FMT: fecal microbiota transplantations; HCC: hepatocellular carcinoma; ICIs: immune checkpoint inhibitors; ITH: intratumoral heterogeneity; KRAS: Kirsten rat sarcoma virus protein; LM: leptomeningeal metastasis; lncRNA: long non-coding RNA; MALAT-1: metastasis-associated lung adenocarcinoma 1; MALBAC: multiple annealing and looping-based amplification cycles; miRNA: microRNA; MMP9: matrix metallopeptidase 9; MPE: malignant pleural effusion; MRD: minimal residual disease; MRI: magnetic resonance imaging; MS: mass spectroscopy; MV: microvesicle; NGS: next-generation sequencing; NSCLC: non-small-cell lung cancer; ONT: Oxford Nanopore Technologies; OS: overall survival; PD-1: programmed cell death protein 1; PD-L1: programmed death ligand 1; PFS: progression-free survival; piRNA: PIWI-interacting RNA; RNA: ribonucleic acid; rRNA: ribosomal RNA; RT-qPCR: reverse-transcription quantitative polymerase chain reaction; SCLC: small-cell lung cancer; SEER: Surveillance, Epidemiology, and End Results Program of the National Cancer Institute; TEP: tumor-educated platelet; TGFβ: transforming growth factor beta; TME: tumor microenvironment; tRF: tRNA-derived fragment; VEGF: vascular endothelial growth factor; WGA: whole-genome amplification; WTA: whole-transcriptome amplification.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  2. Lung and Bronchus Cancer—Cancer Stat Facts. Available online: https://seer.cancer.gov/statfacts/html/lungb.html (accessed on 30 April 2023).
  3. Bade, B.C.; Dela Cruz, C.S. Lung Cancer 2020: Epidemiology, Etiology, and Prevention. Clin. Chest Med. 2020, 41, 1–24. [Google Scholar] [CrossRef] [PubMed]
  4. Ellis, P.M.; Vandermeer, R. Delays in the Diagnosis of Lung Cancer. J. Thorac. Dis. 2011, 3, 183–188. [Google Scholar] [CrossRef] [PubMed]
  5. Riihimäki, M.; Hemminki, A.; Fallah, M.; Thomsen, H.; Sundquist, K.; Sundquist, J.; Hemminki, K. Metastatic Sites and Survival in Lung Cancer. Lung Cancer Amst. Neth. 2014, 86, 78–84. [Google Scholar] [CrossRef] [PubMed]
  6. Popper, H.H. Progression and Metastasis of Lung Cancer. Cancer Metastasis Rev. 2016, 35, 75–91. [Google Scholar] [CrossRef]
  7. Souza, V.G.P.; de Araújo, R.P.; Santesso, M.R.; Seneda, A.L.; Minutentag, I.W.; Felix, T.F.; Hamamoto Filho, P.T.; Pewarchuk, M.E.; Brockley, L.J.; Marchi, F.A.; et al. Advances in the Molecular Landscape of Lung Cancer Brain Metastasis. Cancers 2023, 15, 722. [Google Scholar] [CrossRef]
  8. Chaffer, C.L.; Weinberg, R.A. A Perspective on Cancer Cell Metastasis. Science 2011, 331, 1559–1564. [Google Scholar] [CrossRef] [PubMed]
  9. Gupta, G.P.; Massagué, J. Cancer Metastasis: Building a Framework. Cell 2006, 127, 679–695. [Google Scholar] [CrossRef]
  10. Tamura, T.; Kurishima, K.; Nakazawa, K.; Kagohashi, K.; Ishikawa, H.; Satoh, H.; Hizawa, N. Specific Organ Metastases and Survival in Metastatic Non-Small-Cell Lung Cancer. Mol. Clin. Oncol. 2015, 3, 217–221. [Google Scholar] [CrossRef]
  11. Little, A.G.; Gay, E.G.; Gaspar, L.E.; Stewart, A.K. National Survey of Non-Small Cell Lung Cancer in the United States: Epidemiology, Pathology and Patterns of Care. Lung Cancer Amst. Neth. 2007, 57, 253–260. [Google Scholar] [CrossRef]
  12. Nakazawa, K.; Kurishima, K.; Tamura, T.; Kagohashi, K.; Ishikawa, H.; Satoh, H.; Hizawa, N. Specific Organ Metastases and Survival in Small Cell Lung Cancer. Oncol. Lett. 2012, 4, 617–620. [Google Scholar] [CrossRef]
  13. Lou, F.; Sima, C.S.; Rusch, V.W.; Jones, D.R.; Huang, J. Differences in Patterns of Recurrence in Early-Stage versus Locally Advanced Non-Small Cell Lung Cancer. Ann. Thorac. Surg. 2014, 98, 1755–1760; discussion 1760–1761. [Google Scholar] [CrossRef] [PubMed]
  14. Uramoto, H.; Tanaka, F. Recurrence after Surgery in Patients with NSCLC. Transl. Lung Cancer Res. 2014, 3, 242–249. [Google Scholar] [CrossRef] [PubMed]
  15. Luo, H.; Qiao, L.; Liang, N.; Zhang, J. Risk Factors for Recurrence in Patients with Resected N1 Non-Small Cell Lung Cancer—A Systematic Review and Meta-Analysis. J. BUON Off. J. Balk. Union Oncol. 2015, 20, 791–799. [Google Scholar]
  16. Cruz, C.; Afonso, M.; Oliveiros, B.; Pêgo, A. Recurrence and Risk Factors for Relapse in Patients with Non-Small Cell Lung Cancer Treated by Surgery with Curative Intent. Oncology 2017, 92, 347–352. [Google Scholar] [CrossRef]
  17. al-Kattan, K.; Sepsas, E.; Fountain, S.W.; Townsend, E.R. Disease Recurrence after Resection for Stage I Lung Cancer. Eur. J. Cardio-Thorac. Surg. Off. J. Eur. Assoc. Cardio-Thorac. Surg. 1997, 12, 380–384. [Google Scholar] [CrossRef]
  18. Zhu, J.; Feng, X.; Zhang, X.; Wen, Y.; Lin, P.; Rong, T.; Cai, L.; Zhang, L. Time-Varying Pattern of Postoperative Recurrence Risk of Early-Stage (T1a-T2bN0M0) Non-Small Cell Lung Cancer (NSCLC): Results of a Single-Center Study of 994 Chinese Patients. PLoS ONE 2014, 9, e106668. [Google Scholar] [CrossRef]
  19. Inoue, A.; Kobayashi, K.; Maemondo, M.; Sugawara, S.; Oizumi, S.; Isobe, H.; Gemma, A.; Harada, M.; Yoshizawa, H.; Kinoshita, I.; et al. Updated Overall Survival Results from a Randomized Phase III Trial Comparing Gefitinib with Carboplatin-Paclitaxel for Chemo-Naïve Non-Small Cell Lung Cancer with Sensitive EGFR Gene Mutations (NEJ002). Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2013, 24, 54–59. [Google Scholar] [CrossRef]
  20. Mitsudomi, T.; Morita, S.; Yatabe, Y.; Negoro, S.; Okamoto, I.; Tsurutani, J.; Seto, T.; Satouchi, M.; Tada, H.; Hirashima, T.; et al. Gefitinib versus Cisplatin plus Docetaxel in Patients with Non-Small-Cell Lung Cancer Harbouring Mutations of the Epidermal Growth Factor Receptor (WJTOG3405): An Open Label, Randomised Phase 3 Trial. Lancet Oncol. 2010, 11, 121–128. [Google Scholar] [CrossRef]
  21. Rosell, R.; Carcereny, E.; Gervais, R.; Vergnenegre, A.; Massuti, B.; Felip, E.; Palmero, R.; Garcia-Gomez, R.; Pallares, C.; Sanchez, J.M.; et al. Erlotinib versus Standard Chemotherapy as First-Line Treatment for European Patients with Advanced EGFR Mutation-Positive Non-Small-Cell Lung Cancer (EURTAC): A Multicentre, Open-Label, Randomised Phase 3 Trial. Lancet Oncol. 2012, 13, 239–246. [Google Scholar] [CrossRef]
  22. Blackhall, F.; Kim, D.-W.; Besse, B.; Nokihara, H.; Han, J.-Y.; Wilner, K.D.; Reisman, A.; Iyer, S.; Hirsh, V.; Shaw, A.T. Patient-Reported Outcomes and Quality of Life in PROFILE 1007: A Randomized Trial of Crizotinib Compared with Chemotherapy in Previously Treated Patients with ALK-Positive Advanced Non-Small-Cell Lung Cancer. J. Thorac. Oncol. Off. Publ. Int. Assoc. Study Lung Cancer 2014, 9, 1625–1633. [Google Scholar] [CrossRef]
  23. Shaw, A.T.; Kim, D.-W.; Nakagawa, K.; Seto, T.; Crinó, L.; Ahn, M.-J.; De Pas, T.; Besse, B.; Solomon, B.J.; Blackhall, F.; et al. Crizotinib versus Chemotherapy in Advanced ALK-Positive Lung Cancer. N. Engl. J. Med. 2013, 368, 2385–2394. [Google Scholar] [CrossRef] [PubMed]
  24. Davies, M.J. PD-1/PD-L1 Inhibitors for Non-Small Cell Lung Cancer: Incorporating Care Step Pathways for Effective Side-Effect Management. J. Adv. Pract. Oncol. 2019, 10, 21–35. [Google Scholar] [CrossRef] [PubMed]
  25. Herbst, R.S.; Baas, P.; Kim, D.-W.; Felip, E.; Pérez-Gracia, J.L.; Han, J.-Y.; Molina, J.; Kim, J.-H.; Arvis, C.D.; Ahn, M.-J.; et al. Pembrolizumab versus Docetaxel for Previously Treated, PD-L1-Positive, Advanced Non-Small-Cell Lung Cancer (KEYNOTE-010): A Randomised Controlled Trial. Lancet Lond. Engl. 2016, 387, 1540–1550. [Google Scholar] [CrossRef]
  26. Wang, L.; Ma, Q.; Yao, R.; Liu, J. Current Status and Development of Anti-PD-1/PD-L1 Immunotherapy for Lung Cancer. Int. Immunopharmacol. 2020, 79, 106088. [Google Scholar] [CrossRef]
  27. Chouaid, C.; Dujon, C.; Do, P.; Monnet, I.; Madroszyk, A.; Le Caer, H.; Auliac, J.B.; Berard, H.; Thomas, P.; Lena, H.; et al. Feasibility and Clinical Impact of Re-Biopsy in Advanced Non Small-Cell Lung Cancer: A Prospective Multicenter Study in a Real-World Setting (GFPC Study 12-01). Lung Cancer Amst. Neth. 2014, 86, 170–173. [Google Scholar] [CrossRef] [PubMed]
  28. Fintelmann, F.J.; Troschel, F.M.; Kuklinski, M.W.; McDermott, S.; Petranovic, M.; Digumarthy, S.R.; Sharma, A.; Troschel, A.S.; Price, M.C.; Hariri, L.P.; et al. Safety and Success of Repeat Lung Needle Biopsies in Patients with Epidermal Growth Factor Receptor-Mutant Lung Cancer. Oncologist 2019, 24, 1570–1576. [Google Scholar] [CrossRef]
  29. 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]
  30. Lone, S.N.; Nisar, S.; Masoodi, T.; Singh, M.; Rizwan, A.; Hashem, S.; El-Rifai, W.; Bedognetti, D.; Batra, S.K.; Haris, M.; et al. Liquid Biopsy: A Step Closer to Transform Diagnosis, Prognosis and Future of Cancer Treatments. Mol. Cancer 2022, 21, 79. [Google Scholar] [CrossRef]
  31. Ho, H.-L.; Jiang, Y.; Chiang, C.-L.; Karwowska, S.; Yerram, R.; Sharma, K.; Scudder, S.; Chiu, C.-H.; Tsai, C.-M.; Palma, J.F.; et al. Efficacy of Liquid Biopsy for Disease Monitoring and Early Prediction of Tumor Progression in EGFR Mutation-Positive Non-Small Cell Lung Cancer. PLoS ONE 2022, 17, e0267362. [Google Scholar] [CrossRef]
  32. Liang, H.; Huang, J.; Wang, B.; Liu, Z.; He, J.; Liang, W. The Role of Liquid Biopsy in Predicting Post-Operative Recurrence of Non-Small Cell Lung Cancer. J. Thorac. Dis. 2018, 10, S838–S845. [Google Scholar] [CrossRef]
  33. Bonanno, L.; Dal Maso, A.; Pavan, A.; Zulato, E.; Calvetti, L.; Pasello, G.; Guarneri, V.; Conte, P.; Indraccolo, S. Liquid Biopsy and Non-Small Cell Lung Cancer: Are We Looking at the Tip of the Iceberg? Br. J. Cancer 2022, 127, 383–393. [Google Scholar] [CrossRef] [PubMed]
  34. Fernandes, M.G.O.; Sousa, C.; Pereira Reis, J.; Cruz-Martins, N.; Souto Moura, C.; Guimarães, S.; Justino, A.; Pina, M.J.; Magalhães, A.; Queiroga, H.; et al. Liquid Biopsy for Disease Monitoring in Non-Small Cell Lung Cancer: The Link between Biology and the Clinic. Cells 2021, 10, 1912. [Google Scholar] [CrossRef]
  35. 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]
  36. 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]
  37. Bai, Y.; Zhao, H. Liquid Biopsy in Tumors: Opportunities and Challenges. Ann. Transl. Med. 2018, 6, S89. [Google Scholar] [CrossRef] [PubMed]
  38. Yang, Y.; Liu, H.; Chen, Y.; Xiao, N.; Zheng, Z.; Liu, H.; Wan, J. Liquid Biopsy on the Horizon in Immunotherapy of Non-Small Cell Lung Cancer: Current Status, Challenges, and Perspectives. Cell Death Dis. 2023, 14, 230. [Google Scholar] [CrossRef]
  39. Zhu, W.; Love, K.; Gray, S.W.; Raz, D.J. Liquid Biopsy Screening for Early Detection of Lung Cancer: Current State and Future Directions. Clin. Lung Cancer 2023, 24, 209–217. [Google Scholar] [CrossRef] [PubMed]
  40. Tomasik, B.; Skrzypski, M.; Bieńkowski, M.; Dziadziuszko, R.; Jassem, J. Current and Future Applications of Liquid Biopsy in Non-Small-Cell Lung Cancer-a Narrative Review. Transl. Lung Cancer Res. 2023, 12, 594–614. [Google Scholar] [CrossRef]
  41. Melichar, B. Biomarkers in the Management of Lung Cancer: Changing the Practice of Thoracic Oncology. Clin. Chem. Lab. Med. 2023, 61, 906–920. [Google Scholar] [CrossRef]
  42. Khan, S.R.; Scheffler, M.; Soomar, S.M.; Rashid, Y.A.; Moosajee, M.; Ahmad, A.; Raza, A.; Uddin, S. Role of Circulating-Tumor DNA in the Early-Stage Non-Small Cell Lung Carcinoma as a Predictive Biomarker. Pathol. Res. Pract. 2023, 245, 154455. [Google Scholar] [CrossRef]
  43. Lianidou, E.; Pantel, K. Liquid Biopsies. Genes. Chromosomes Cancer 2019, 58, 219–232. [Google Scholar] [CrossRef]
  44. 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]
  45. Brockley, L.J.; Souza, V.G.P.; Forder, A.; Pewarchuk, M.E.; Erkan, M.; Telkar, N.; Benard, K.; Trejo, J.; Stewart, M.D.; Stewart, G.L.; et al. Sequence-Based Platforms for Discovering Biomarkers in Liquid Biopsy of Non-Small-Cell Lung Cancer. Cancers 2023, 15, 2275. [Google Scholar] [CrossRef]
  46. Fernandes, M.G.O.; Cruz-Martins, N.; Souto Moura, C.; Guimarães, S.; Pereira Reis, J.; Justino, A.; Pina, M.J.; Magalhães, A.; Queiroga, H.; Machado, J.C.; et al. Clinical Application of Next-Generation Sequencing of Plasma Cell-Free DNA for Genotyping Untreated Advanced Non-Small Cell Lung Cancer. Cancers 2021, 13, 2707. [Google Scholar] [CrossRef]
  47. Li, Y.; Jiang, G.; Wu, W.; Yang, H.; Jin, Y.; Wu, M.; Liu, W.; Yang, A.; Chervova, O.; Zhang, S.; et al. Multi-Omics Integrated Circulating Cell-Free DNA Genomic Signatures Enhanced the Diagnostic Performance of Early-Stage Lung Cancer and Postoperative Minimal Residual Disease. eBioMedicine 2023, 91, 104553. [Google Scholar] [CrossRef]
  48. Aggarwal, C.; Rolfo, C.D.; Oxnard, G.R.; Gray, J.E.; Sholl, L.M.; Gandara, D.R. Strategies for the Successful Implementation of Plasma-Based NSCLC Genotyping in Clinical Practice. Nat. Rev. Clin. Oncol. 2021, 18, 56–62. [Google Scholar] [CrossRef]
  49. Mack, P.C.; Miao, J.; Redman, M.W.; Moon, J.; Goldberg, S.B.; Herbst, R.S.; Melnick, M.A.; Walther, Z.; Hirsch, F.R.; Politi, K.; et al. Circulating Tumor DNA Kinetics Predict Progression-Free and Overall Survival in EGFR TKI-Treated Patients with EGFR-Mutant NSCLC (SWOG S1403). Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2022, 28, 3752–3760. [Google Scholar] [CrossRef] [PubMed]
  50. Rolfo, C.; Mack, P.; Scagliotti, G.V.; Aggarwal, C.; Arcila, M.E.; Barlesi, F.; Bivona, T.; Diehn, M.; Dive, C.; Dziadziuszko, R.; et al. Liquid Biopsy for Advanced NSCLC: A Consensus Statement From the International Association for the Study of Lung Cancer. J. Thorac. Oncol. Off. Publ. Int. Assoc. Study Lung Cancer 2021, 16, 1647–1662. [Google Scholar] [CrossRef] [PubMed]
  51. Liu, Q.; Yu, Z.; Yuan, S.; Xie, W.; Li, C.; Hu, Z.; Xiang, Y.; Wu, N.; Wu, L.; Bai, L.; et al. Circulating Exosomal MicroRNAs as Prognostic Biomarkers for Non-Small-Cell Lung Cancer. Oncotarget 2017, 8, 13048–13058. [Google Scholar] [CrossRef] [PubMed]
  52. Hallermayr, A.; Benet-Pagès, A.; Steinke-Lange, V.; Mansmann, U.; Rentsch, M.; Holinski-Feder, E.; Pickl, J.M.A. Liquid Biopsy Hotspot Variant Assays: Analytical Validation for Application in Residual Disease Detection and Treatment Monitoring. Clin. Chem. 2021, 67, 1483–1491. [Google Scholar] [CrossRef] [PubMed]
  53. Taguchi, A.; Politi, K.; Pitteri, S.J.; Lockwood, W.W.; Faça, V.M.; Kelly-Spratt, K.; Wong, C.-H.; Zhang, Q.; Chin, A.; Park, K.-S.; et al. Lung Cancer Signatures in Plasma Based on Proteome Profiling of Mouse Tumor Models. Cancer Cell 2011, 20, 289–299. [Google Scholar] [CrossRef]
  54. Chen, Q.; Zhu, C.; Jin, Y.; Si, X.; Jiao, W.; He, W.; Mao, W.; Li, M.; Luo, G. Plasma Long Non-Coding RNA RP11-438N5.3 as a Novel Biomarker for Non-Small Cell Lung Cancer. Cancer Manag. Res. 2020, 12, 1513–1521. [Google Scholar] [CrossRef]
  55. Giallombardo, M.; Jorge Chacartegui, J.; Reclusa, P.; Van Meerbeeck, J.P.; Alessandro, R.; Peeters, M.; Pauwels, P.; Rolfo, C.D. Follow up Analysis by Exosomal MiRNAs in EGFR Mutated Non-Small Cell Lung Cancer (NSCLC) Patients during Osimertinib (AZD9291) Treatment: A Potential Prognostic Biomarker Tool. J. Clin. Oncol. 2016, 34, e23035. [Google Scholar] [CrossRef]
  56. Hori, S.; Nishiumi, S.; Kobayashi, K.; Shinohara, M.; Hatakeyama, Y.; Kotani, Y.; Hatano, N.; Maniwa, Y.; Nishio, W.; Bamba, T.; et al. A Metabolomic Approach to Lung Cancer. Lung Cancer Amst. Neth. 2011, 74, 284–292. [Google Scholar] [CrossRef] [PubMed]
  57. Caponnetto, S.; Iannantuono, G.M.; Barchiesi, G.; Magri, V.; Gelibter, A.; Cortesi, E. Prolactin as a Potential Early Predictive Factor in Metastatic Non-Small Cell Lung Cancer Patients Treated with Nivolumab. Oncology 2017, 93, 62–66. [Google Scholar] [CrossRef] [PubMed]
  58. Duréndez-Sáez, E.; Torres-Martinez, S.; Calabuig-Fariñas, S.; Meri-Abad, M.; Ferrero-Gimeno, M.; Camps, C. Exosomal MicroRNAs in Non-Small Cell Lung Cancer. Transl. Cancer Res. 2021, 10, 3128–3139. [Google Scholar] [CrossRef]
  59. Wang, C.-F.; Peng, S.-J.; Liu, R.-Q.; Yu, Y.-J.; Ge, Q.-M.; Liang, R.-B.; Li, Q.-Y.; Li, B.; Shao, Y. The Combination of CA125 and NSE Is Useful for Predicting Liver Metastasis of Lung Cancer. Dis. Mark. 2020, 2020, 8850873. [Google Scholar] [CrossRef]
  60. Lou, E.; Johnson, M.; Sima, C.; Gonzalez-Espinoza, R.; Fleisher, M.; Kris, M.G.; Azzoli, C.G. Serum Biomarkers for Assessing Histology and Outcomes in Patients with Metastatic Lung Cancer. Cancer Biomark. Sect. Dis. Mark. 2014, 14, 207–214. [Google Scholar] [CrossRef]
  61. Varella-Garcia, M.; Schulte, A.P.; Wolf, H.J.; Feser, W.J.; Zeng, C.; Braudrick, S.; Yin, X.; Hirsch, F.R.; Kennedy, T.C.; Keith, R.L.; et al. The Detection of Chromosomal Aneusomy by Fluorescence in Situ Hybridization in Sputum Predicts Lung Cancer Incidence. Cancer Prev. Res. Phila. Pa 2010, 3, 447–453. [Google Scholar] [CrossRef]
  62. Guo, T.; Hu, C.; Qin, L.; Yang, H.; Gu, Q.; Cao, L.; Deng, P.; Han-Zhang, H.; Li, B.; Ye, J.; et al. Sputum Supernatant as an Alternative Source for Liquid Biopsy in Patients with Advanced Lung Cancer. J. Clin. Oncol. 2020, 38, e21617. [Google Scholar] [CrossRef]
  63. Xie, X.; Wu, J.; Guo, B.; Wang, L.; Deng, H.; Lin, X.; Liu, M.; Qin, Y.; Luo, W.; Yang, Y.; et al. Comprehensive Characterization Reveals Sputum Supernatant as a Valuable Alternative Liquid Biopsy for Genome Profiling in Advanced Non-Small Cell Lung Cancer. Respir. Res. 2022, 23, 175. [Google Scholar] [CrossRef] [PubMed]
  64. Qin, L.; Guo, T.; Yang, H.; Deng, P.; Gu, Q.; Liu, C.; Wu, M.; Lizaso, A.; Li, B.; Zhang, S.; et al. The Utility of Sputum Supernatant as an Alternative Liquid Biopsy Specimen for Next-Generation Sequencing-Based Somatic Variation Profiling. Ann. Transl. Med. 2022, 10, 462. [Google Scholar] [CrossRef]
  65. Rennard, S.I. Bronchoalveolar Lavage in the Assessment of Primary and Metastatic Lung Cancer. Respir. Int. Rev. Thorac. Dis. 1992, 59 (Suppl. 1), 41–43. [Google Scholar] [CrossRef]
  66. Matthiesen, R. MS-Based Biomarker Discovery in Bronchoalveolar Lavage Fluid for Lung Cancer. Proteomics Clin. Appl. 2020, 14, e1900077. [Google Scholar] [CrossRef]
  67. Porcel, J.M.; Esquerda, A.; Martínez-Alonso, M.; Bielsa, S.; Salud, A. Identifying Thoracic Malignancies Through Pleural Fluid Biomarkers: A Predictive Multivariate Model. Medicine 2016, 95, e3044. [Google Scholar] [CrossRef] [PubMed]
  68. Jany, B.; Welte, T. Pleural Effusion in Adults-Etiology, Diagnosis, and Treatment. Dtsch. Arztebl. Int. 2019, 116, 377–386. [Google Scholar] [CrossRef] [PubMed]
  69. Lee, J.S.; Hur, J.Y.; Kim, I.A.; Kim, H.J.; Choi, C.M.; Lee, J.C.; Kim, W.S.; Lee, K.Y. Liquid Biopsy Using the Supernatant of a Pleural Effusion for EGFR Genotyping in Pulmonary Adenocarcinoma Patients: A Comparison between Cell-Free DNA and Extracellular Vesicle-Derived DNA. BMC Cancer 2018, 18, 1236. [Google Scholar] [CrossRef]
  70. Oshi, M.; Murthy, V.; Takahashi, H.; Huyser, M.; Okano, M.; Tokumaru, Y.; Rashid, O.M.; Matsuyama, R.; Endo, I.; Takabe, K. Urine as a Source of Liquid Biopsy for Cancer. Cancers 2021, 13, 2652. [Google Scholar] [CrossRef]
  71. Huang, W.-L.; Chen, Y.-L.; Yang, S.-C.; Ho, C.-L.; Wei, F.; Wong, D.T.; Su, W.-C.; Lin, C.-C. Liquid Biopsy Genotyping in Lung Cancer: Ready for Clinical Utility? Oncotarget 2017, 8, 18590–18608. [Google Scholar] [CrossRef]
  72. Rolfo, C.; Mack, P.C.; Scagliotti, G.V.; Baas, P.; Barlesi, F.; Bivona, T.G.; Herbst, R.S.; Mok, T.S.; Peled, N.; Pirker, R.; et al. Liquid Biopsy for Advanced Non-Small Cell Lung Cancer (NSCLC): A Statement Paper from the IASLC. J. Thorac. Oncol. Off. Publ. Int. Assoc. Study Lung Cancer 2018, 13, 1248–1268. [Google Scholar] [CrossRef] [PubMed]
  73. Doyle, L.M.; Wang, M.Z. Overview of Extracellular Vesicles, Their Origin, Composition, Purpose, and Methods for Exosome Isolation and Analysis. Cells 2019, 8, 727. [Google Scholar] [CrossRef] [PubMed]
  74. Zaborowski, M.P.; Balaj, L.; Breakefield, X.O.; Lai, C.P. Extracellular Vesicles: Composition, Biological Relevance, and Methods of Study. Bioscience 2015, 65, 783–797. [Google Scholar] [CrossRef] [PubMed]
  75. Yáñez-Mó, M.; Siljander, P.R.-M.; Andreu, Z.; Zavec, A.B.; Borràs, F.E.; Buzas, E.I.; Buzas, K.; Casal, E.; Cappello, F.; Carvalho, J.; et al. Biological Properties of Extracellular Vesicles and Their Physiological Functions. J. Extracell. Vesicles 2015, 4, 27066. [Google Scholar] [CrossRef]
  76. Sandúa, A.; Alegre, E.; González, Á. Exosomes in Lung Cancer: Actors and Heralds of Tumor Development. Cancers 2021, 13, 4330. [Google Scholar] [CrossRef]
  77. Rizwan, M.N.; Ma, Y.; Nenkov, M.; Jin, L.; Schröder, D.C.; Westermann, M.; Gaßler, N.; Chen, Y. Tumor-Derived Exosomes: Key Players in Non-Small Cell Lung Cancer Metastasis and Their Implication for Targeted Therapy. Mol. Carcinog. 2022, 61, 269–280. [Google Scholar] [CrossRef]
  78. Li, M.-Y.; Liu, L.-Z.; Dong, M. Progress on Pivotal Role and Application of Exosome in Lung Cancer Carcinogenesis, Diagnosis, Therapy and Prognosis. Mol. Cancer 2021, 20, 22. [Google Scholar] [CrossRef]
  79. Xu, K.; Zhang, C.; Du, T.; Gabriel, A.N.A.; Wang, X.; Li, X.; Sun, L.; Wang, N.; Jiang, X.; Zhang, Y. Progress of Exosomes in the Diagnosis and Treatment of Lung Cancer. Biomed. Pharmacother. Biomedecine Pharmacother. 2021, 134, 111111. [Google Scholar] [CrossRef]
  80. Kalluri, R.; LeBleu, V.S. The Biology, Function, and Biomedical Applications of Exosomes. Science 2020, 367, eaau6977. [Google Scholar] [CrossRef]
  81. Forder, A.; Hsing, C.-Y.; Trejo Vazquez, J.; Garnis, C. Emerging Role of Extracellular Vesicles and Cellular Communication in Metastasis. Cells 2021, 10, 3429. [Google Scholar] [CrossRef]
  82. Kwok, H.-H.; Ning, Z.; Chong, P.W.-C.; Wan, T.S.-K.; Ng, M.H.-L.; Ho, G.Y.F.; Ip, M.S.-M.; Lam, D.C.-L. Transfer of Extracellular Vesicle-Associated-RNAs Induces Drug Resistance in ALK-Translocated Lung Adenocarcinoma. Cancers 2019, 11, 104. [Google Scholar] [CrossRef]
  83. Pasini, L.; Ulivi, P. Extracellular Vesicles in Non-Small-Cell Lung Cancer: Functional Role and Involvement in Resistance to Targeted Treatment and Immunotherapy. Cancers 2019, 12, 40. [Google Scholar] [CrossRef] [PubMed]
  84. Hur, J.Y.; Lee, J.S.; Kim, I.A.; Kim, H.J.; Kim, W.S.; Lee, K.Y. Extracellular Vesicle-Based EGFR Genotyping in Bronchoalveolar Lavage Fluid from Treatment-Naive Non-Small Cell Lung Cancer Patients. Transl. Lung Cancer Res. 2019, 8, 1051–1060. [Google Scholar] [CrossRef] [PubMed]
  85. Sánchez-Herrero, E.; Campos-Silva, C.; Cáceres-Martell, Y.; Robado de Lope, L.; Sanz-Moreno, S.; Serna-Blasco, R.; Rodríguez-Festa, A.; Ares Trotta, D.; Martín-Acosta, P.; Patiño, C.; et al. ALK-Fusion Transcripts Can Be Detected in Extracellular Vesicles (EVs) from Nonsmall Cell Lung Cancer Cell Lines and Patient Plasma: Toward EV-Based Noninvasive Testing. Clin. Chem. 2022, 68, 668–679. [Google Scholar] [CrossRef]
  86. Yuwen, D.-L.; Sheng, B.-B.; Liu, J.; Wenyu, W.; Shu, Y.-Q. MiR-146a-5p Level in Serum Exosomes Predicts Therapeutic Effect of Cisplatin in Non-Small Cell Lung Cancer. Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 2650–2658. [Google Scholar]
  87. Han, B.; Marrades, R.M.; Viñolas, N.; He, Y.; Canals, J.; Díaz, T.; Molins, L.; Martinez, D.; Moisés, J.; Sánchez, D.; et al. Monitoring HOTTIP Levels on Extracellular Vesicles for Predicting Recurrence in Surgical Non-Small Cell Lung Cancer Patients. Transl. Oncol. 2021, 14, 101144. [Google Scholar] [CrossRef] [PubMed]
  88. Whiteside, T.L. Exosomes and Tumor-Mediated Immune Suppression. J. Clin. Investig. 2016, 126, 1216–1223. [Google Scholar] [CrossRef]
  89. Tian, W.; Liu, S.; Li, B. Potential Role of Exosomes in Cancer Metastasis. BioMed Res. Int. 2019, 2019, 4649705. [Google Scholar] [CrossRef]
  90. Jiang, C.; Zhang, N.; Hu, X.; Wang, H. Tumor-Associated Exosomes Promote Lung Cancer Metastasis through Multiple Mechanisms. Mol. Cancer 2021, 20, 117. [Google Scholar] [CrossRef]
  91. Kuriyama, N.; Yoshioka, Y.; Kikuchi, S.; Azuma, N.; Ochiya, T. Extracellular Vesicles Are Key Regulators of Tumor Neovasculature. Front. Cell Dev. Biol. 2020, 8, 611039. [Google Scholar] [CrossRef]
  92. Zhao, L.; Ma, X.; Yu, J. Exosomes and Organ-Specific Metastasis. Mol. Ther. Methods Clin. Dev. 2021, 22, 133–147. [Google Scholar] [CrossRef]
  93. Gao, J.; Zhang, X.; Jiang, L.; Li, Y.; Zheng, Q. Tumor Endothelial Cell-Derived Extracellular Vesicles Contribute to Tumor Microenvironment Remodeling. Cell Commun. Signal. CCS 2022, 20, 97. [Google Scholar] [CrossRef] [PubMed]
  94. Tao, S.-C.; Guo, S.-C.; Zhang, C.-Q. Platelet-Derived Extracellular Vesicles: An Emerging Therapeutic Approach. Int. J. Biol. Sci. 2017, 13, 828–834. [Google Scholar] [CrossRef] [PubMed]
  95. Labelle, M.; Hynes, R.O. The Initial Hours of Metastasis: The Importance of Cooperative Host-Tumor Cell Interactions during Hematogenous Dissemination. Cancer Discov. 2012, 2, 1091–1099. [Google Scholar] [CrossRef] [PubMed]
  96. Nikanjam, M.; Kato, S.; Kurzrock, R. Liquid Biopsy: Current Technology and Clinical Applications. J. Hematol. Oncol. 2022, 15, 131. [Google Scholar] [CrossRef]
  97. Stroun, M.; Anker, P.; Maurice, P.; Lyautey, J.; Lederrey, C.; Beljanski, M. Neoplastic Characteristics of the DNA Found in the Plasma of Cancer Patients. Oncology 1989, 46, 318–322. [Google Scholar] [CrossRef]
  98. Sánchez-Herrero, E.; Serna-Blasco, R.; Robado de Lope, L.; González-Rumayor, V.; Romero, A.; Provencio, M. Circulating Tumor DNA as a Cancer Biomarker: An Overview of Biological Features and Factors That May Impact on CtDNA Analysis. Front. Oncol. 2022, 12, 943253. [Google Scholar] [CrossRef]
  99. Wong, F.C.K.; Sun, K.; Jiang, P.; Cheng, Y.K.Y.; Chan, K.C.A.; Leung, T.Y.; Chiu, R.W.K.; Lo, Y.M.D. Cell-Free DNA in Maternal Plasma and Serum: A Comparison of Quantity, Quality and Tissue Origin Using Genomic and Epigenomic Approaches. Clin. Biochem. 2016, 49, 1379–1386. [Google Scholar] [CrossRef]
  100. Devonshire, A.S.; Whale, A.S.; Gutteridge, A.; Jones, G.; Cowen, S.; Foy, C.A.; Huggett, J.F. Towards Standardisation of Cell-Free DNA Measurement in Plasma: Controls for Extraction Efficiency, Fragment Size Bias and Quantification. Anal. Bioanal. Chem. 2014, 406, 6499–6512. [Google Scholar] [CrossRef]
  101. Parsons, H.A.; Beaver, J.A.; Park, B.H. Circulating Plasma Tumor DNA. Adv. Exp. Med. Biol. 2016, 882, 259–276. [Google Scholar] [CrossRef]
  102. Danesi, R.; Lo, Y.M.D.; Oellerich, M.; Beck, J.; Galbiati, S.; Re, M.D.; Lianidou, E.; Neumaier, M.; van Schaik, R.H.N. What Do We Need to Obtain High Quality Circulating Tumor DNA (CtDNA) for Routine Diagnostic Test in Oncology?—Considerations on Pre-Analytical Aspects by the IFCC Workgroup CfDNA. Clin. Chim. Acta Int. J. Clin. Chem. 2021, 520, 168–171. [Google Scholar] [CrossRef] [PubMed]
  103. Elazezy, M.; Joosse, S.A. Techniques of Using Circulating Tumor DNA as a Liquid Biopsy Component in Cancer Management. Comput. Struct. Biotechnol. J. 2018, 16, 370–378. [Google Scholar] [CrossRef]
  104. Heitzer, E.; van den Broek, D.; Denis, M.G.; Hofman, P.; Hubank, M.; Mouliere, F.; Paz-Ares, L.; Schuuring, E.; Sültmann, H.; Vainer, G.; et al. Recommendations for a Practical Implementation of Circulating Tumor DNA Mutation Testing in Metastatic Non-Small-Cell Lung Cancer. ESMO Open 2022, 7, 100399. [Google Scholar] [CrossRef] [PubMed]
  105. Assaf, Z.J.F.; Zou, W.; Fine, A.D.; Socinski, M.A.; Young, A.; Lipson, D.; Freidin, J.F.; Kennedy, M.; Polisecki, E.; Nishio, M.; et al. A Longitudinal Circulating Tumor DNA-Based Model Associated with Survival in Metastatic Non-Small-Cell Lung Cancer. Nat. Med. 2023, 29, 859–868. [Google Scholar] [CrossRef] [PubMed]
  106. Zou, W.; Yaung, S.J.; Fuhlbrück, F.; Ballinger, M.; Peters, E.; Palma, J.F.; Shames, D.S.; Gandara, D.; Jiang, Y.; Patil, N.S. CtDNA Predicts Overall Survival in Patients With NSCLC Treated With PD-L1 Blockade or With Chemotherapy. JCO Precis. Oncol. 2021, 5, 827–838. [Google Scholar] [CrossRef]
  107. Buder, A.; Hochmair, M.J.; Setinek, U.; Pirker, R.; Filipits, M. EGFR Mutation Tracking Predicts Survival in Advanced EGFR-Mutated Non-Small Cell Lung Cancer Patients Treated with Osimertinib. Transl. Lung Cancer Res. 2020, 9, 239–245. [Google Scholar] [CrossRef]
  108. Cheng, M.L.; Lau, C.J.; Milan, M.S.D.; Supplee, J.G.; Riess, J.W.; Bradbury, P.A.; Jänne, P.A.; Oxnard, G.R.; Paweletz, C.P. Plasma CtDNA Response Is an Early Marker of Treatment Effect in Advanced NSCLC. JCO Precis. Oncol. 2021, 5, 393–402. [Google Scholar] [CrossRef]
  109. Bratman, S.V.; Yang, S.Y.C.; Iafolla, M.A.J.; Liu, Z.; Hansen, A.R.; Bedard, P.L.; Lheureux, S.; Spreafico, A.; Razak, A.A.; Shchegrova, S.; et al. Personalized Circulating Tumor DNA Analysis as a Predictive Biomarker in Solid Tumor Patients Treated with Pembrolizumab. Nat. Cancer 2020, 1, 873–881. [Google Scholar] [CrossRef]
  110. Ricciuti, B.; Jones, G.; Severgnini, M.; Alessi, J.V.; Recondo, G.; Lawrence, M.; Forshew, T.; Lydon, C.; Nishino, M.; Cheng, M.; et al. Early Plasma Circulating Tumor DNA (CtDNA) Changes Predict Response to First-Line Pembrolizumab-Based Therapy in Non-Small Cell Lung Cancer (NSCLC). J. Immunother. Cancer 2021, 9, e001504. [Google Scholar] [CrossRef]
  111. Li, N.; Wang, B.-X.; Li, J.; Shao, Y.; Li, M.-T.; Li, J.-J.; Kuang, P.-P.; Liu, Z.; Sun, T.-Y.; Wu, H.-Q.; et al. Perioperative Circulating Tumor DNA as a Potential Prognostic Marker for Operable Stage I to IIIA Non-Small Cell Lung Cancer. Cancer 2022, 128, 708–718. [Google Scholar] [CrossRef]
  112. Gale, D.; Heider, K.; Ruiz-Valdepenas, A.; Hackinger, S.; Perry, M.; Marsico, G.; Rundell, V.; Wulff, J.; Sharma, G.; Knock, H.; et al. Residual CtDNA after Treatment Predicts Early Relapse in Patients with Early-Stage Non-Small Cell Lung Cancer. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2022, 33, 500–510. [Google Scholar] [CrossRef] [PubMed]
  113. Wang, S.; Li, M.; Zhang, J.; Xing, P.; Wu, M.; Meng, F.; Jiang, F.; Wang, J.; Bao, H.; Huang, J.; et al. Circulating Tumor DNA Integrating Tissue Clonality Detects Minimal Residual Disease in Resectable Non-Small-Cell Lung Cancer. J. Hematol. Oncol. 2022, 15, 137. [Google Scholar] [CrossRef]
  114. Zhang, R.; Zhang, X.; Huang, Z.; Wang, F.; Lin, Y.; Wen, Y.; Liu, L.; Li, J.; Liu, X.; Xie, W.; et al. Development and Validation of a Preoperative Noninvasive Predictive Model Based on Circular Tumor DNA for Lymph Node Metastasis in Resectable Non-Small Cell Lung Cancer. Transl. Lung Cancer Res. 2020, 9, 722–730. [Google Scholar] [CrossRef] [PubMed]
  115. Tzimagiorgis, G.; Michailidou, E.Z.; Kritis, A.; Markopoulos, A.K.; Kouidou, S. Recovering Circulating Extracellular or Cell-Free RNA from Bodily Fluids. Cancer Epidemiol. 2011, 35, 580–589. [Google Scholar] [CrossRef] [PubMed]
  116. Liu, Z.; Wang, T.; Yang, X.; Zhou, Q.; Zhu, S.; Zeng, J.; Chen, H.; Sun, J.; Li, L.; Xu, J.; et al. Polyadenylation Ligation-Mediated Sequencing (PALM-Seq) Characterizes Cell-Free Coding and Non-Coding RNAs in Human Biofluids. Clin. Transl. Med. 2022, 12, e987. [Google Scholar] [CrossRef]
  117. Cabús, L.; Lagarde, J.; Curado, J.; Lizano, E.; Pérez-Boza, J. Current Challenges and Best Practices for Cell-Free Long RNA Biomarker Discovery. Biomark. Res. 2022, 10, 62. [Google Scholar] [CrossRef] [PubMed]
  118. Kolenda, T.; Guglas, K.; Baranowski, D.; Sobocińska, J.; Kopczyńska, M.; Teresiak, A.; Bliźniak, R.; Lamperska, K. CfRNAs as Biomarkers in Oncology—Still Experimental or Applied Tool for Personalized Medicine Already? Rep. Pract. Oncol. Radiother. J. Gt. Cancer Cent. Poznan Pol. Soc. Radiat. Oncol. 2020, 25, 783–792. [Google Scholar] [CrossRef] [PubMed]
  119. Bruno, R.; Fontanini, G. Next Generation Sequencing for Gene Fusion Analysis in Lung Cancer: A Literature Review. Diagnostics 2020, 10, 521. [Google Scholar] [CrossRef]
  120. Mensah, M.; Borzi, C.; Verri, C.; Suatoni, P.; Conte, D.; Pastorino, U.; Orazio, F.; Sozzi, G.; Boeri, M. MicroRNA Based Liquid Biopsy: The Experience of the Plasma MiRNA Signature Classifier (MSC) for Lung Cancer Screening. J. Vis. Exp. JoVE 2017, 128, 56326. [Google Scholar] [CrossRef]
  121. Ishiba, T.; Hoffmann, A.-C.; Usher, J.; Elshimali, Y.; Sturdevant, T.; Dang, M.; Jaimes, Y.; Tyagi, R.; Gonzales, R.; Grino, M.; et al. Frequencies and Expression Levels of Programmed Death Ligand 1 (PD-L1) in Circulating Tumor RNA (CtRNA) in Various Cancer Types. Biochem. Biophys. Res. Commun. 2018, 500, 621–625. [Google Scholar] [CrossRef]
  122. Gu, X.; He, J.; Ji, G. Combined Use of Circulating Tumor Cells and Salivary MRNA to Detect Non-Small-Cell Lung Cancer. Medicine 2020, 99, e19097. [Google Scholar] [CrossRef] [PubMed]
  123. Pös, O.; Biró, O.; Szemes, T.; Nagy, B. Circulating Cell-Free Nucleic Acids: Characteristics and Applications. Eur. J. Hum. Genet. EJHG 2018, 26, 937–945. [Google Scholar] [CrossRef] [PubMed]
  124. Xu, Q.; Ye, L.; Huang, L.; Zhou, L.; Chen, X.; Ye, M.; Wu, G.; Zhan, P.; Lv, T.; Song, Y. Serum Exosomal MiRNA Might Be a Novel Liquid Biopsy to Identify Leptomeningeal Metastasis in Non-Small Cell Lung Cancer. OncoTargets Ther. 2021, 14, 2327–2335. [Google Scholar] [CrossRef] [PubMed]
  125. Wu, L.; Hu, B.; Zhao, B.; Liu, Y.; Yang, Y.; Zhang, L.; Chen, J. Circulating MicroRNA-422a Is Associated with Lymphatic Metastasis in Lung Cancer. Oncotarget 2017, 8, 42173–42188. [Google Scholar] [CrossRef] [PubMed]
  126. Freitas, C.; Sousa, C.; Machado, F.; Serino, M.; Santos, V.; Cruz-Martins, N.; Teixeira, A.; Cunha, A.; Pereira, T.; Oliveira, H.P.; et al. The Role of Liquid Biopsy in Early Diagnosis of Lung Cancer. Front. Oncol. 2021, 11, 634316. [Google Scholar] [CrossRef]
  127. Hubaux, R.; Becker-Santos, D.D.; Enfield, K.S.S.; Lam, S.; Lam, W.L.; Martinez, V.D. MicroRNAs As Biomarkers For Clinical Features Of Lung Cancer. Metab. Open Access 2012, 2, 1000108. [Google Scholar] [CrossRef]
  128. Shen, L.; Chen, L.; Wang, Y.; Jiang, X.; Xia, H.; Zhuang, Z. Long Noncoding RNA MALAT1 Promotes Brain Metastasis by Inducing Epithelial-Mesenchymal Transition in Lung Cancer. J. Neurooncol. 2015, 121, 101–108. [Google Scholar] [CrossRef]
  129. Zhang, R.; Xia, Y.; Wang, Z.; Zheng, J.; Chen, Y.; Li, X.; Wang, Y.; Ming, H. Serum Long Non Coding RNA MALAT-1 Protected by Exosomes Is up-Regulated and Promotes Cell Proliferation and Migration in Non-Small Cell Lung Cancer. Biochem. Biophys. Res. Commun. 2017, 490, 406–414. [Google Scholar] [CrossRef]
  130. Weber, D.G.; Johnen, G.; Casjens, S.; Bryk, O.; Pesch, B.; Jöckel, K.-H.; Kollmeier, J.; Brüning, T. Evaluation of Long Noncoding RNA MALAT1 as a Candidate Blood-Based Biomarker for the Diagnosis of Non-Small Cell Lung Cancer. BMC Res. Notes 2013, 6, 518. [Google Scholar] [CrossRef]
  131. Siravegna, G.; Marsoni, S.; Siena, S.; Bardelli, A. Integrating Liquid Biopsies into the Management of Cancer. Nat. Rev. Clin. Oncol. 2017, 14, 531–548. [Google Scholar] [CrossRef] [PubMed]
  132. Malczewska, A.; Bodei, L.; Kidd, M.; Modlin, I.M. Blood MRNA Measurement (NETest) for Neuroendocrine Tumor Diagnosis of Image-Negative Liver Metastatic Disease. J. Clin. Endocrinol. Metab. 2019, 104, 867–872. [Google Scholar] [CrossRef] [PubMed]
  133. Ibrahim, J.; Peeters, M.; Van Camp, G.; Op de Beeck, K. Methylation Biomarkers for Early Cancer Detection and Diagnosis: Current and Future Perspectives. Eur. J. Cancer Oxf. Engl. 1990 2023, 178, 91–113. [Google Scholar] [CrossRef] [PubMed]
  134. Locke, W.J.; Guanzon, D.; Ma, C.; Liew, Y.J.; Duesing, K.R.; Fung, K.Y.C.; Ross, J.P. DNA Methylation Cancer Biomarkers: Translation to the Clinic. Front. Genet. 2019, 10, 1150. [Google Scholar] [CrossRef] [PubMed]
  135. Constantin, N.; Sina, A.A.I.; Korbie, D.; Trau, M. Opportunities for Early Cancer Detection: The Rise of CtDNA Methylation-Based Pan-Cancer Screening Technologies. Epigenomes 2022, 6, 6. [Google Scholar] [CrossRef] [PubMed]
  136. Liu, M.C.; Oxnard, G.R.; Klein, E.A.; Swanton, C.S.M.C.C.; Seiden, M.V.; Liu, M.C.; Oxnard, G.R.; Klein, E.A.; Smith, D.; Richards, D.; et al. Sensitive and Specific Multi-Cancer Detection and Localization Using Methylation Signatures in Cell-Free DNA. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2020, 31, 745–759. [Google Scholar] [CrossRef]
  137. Zhao, H.; Zhang, H.; Xu, W.; Wen, B.; Kang, Y. A Sight of the Diagnostic Value of Aberrant Cell-Free DNA Methylation in Lung Cancer. Dis. Markers 2022, 2022, 9619357. [Google Scholar] [CrossRef]
  138. Janke, F.; Angeles, A.K.; Riediger, A.L.; Bauer, S.; Reck, M.; Stenzinger, A.; Schneider, M.A.; Muley, T.; Thomas, M.; Christopoulos, P.; et al. Longitudinal Monitoring of Cell-Free DNA Methylation in ALK-Positive Non-Small Cell Lung Cancer Patients. Clin. Epigenetics 2022, 14, 163. [Google Scholar] [CrossRef]
  139. Mo, S.; Dai, W.; Wang, H.; Lan, X.; Ma, C.; Su, Z.; Xiang, W.; Han, L.; Luo, W.; Zhang, L.; et al. Early Detection and Prognosis Prediction for Colorectal Cancer by Circulating Tumour DNA Methylation Haplotypes: A Multicentre Cohort Study. eClinicalMedicine 2023, 55, 101717. [Google Scholar] [CrossRef]
  140. Kandimalla, R.; Xu, J.; Link, A.; Matsuyama, T.; Yamamura, K.; Parker, M.I.; Uetake, H.; Balaguer, F.; Borazanci, E.; Tsai, S.; et al. EpiPanGI Dx: A Cell-Free DNA Methylation Fingerprint for the Early Detection of Gastrointestinal Cancers. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2021, 27, 6135–6144. [Google Scholar] [CrossRef]
  141. Liang, R.; Li, X.; Li, W.; Zhu, X.; Li, C. DNA Methylation in Lung Cancer Patients: Opening a “Window of Life” under Precision Medicine. Biomed. Pharmacother. 2021, 144, 112202. [Google Scholar] [CrossRef]
  142. Li, P.; Liu, S.; Du, L.; Mohseni, G.; Zhang, Y.; Wang, C. Liquid Biopsies Based on DNA Methylation as Biomarkers for the Detection and Prognosis of Lung Cancer. Clin. Epigenetics 2022, 14, 118. [Google Scholar] [CrossRef]
  143. Liu, Z.; Cheng, X.; Zhang, L.; Zhou, J.; Deng, D.; Ji, J. A Panel of DNA Methylated Markers Predicts Metastasis of PN0M0 Gastric Carcinoma: A Prospective Cohort Study. Br. J. Cancer 2019, 121, 529–536. [Google Scholar] [CrossRef]
  144. Liu, L.; Toung, J.M.; Jassowicz, A.F.; Vijayaraghavan, R.; Kang, H.; Zhang, R.; Kruglyak, K.M.; Huang, H.J.; Hinoue, T.; Shen, H.; et al. Targeted Methylation Sequencing of Plasma Cell-Free DNA for Cancer Detection and Classification. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2018, 29, 1445–1453. [Google Scholar] [CrossRef] [PubMed]
  145. Peng, Y.; Mei, W.; Ma, K.; Zeng, C. Circulating Tumor DNA and Minimal Residual Disease (MRD) in Solid Tumors: Current Horizons and Future Perspectives. Front. Oncol. 2021, 11, 763790. [Google Scholar] [CrossRef] [PubMed]
  146. Früh, M.; Rolland, E.; Pignon, J.-P.; Seymour, L.; Ding, K.; Tribodet, H.; Winton, T.; Le Chevalier, T.; Scagliotti, G.V.; Douillard, J.Y.; et al. Pooled Analysis of the Effect of Age on Adjuvant Cisplatin-Based Chemotherapy for Completely Resected Non-Small-Cell Lung Cancer. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2008, 26, 3573–3581. [Google Scholar] [CrossRef] [PubMed]
  147. Dagogo-Jack, I.; Shaw, A.T. Tumour Heterogeneity and Resistance to Cancer Therapies. Nat. Rev. Clin. Oncol. 2018, 15, 81–94. [Google Scholar] [CrossRef] [PubMed]
  148. Gainor, J.F.; Dardaei, L.; Yoda, S.; Friboulet, L.; Leshchiner, I.; Katayama, R.; Dagogo-Jack, I.; Gadgeel, S.; Schultz, K.; Singh, M.; et al. Molecular Mechanisms of Resistance to First- and Second-Generation ALK Inhibitors in ALK-Rearranged Lung Cancer. Cancer Discov. 2016, 6, 1118–1133. [Google Scholar] [CrossRef]
  149. Sequist, L.V.; Waltman, B.A.; Dias-Santagata, D.; Digumarthy, S.; Turke, A.B.; Fidias, P.; Bergethon, K.; Shaw, A.T.; Gettinger, S.; Cosper, A.K.; et al. Genotypic and Histological Evolution of Lung Cancers Acquiring Resistance to EGFR Inhibitors. Sci. Transl. Med. 2011, 3, 75ra26. [Google Scholar] [CrossRef]
  150. Yu, H.A.; Arcila, M.E.; Rekhtman, N.; Sima, C.S.; Zakowski, M.F.; Pao, W.; Kris, M.G.; Miller, V.A.; Ladanyi, M.; Riely, G.J. Analysis of Tumor Specimens at the Time of Acquired Resistance to EGFR-TKI Therapy in 155 Patients with EGFR-Mutant Lung Cancers. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2013, 19, 2240–2247. [Google Scholar] [CrossRef]
  151. Black, J.R.M.; McGranahan, N. Genetic and Non-Genetic Clonal Diversity in Cancer Evolution. Nat. Rev. Cancer 2021, 21, 379–392. [Google Scholar] [CrossRef]
  152. McGranahan, N.; Swanton, C. Clonal Heterogeneity and Tumor Evolution: Past, Present, and the Future. Cell 2017, 168, 613–628. [Google Scholar] [CrossRef] [PubMed]
  153. Roper, N.; Brown, A.-L.; Wei, J.S.; Pack, S.; Trindade, C.; Kim, C.; Restifo, O.; Gao, S.; Sindiri, S.; Mehrabadi, F.; et al. Clonal Evolution and Heterogeneity of Osimertinib Acquired Resistance Mechanisms in EGFR Mutant Lung Cancer. Cell Rep. Med. 2020, 1, 100007. [Google Scholar] [CrossRef] [PubMed]
  154. Chen, J.; Facchinetti, F.; Braye, F.; Yurchenko, A.A.; Bigot, L.; Ponce, S.; Planchard, D.; Gazzah, A.; Nikolaev, S.; Michiels, S.; et al. Single-Cell DNA-Seq Depicts Clonal Evolution of Multiple Driver Alterations in Osimertinib-Resistant Patients. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2022, 33, 434–444. [Google Scholar] [CrossRef] [PubMed]
  155. Guibert, N.; Pradines, A.; Favre, G.; Mazieres, J. Current and Future Applications of Liquid Biopsy in Nonsmall Cell Lung Cancer from Early to Advanced Stages. Eur. Respir. Rev. Off. J. Eur. Respir. Soc. 2020, 29, 190052. [Google Scholar] [CrossRef] [PubMed]
  156. Crowley, E.; Di Nicolantonio, F.; Loupakis, F.; Bardelli, A. Liquid Biopsy: Monitoring Cancer-Genetics in the Blood. Nat. Rev. Clin. Oncol. 2013, 10, 472–484. [Google Scholar] [CrossRef] [PubMed]
  157. Honoré, N.; Galot, R.; van Marcke, C.; Limaye, N.; Machiels, J.-P. Liquid Biopsy to Detect Minimal Residual Disease: Methodology and Impact. Cancers 2021, 13, 5364. [Google Scholar] [CrossRef]
  158. Verzè, M.; Pluchino, M.; Leonetti, A.; Corianò, M.; Bonatti, F.; Armillotta, M.P.; Perrone, F.; Casali, M.; Minari, R.; Tiseo, M. Role of CtDNA for the Detection of Minimal Residual Disease in Resected Non-Small Cell Lung Cancer: A Systematic Review. Transl. Lung Cancer Res. 2022, 11, 2588–2600. [Google Scholar] [CrossRef] [PubMed]
  159. Markou, A.; Tzanikou, E.; Lianidou, E. The Potential of Liquid Biopsy in the Management of Cancer Patients. Semin. Cancer Biol. 2022, 84, 69–79. [Google Scholar] [CrossRef]
  160. Xia, L.; Mei, J.; Kang, R.; Deng, S.; Chen, Y.; Yang, Y.; Feng, G.; Deng, Y.; Gan, F.; Lin, Y.; et al. Perioperative CtDNA-Based Molecular Residual Disease Detection for Non–Small Cell Lung Cancer: A Prospective Multicenter Cohort Study (LUNGCA-1). Clin. Cancer Res. 2022, 28, 3308–3317. [Google Scholar] [CrossRef]
  161. 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]
  162. Yue, D.; Liu, W.; Chen, C.; Zhang, T.; Ma, Y.; Cui, L.; Gu, Y.; Bei, T.; Zhao, X.; Zhang, B.; et al. Circulating Tumor DNA Predicts Neoadjuvant Immunotherapy Efficacy and Recurrence-Free Survival in Surgical Non-Small Cell Lung Cancer Patients. Transl. Lung Cancer Res. 2022, 11, 263–276. [Google Scholar] [CrossRef]
  163. O’Sullivan, H.M.; Feber, A.; Popat, S. Minimal Residual Disease Monitoring in Radically Treated Non-Small Cell Lung Cancer: Challenges and Future Directions. OncoTargets Ther. 2023, 16, 249–259. [Google Scholar] [CrossRef]
  164. Marsico, G.; Sharma, G.; Perry, M.; Hackinger, S.; Forshew, T.; Howarth, K.; Platt, J.; Rosenfeld, N.; Osborne, R. Abstract 3097: Analytical Development of the RaDaRTM Assay, a Highly Sensitive and Specific Assay for the Monitoring of Minimal Residual Disease. Cancer Res. 2020, 80, 3097. [Google Scholar] [CrossRef]
  165. Sethi, H.; Salari, R.; Navarro, S.; Natarajan, P.; Srinivasan, R.; Dashner, S.; Tin, T.; Balcioglu, M.; Swenerton, R.; Zimmermann, B. Abstract 4542: Analytical Validation of the SignateraTM RUO Assay, a Highly Sensitive Patient-Specific Multiplex PCR NGS-Based Noninvasive Cancer Recurrence Detection and Therapy Monitoring Assay. Cancer Res. 2018, 78, 4542. [Google Scholar] [CrossRef]
  166. Kohabir, K.; Wolthuis, R.; Sistermans, E.A. Fragmentomic CfDNA Patterns in Noninvasive Prenatal Testing and Beyond. J. Biomed. Transl. Res. 2021, 7, 38–47. [Google Scholar] [CrossRef]
  167. Majidpoor, J.; Mortezaee, K. Steps in Metastasis: An Updated Review. Med. Oncol. Northwood Lond. Engl. 2021, 38, 3. [Google Scholar] [CrossRef] [PubMed]
  168. Chambers, A.F.; Naumov, G.N.; Vantyghem, S.A.; Tuck, A.B. Molecular Biology of Breast Cancer Metastasis. Clinical Implications of Experimental Studies on Metastatic Inefficiency. Breast Cancer Res. BCR 2000, 2, 400–407. [Google Scholar] [CrossRef] [PubMed]
  169. Wood, S.L.; Pernemalm, M.; Crosbie, P.A.; Whetton, A.D. The Role of the Tumor-Microenvironment in Lung Cancer-Metastasis and Its Relationship to Potential Therapeutic Targets. Cancer Treat. Rev. 2014, 40, 558–566. [Google Scholar] [CrossRef]
  170. Welch, D.R.; Hurst, D.R. Defining the Hallmarks of Metastasis. Cancer Res. 2019, 79, 3011–3027. [Google Scholar] [CrossRef] [PubMed]
  171. Fares, J.; Fares, M.Y.; Khachfe, H.H.; Salhab, H.A.; Fares, Y. Molecular Principles of Metastasis: A Hallmark of Cancer Revisited. Signal Transduct. Target. Ther. 2020, 5, 28. [Google Scholar] [CrossRef]
  172. Eslami-S, Z.; Cortés-Hernández, L.E.; Alix-Panabières, C. The Metastatic Cascade as the Basis for Liquid Biopsy Development. Front. Oncol. 2020, 10, 1055. [Google Scholar] [CrossRef] [PubMed]
  173. Todorova, D.; Simoncini, S.; Lacroix, R.; Sabatier, F.; Dignat-George, F. Extracellular Vesicles in Angiogenesis. Circ. Res. 2017, 120, 1658–1673. [Google Scholar] [CrossRef] [PubMed]
  174. Schillaci, O.; Fontana, S.; Monteleone, F.; Taverna, S.; Di Bella, M.A.; Di Vizio, D.; Alessandro, R. Exosomes from Metastatic Cancer Cells Transfer Amoeboid Phenotype to Non-Metastatic Cells and Increase Endothelial Permeability: Their Emerging Role in Tumor Heterogeneity. Sci. Rep. 2017, 7, 4711. [Google Scholar] [CrossRef] [PubMed]
  175. Peinado, H.; Alečković, M.; Lavotshkin, S.; Matei, I.; Costa-Silva, B.; Moreno-Bueno, G.; Hergueta-Redondo, M.; Williams, C.; García-Santos, G.; Ghajar, C.; et al. Melanoma Exosomes Educate Bone Marrow Progenitor Cells toward a Pro-Metastatic Phenotype through MET. Nat. Med. 2012, 18, 883–891. [Google Scholar] [CrossRef]
  176. Ward, Y.; Lake, R.; Faraji, F.; Sperger, J.; Martin, P.; Gilliard, C.; Ku, K.P.; Rodems, T.; Niles, D.; Tillman, H.; et al. Platelets Promote Metastasis via Binding Tumor CD97 Leading to Bidirectional Signaling That Coordinates Transendothelial Migration. Cell Rep. 2018, 23, 808–822. [Google Scholar] [CrossRef] [PubMed]
  177. Varkey, J.; Nicolaides, T. Tumor-Educated Platelets: A Review of Current and Potential Applications in Solid Tumors. Cureus 2021, 13, e19189. [Google Scholar] [CrossRef]
  178. Jiang, X.; Wong, K.H.K.; Khankhel, A.H.; Zeinali, M.; Reategui, E.; Phillips, M.J.; Luo, X.; Aceto, N.; Fachin, F.; Hoang, A.N.; et al. Microfluidic Isolation of Platelet-Covered Circulating Tumor Cells. Lab Chip 2017, 17, 3498–3503. [Google Scholar] [CrossRef]
  179. Szczerba, B.M.; Castro-Giner, F.; Vetter, M.; Krol, I.; Gkountela, S.; Landin, J.; Scheidmann, M.C.; Donato, C.; Scherrer, R.; Singer, J.; et al. Neutrophils Escort Circulating Tumour Cells to Enable Cell Cycle Progression. Nature 2019, 566, 553–557. [Google Scholar] [CrossRef]
  180. Hoshino, A.; Costa-Silva, B.; Shen, T.-L.; Rodrigues, G.; Hashimoto, A.; Tesic Mark, M.; Molina, H.; Kohsaka, S.; Di Giannatale, A.; Ceder, S.; et al. Tumour Exosome Integrins Determine Organotropic Metastasis. Nature 2015, 527, 329–335. [Google Scholar] [CrossRef]
  181. Costa-Silva, B.; Aiello, N.M.; Ocean, A.J.; Singh, S.; Zhang, H.; Thakur, B.K.; Becker, A.; Hoshino, A.; Mark, M.T.; Molina, H.; et al. Pancreatic Cancer Exosomes Initiate Pre-Metastatic Niche Formation in the Liver. Nat. Cell Biol. 2015, 17, 816–826. [Google Scholar] [CrossRef]
  182. Kleiner, D.E.; Stetler-Stevenson, W.G. Matrix Metalloproteinases and Metastasis. Cancer Chemother. Pharmacol. 1999, 43, S42–S51. [Google Scholar] [CrossRef]
  183. Deryugina, E.I.; Quigley, J.P. Matrix Metalloproteinases and Tumor Metastasis. Cancer Metastasis Rev. 2006, 25, 9–34. [Google Scholar] [CrossRef] [PubMed]
  184. Quintero-Fabián, S.; Arreola, R.; Becerril-Villanueva, E.; Torres-Romero, J.C.; Arana-Argáez, V.; Lara-Riegos, J.; Ramírez-Camacho, M.A.; Alvarez-Sánchez, M.E. Role of Matrix Metalloproteinases in Angiogenesis and Cancer. Front. Oncol. 2019, 9, 1370. [Google Scholar] [CrossRef]
  185. Gonzalez-Avila, G.; Sommer, B.; Mendoza-Posada, D.A.; Ramos, C.; Garcia-Hernandez, A.A.; Falfan-Valencia, R. Matrix Metalloproteinases Participation in the Metastatic Process and Their Diagnostic and Therapeutic Applications in Cancer. Crit. Rev. Oncol. Hematol. 2019, 137, 57–83. [Google Scholar] [CrossRef]
  186. Kong, D.; Zhou, H.; Neelakantan, D.; Hughes, C.J.; Hsu, J.Y.; Srinivasan, R.R.; Lewis, M.T.; Ford, H.L. VEGF-C Mediates Tumor Growth and Metastasis through Promoting EMT-Epithelial Breast Cancer Cell Crosstalk. Oncogene 2021, 40, 964–979. [Google Scholar] [CrossRef] [PubMed]
  187. Valenti, R.; Huber, V.; Iero, M.; Filipazzi, P.; Parmiani, G.; Rivoltini, L. Tumor-Released Microvesicles as Vehicles of Immunosuppression. Cancer Res. 2007, 67, 2912–2915. [Google Scholar] [CrossRef]
  188. Yang, Y.; Cao, Y. The Impact of VEGF on Cancer Metastasis and Systemic Disease. Semin. Cancer Biol. 2022, 86, 251–261. [Google Scholar] [CrossRef]
  189. Yang, X.; Zhang, Y.; Hosaka, K.; Andersson, P.; Wang, J.; Tholander, F.; Cao, Z.; Morikawa, H.; Tegnér, J.; Yang, Y.; et al. VEGF-B Promotes Cancer Metastasis through a VEGF-A-Independent Mechanism and Serves as a Marker of Poor Prognosis for Cancer Patients. Proc. Natl. Acad. Sci. USA 2015, 112, E2900–E2909. [Google Scholar] [CrossRef]
  190. Cheng, H.; Perez-Soler, R. Leptomeningeal Metastases in Non-Small-Cell Lung Cancer. Lancet Oncol. 2018, 19, e43–e55. [Google Scholar] [CrossRef] [PubMed]
  191. Li, Y.S.; Jiang, B.Y.; Yang, J.J.; Zhang, X.C.; Zhang, Z.; Ye, J.Y.; Zhong, W.Z.; Tu, H.Y.; Chen, H.J.; Wang, Z.; et al. Unique Genetic Profiles from Cerebrospinal Fluid Cell-Free DNA in Leptomeningeal Metastases of EGFR-Mutant Non-Small-Cell Lung Cancer: A New Medium of Liquid Biopsy. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2018, 29, 945–952. [Google Scholar] [CrossRef]
  192. Brower, J.V.; Saha, S.; Rosenberg, S.A.; Hullett, C.R.; Ian Robins, H. Management of Leptomeningeal Metastases: Prognostic Factors and Associated Outcomes. J. Clin. Neurosci. Off. J. Neurosurg. Soc. Australas. 2016, 27, 130–137. [Google Scholar] [CrossRef] [PubMed]
  193. De Mattos-Arruda, L.; Mayor, R.; Ng, C.K.Y.; Weigelt, B.; Martínez-Ricarte, F.; Torrejon, D.; Oliveira, M.; Arias, A.; Raventos, C.; Tang, J.; et al. Cerebrospinal Fluid-Derived Circulating Tumour DNA Better Represents the Genomic Alterations of Brain Tumours than Plasma. Nat. Commun. 2015, 6, 8839. [Google Scholar] [CrossRef] [PubMed]
  194. Yang, H.; Wen, L.; Zhao, C.; Chen, J.; Zhou, Z.; Zhou, C.; Cai, L.; Zhou, C. Cerebrospinal Fluid-Derived Circulating Tumor DNA Is More Comprehensive than Plasma in NSCLC Patients with Leptomeningeal Metastases Regardless of Extracranial Evolution. Heliyon 2022, 8, e12374. [Google Scholar] [CrossRef] [PubMed]
  195. Shen, F.; Liang, N.; Fan, Z.; Zhao, M.; Kang, J.; Wang, X.; Hu, Q.; Mu, Y.; Wang, K.; Yuan, M.; et al. Genomic Alterations Identification and Resistance Mechanisms Exploration of NSCLC With Central Nervous System Metastases Using Liquid Biopsy of Cerebrospinal Fluid: A Real-World Study. Front. Oncol. 2022, 12, 889591. [Google Scholar] [CrossRef] [PubMed]
  196. Wei, C.; Zhang, R.; Cai, Q.; Gao, X.; Tong, F.; Dong, J.; Hu, Y.; Wu, G.; Dong, X. MicroRNA-330-3p Promotes Brain Metastasis and Epithelial-Mesenchymal Transition via GRIA3 in Non-Small Cell Lung Cancer. Aging 2019, 11, 6734–6761. [Google Scholar] [CrossRef]
  197. Zhang, X.; Sai, B.; Wang, F.; Wang, L.; Wang, Y.; Zheng, L.; Li, G.; Tang, J.; Xiang, J. Hypoxic BMSC-Derived Exosomal MiRNAs Promote Metastasis of Lung Cancer Cells via STAT3-Induced EMT. Mol. Cancer 2019, 18, 40. [Google Scholar] [CrossRef]
  198. Yang, X.-R.; Pi, C.; Yu, R.; Fan, X.-J.; Peng, X.-X.; Zhang, X.-C.; Chen, Z.-H.; Wu, X.; Shao, Y.; Wu, Y.-L.; et al. Correlation of Exosomal MicroRNA Clusters with Bone Metastasis in Non-Small Cell Lung Cancer. Clin. Exp. Metastasis 2021, 38, 109–117. [Google Scholar] [CrossRef]
  199. Couraud, S.; Vaca-Paniagua, F.; Villar, S.; Oliver, J.; Schuster, T.; Blanché, H.; Girard, N.; Trédaniel, J.; Guilleminault, L.; Gervais, R.; et al. Noninvasive Diagnosis of Actionable Mutations by Deep Sequencing of Circulating Free DNA in Lung Cancer from Never-Smokers: A Proof-of-Concept Study from BioCAST/IFCT-1002. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2014, 20, 4613–4624. [Google Scholar] [CrossRef]
  200. Zhou, J.; Dong, F.; Cui, F.; Xu, R.; Tang, X. The Role of Circulating Tumor Cells in Evaluation of Prognosis and Treatment Response in Advanced Non-Small-Cell Lung Cancer. Cancer Chemother. Pharmacol. 2017, 79, 825–833. [Google Scholar] [CrossRef] [PubMed]
  201. Abbosh, C.; Frankell, A.M.; Harrison, T.; Kisistok, J.; Garnett, A.; Johnson, L.; Veeriah, S.; Moreau, M.; Chesh, A.; Chaunzwa, T.L.; et al. Tracking Early Lung Cancer Metastatic Dissemination in TRACERx Using CtDNA. Nature 2023, 616, 553–562. [Google Scholar] [CrossRef] [PubMed]
  202. Aggarwal, C.; Thompson, J.C.; Black, T.A.; Katz, S.I.; Fan, R.; Yee, S.S.; Chien, A.L.; Evans, T.L.; Bauml, J.M.; Alley, E.W.; et al. Clinical Implications of Plasma-Based Genotyping With the Delivery of Personalized Therapy in Metastatic Non–Small Cell Lung Cancer. JAMA Oncol. 2019, 5, 173. [Google Scholar] [CrossRef] [PubMed]
  203. Thress, K.S.; Paweletz, C.P.; Felip, E.; Cho, B.C.; Stetson, D.; Dougherty, B.; Lai, Z.; Markovets, A.; Vivancos, A.; Kuang, Y.; et al. Acquired EGFR C797S Mutation Mediates Resistance to AZD9291 in Non-Small Cell Lung Cancer Harboring EGFR T790M. Nat. Med. 2015, 21, 560–562. [Google Scholar] [CrossRef] [PubMed]
  204. Osumi, H.; Shinozaki, E.; Yamaguchi, K.; Zembutsu, H. Early Change in Circulating Tumor DNA as a Potential Predictor of Response to Chemotherapy in Patients with Metastatic Colorectal Cancer. Sci. Rep. 2019, 9, 17358. [Google Scholar] [CrossRef]
  205. Sinoquet, L.; Jacot, W.; Quantin, X.; Alix-Panabières, C. Liquid Biopsy and Immuno-Oncology for Advanced Nonsmall Cell Lung Cancer. Clin. Chem. 2023, 69, 23–40. [Google Scholar] [CrossRef]
  206. Sama, S.; Le, T.; Ullah, A.; Elhelf, I.A.; Kavuri, S.K.; Karim, N.A. The Role of Serial Liquid Biopsy in the Management of Metastatic Non-Small Cell Lung Cancer (NSCLC). Clin. Pract. 2022, 12, 419–424. [Google Scholar] [CrossRef] [PubMed]
  207. Crucitta, S.; Ruglioni, M.; Novi, C.; Manganiello, M.; Arici, R.; Petrini, I.; Pardini, E.; Cucchiara, F.; Marmorino, F.; Cremolini, C.; et al. Comparison of Digital PCR Systems for the Analysis of Liquid Biopsy Samples of Patients Affected by Lung and Colorectal Cancer. Clin. Chim. Acta Int. J. Clin. Chem. 2023, 541, 117239. [Google Scholar] [CrossRef]
  208. 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]
  209. Solutions—Guardant360. Available online: https://www.guardanthealthamea.com/solutions/ (accessed on 1 March 2023).
  210. FoundationOne CDx|Foundation Medicine. Available online: https://www.foundationmedicine.com/test/foundationone-cdx (accessed on 1 March 2023).
  211. Chen, M.; Zhao, H. Next-Generation Sequencing in Liquid Biopsy: Cancer Screening and Early Detection. Hum. Genom. 2019, 13, 34. [Google Scholar] [CrossRef]
  212. NCounter® Pro Analysis System|Gene Expression|NanoString. Available online: https://nanostring.com/products/ncounter-analysis-system/ncounter-pro/?utm_source=google&utm_medium=cpc&utm_campaign=ncounterpro&utm_id=Instruments_CombinedTier_Search&gad=1&gclid=CjwKCAjwge2iBhBBEiwAfXDBRx2KQC1n2oqYorQMQD9kIQvcKUQ99ybmkm5693ugCR2WYENahNuUkBoCx9UQAvD_BwE (accessed on 10 May 2023).
  213. Marin, E.; Reyes, R.; Arcocha, A.; Viñolas, N.; Mezquita, L.; Gonzalvo, E.; Saez de Gordoa, K.; Jares, P.; Reguart, N.; Teixido, C. Prospective Evaluation of Single Nucleotide Variants by Two Different Technologies in Paraffin Samples of Advanced Non-Small Cell Lung Cancer Patients. Diagnostics 2020, 10, 902. [Google Scholar] [CrossRef]
  214. Giménez-Capitán, A.; Bracht, J.; García, J.J.; Jordana-Ariza, N.; García, B.; Garzón, M.; Mayo-de-Las-Casas, C.; Viteri-Ramirez, S.; Martinez-Bueno, A.; Aguilar, A.; et al. Multiplex Detection of Clinically Relevant Mutations in Liquid Biopsies of Cancer Patients Using a Hybridization-Based Platform. Clin. Chem. 2021, 67, 554–563. [Google Scholar] [CrossRef]
  215. Wilkening, S.; Hemminki, K.; Thirumaran, R.K.; Bermejo, J.L.; Bonn, S.; Försti, A.; Kumar, R. Determination of Allele Frequency in Pooled DNA: Comparison of Three PCR-Based Methods. BioTechniques 2005, 39, 853–858. [Google Scholar] [CrossRef] [PubMed]
  216. Iwama, E.; Takayama, K.; Harada, T.; Okamoto, I.; Ookubo, F.; Kishimoto, J.; Baba, E.; Oda, Y.; Nakanishi, Y. Highly Sensitive and Quantitative Evaluation of the EGFR T790M Mutation by Nanofluidic Digital PCR. Oncotarget 2015, 6, 20466–20473. [Google Scholar] [CrossRef] [PubMed]
  217. Wei, B.; Kang, J.; Kibukawa, M.; Arreaza, G.; Maguire, M.; Chen, L.; Qiu, P.; Lang, L.; Aurora-Garg, D.; Cristescu, R.; et al. Evaluation of the TruSight Oncology 500 Assay for Routine Clinical Testing of Tumor Mutational Burden and Clinical Utility for Predicting Response to Pembrolizumab. J. Mol. Diagn. JMD 2022, 24, 600–608. [Google Scholar] [CrossRef] [PubMed]
  218. Lee, Y.; Clark, E.W.; Milan, M.S.D.; Champagne, C.; Michael, K.S.; Awad, M.M.; Barbie, D.A.; Cheng, M.L.; Kehl, K.L.; Marcoux, J.P.; et al. Turnaround Time of Plasma Next-Generation Sequencing in Thoracic Oncology Patients: A Quality Improvement Analysis. JCO Precis. Oncol. 2020, 4, 1098–1108. [Google Scholar] [CrossRef] [PubMed]
  219. Ivanov, M.; Baranova, A.; Butler, T.; Spellman, P.; Mileyko, V. Non-Random Fragmentation Patterns in Circulating Cell-Free DNA Reflect Epigenetic Regulation. BMC Genom. 2015, 16 (Suppl. 13), S1. [Google Scholar] [CrossRef]
  220. Lo, Y.M.D.; Han, D.S.C.; Jiang, P.; Chiu, R.W.K. Epigenetics, Fragmentomics, and Topology of Cell-Free DNA in Liquid Biopsies. Science 2021, 372, eaaw3616. [Google Scholar] [CrossRef]
  221. Ding, S.C.; Lo, Y.M.D. Cell-Free DNA Fragmentomics in Liquid Biopsy. Diagnostics 2022, 12, 978. [Google Scholar] [CrossRef]
  222. Zheng, G.X.Y.; Lau, B.T.; Schnall-Levin, M.; Jarosz, M.; Bell, J.M.; Hindson, C.M.; Kyriazopoulou-Panagiotopoulou, S.; Masquelier, D.A.; Merrill, L.; Terry, J.M.; et al. Haplotyping Germline and Cancer Genomes with High-Throughput Linked-Read Sequencing. Nat. Biotechnol. 2016, 34, 303–311. [Google Scholar] [CrossRef]
  223. Cristiano, S.; Leal, A.; Phallen, J.; Fiksel, J.; Adleff, V.; Bruhm, D.C.; Jensen, S.Ø.; Medina, J.E.; Hruban, C.; White, J.R.; et al. Genome-Wide Cell-Free DNA Fragmentation in Patients with Cancer. Nature 2019, 570, 385–389. [Google Scholar] [CrossRef]
  224. Foda, Z.H.; Annapragada, A.V.; Boyapati, K.; Bruhm, D.C.; Vulpescu, N.A.; Medina, J.E.; Mathios, D.; Cristiano, S.; Niknafs, N.; Luu, H.T.; et al. Detecting Liver Cancer Using Cell-Free DNA Fragmentomes. Cancer Discov. 2023, 13, 616–631. [Google Scholar] [CrossRef]
  225. Vessies, D.C.L.; Schuurbiers, M.M.F.; van der Noort, V.; Schouten, I.; Linders, T.C.; Lanfermeijer, M.; Ramkisoensing, K.L.; Hartemink, K.J.; Monkhorst, K.; van den Heuvel, M.M.; et al. Combining Variant Detection and Fragment Length Analysis Improves Detection of Minimal Residual Disease in Postsurgery Circulating Tumour DNA of Stage II-IIIA NSCLC Patients. Mol. Oncol. 2022, 16, 2719–2732. [Google Scholar] [CrossRef]
  226. Jain, M.; Olsen, H.E.; Paten, B.; Akeson, M. The Oxford Nanopore MinION: Delivery of Nanopore Sequencing to the Genomics Community. Genome Biol. 2016, 17, 239. [Google Scholar] [CrossRef]
  227. Eid, J.; Fehr, A.; Gray, J.; Luong, K.; Lyle, J.; Otto, G.; Peluso, P.; Rank, D.; Baybayan, P.; Bettman, B.; et al. Real-Time DNA Sequencing from Single Polymerase Molecules. Science 2009, 323, 133–138. [Google Scholar] [CrossRef] [PubMed]
  228. Wang, Y.; Zhao, Y.; Bollas, A.; Wang, Y.; Au, K.F. Nanopore Sequencing Technology, Bioinformatics and Applications. Nat. Biotechnol. 2021, 39, 1348–1365. [Google Scholar] [CrossRef] [PubMed]
  229. Martignano, F.; Munagala, U.; Crucitta, S.; Mingrino, A.; Semeraro, R.; Del Re, M.; Petrini, I.; Magi, A.; Conticello, S.G. Nanopore Sequencing from Liquid Biopsy: Analysis of Copy Number Variations from Cell-Free DNA of Lung Cancer Patients. Mol. Cancer 2021, 20, 32. [Google Scholar] [CrossRef]
  230. Choy, L.Y.L.; Peng, W.; Jiang, P.; Cheng, S.H.; Yu, S.C.Y.; Shang, H.; Olivia Tse, O.Y.; Wong, J.; Wong, V.W.S.; Wong, G.L.H.; et al. Single-Molecule Sequencing Enables Long Cell-Free DNA Detection and Direct Methylation Analysis for Cancer Patients. Clin. Chem. 2022, 68, 1151–1163. [Google Scholar] [CrossRef] [PubMed]
  231. Lim, S.B.; Di Lee, W.; Vasudevan, J.; Lim, W.-T.; Lim, C.T. Liquid Biopsy: One Cell at a Time. NPJ Precis. Oncol. 2019, 3, 23. [Google Scholar] [CrossRef]
  232. Lu, S.; Chang, C.-J.; Guan, Y.; Szafer-Glusman, E.; Punnoose, E.; Do, A.; Suttmann, B.; Gagnon, R.; Rodriguez, A.; Landers, M.; et al. Genomic Analysis of Circulating Tumor Cells at the Single-Cell Level. J. Mol. Diagn. JMD 2020, 22, 770–781. [Google Scholar] [CrossRef]
  233. Ni, X.; Zhuo, M.; Su, Z.; Duan, J.; Gao, Y.; Wang, Z.; Zong, C.; Bai, H.; Chapman, A.R.; Zhao, J.; et al. Reproducible Copy Number Variation Patterns among Single Circulating Tumor Cells of Lung Cancer Patients. Proc. Natl. Acad. Sci. USA 2013, 110, 21083–21088. [Google Scholar] [CrossRef]
  234. Su, Z.; Wang, Z.; Ni, X.; Duan, J.; Gao, Y.; Zhuo, M.; Li, R.; Zhao, J.; Ma, Q.; Bai, H.; et al. Inferring the Evolution and Progression of Small-Cell Lung Cancer by Single-Cell Sequencing of Circulating Tumor Cells. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2019, 25, 5049–5060. [Google Scholar] [CrossRef]
  235. Zheng, D.; Liwinski, T.; Elinav, E. Interaction between Microbiota and Immunity in Health and Disease. Cell Res. 2020, 30, 492–506. [Google Scholar] [CrossRef] [PubMed]
  236. Chen, H.; Ma, Y.; Liu, Z.; Li, J.; Li, X.; Yang, F.; Qiu, M. Circulating Microbiome DNA: An Emerging Paradigm for Cancer Liquid Biopsy. Cancer Lett. 2021, 521, 82–87. [Google Scholar] [CrossRef] [PubMed]
  237. Yusuf, K.; Sampath, V.; Umar, S. Bacterial Infections and Cancer: Exploring This Association and Its Implications for Cancer Patients. Int. J. Mol. Sci. 2023, 24, 3110. [Google Scholar] [CrossRef] [PubMed]
  238. Tateishi, A.T.; Okuma, Y. Onco-Biome in Pharmacotherapy for Lung Cancer: A Narrative Review. Transl. Lung Cancer Res. 2022, 11, 2332–2345. [Google Scholar] [CrossRef] [PubMed]
  239. Lam, K.C.; Araya, R.E.; Huang, A.; Chen, Q.; Di Modica, M.; Rodrigues, R.R.; Lopès, A.; Johnson, S.B.; Schwarz, B.; Bohrnsen, E.; et al. Microbiota Triggers STING-Type I IFN-Dependent Monocyte Reprogramming of the Tumor Microenvironment. Cell 2021, 184, 5338–5356.e21. [Google Scholar] [CrossRef]
  240. Hilmi, M.; Kamal, M.; Vacher, S.; Dupain, C.; Ibadioune, S.; Halladjian, M.; Sablin, M.P.; Marret, G.; Ajgal, Z.C.; Nijnikoff, M.; et al. Intratumoral Microbiome Is Driven by Metastatic Site and Associated with Immune Histopathological Parameters: An Ancillary Study of the SHIVA Clinical Trial. Eur. J. Cancer Oxf. Engl. 1990 2023, 183, 152–161. [Google Scholar] [CrossRef]
  241. Liu, B.; Chau, J.; Dai, Q.; Zhong, C.; Zhang, J. Exploring Gut Microbiome in Predicting the Efficacy of Immunotherapy in Non-Small Cell Lung Cancer. Cancers 2022, 14, 5401. [Google Scholar] [CrossRef]
  242. Verschueren, M.V.; van der Welle, C.C.; Tonn, M.; Schramel, F.M.N.H.; Peters, B.J.M.; van de Garde, E.M.W. The Association between Gut Microbiome Affecting Concomitant Medication and the Effectiveness of Immunotherapy in Patients with Stage IV NSCLC. Sci. Rep. 2021, 11, 23331. [Google Scholar] [CrossRef]
  243. Bredin, P.; Naidoo, J. The Gut Microbiome, Immune Check Point Inhibition and Immune-Related Adverse Events in Non-Small Cell Lung Cancer. Cancer Metastasis Rev. 2022, 41, 347–366. [Google Scholar] [CrossRef]
  244. Dora, D.; Bokhari, S.M.Z.; Aloss, K.; Takacs, P.; Desnoix, J.Z.; Szklenárik, G.; Hurley, P.D.; Lohinai, Z. Implication of the Gut Microbiome and Microbial-Derived Metabolites in Immune-Related Adverse Events: Emergence of Novel Biomarkers for Cancer Immunotherapy. Int. J. Mol. Sci. 2023, 24, 2769. [Google Scholar] [CrossRef]
  245. Marshall, E.A.; Filho, F.S.L.; Sin, D.D.; Lam, S.; Leung, J.M.; Lam, W.L. Distinct Bronchial Microbiome Precedes Clinical Diagnosis of Lung Cancer. Mol. Cancer 2022, 21, 68. [Google Scholar] [CrossRef] [PubMed]
  246. Peters, B.A.; Pass, H.I.; Burk, R.D.; Xue, X.; Goparaju, C.; Sollecito, C.C.; Grassi, E.; Segal, L.N.; Tsay, J.-C.J.; Hayes, R.B.; et al. The Lung Microbiome, Peripheral Gene Expression, and Recurrence-Free Survival after Resection of Stage II Non-Small Cell Lung Cancer. Genome Med. 2022, 14, 121. [Google Scholar] [CrossRef] [PubMed]
  247. Yuan, X.; Wang, Z.; Li, C.; Lv, K.; Tian, G.; Tang, M.; Ji, L.; Yang, J. Bacterial Biomarkers Capable of Identifying Recurrence or Metastasis Carry Disease Severity Information for Lung Cancer. Front. Microbiol. 2022, 13, 1007831. [Google Scholar] [CrossRef]
  248. Chen, S.; Jin, Y.; Wang, S.; Xing, S.; Wu, Y.; Tao, Y.; Ma, Y.; Zuo, S.; Liu, X.; Hu, Y.; et al. Cancer Type Classification Using Plasma Cell-Free RNAs Derived from Human and Microbes. eLife 2022, 11, e75181. [Google Scholar] [CrossRef]
  249. Yuan, Q.; Wang, X.; Li, Z.; Guo, W.; Cheng, H.; Cao, Q. A Preliminary Study on Microbiota Characteristics of Bronchoalveolar Lavage Fluid in Patients with Pulmonary Nodules Based on Metagenomic Next-Generation Sequencing. Biomedicines 2023, 11, 631. [Google Scholar] [CrossRef]
  250. Kwok, B.; Wu, B.G.; Kocak, I.F.; Sulaiman, I.; Schluger, R.; Li, Y.; Anwer, R.; Goparaju, C.; Ryan, D.J.; Sagatelian, M.; et al. Pleural Fluid Microbiota as a Biomarker for Malignancy and Prognosis. Sci. Rep. 2023, 13, 2229. [Google Scholar] [CrossRef] [PubMed]
  251. Zhuo, M.; Chen, H.; Zhang, T.; Yang, X.; Zhong, J.; Wang, Y.; An, T.; Wu, M.; Wang, Z.; Huang, J.; et al. The Potential Predictive Value of Circulating Immune Cell Ratio and Tumor Marker in Atezolizumab Treated Advanced Non-Small Cell Lung Cancer Patients. Cancer Biomark. 2018, 22, 467–476. [Google Scholar] [CrossRef] [PubMed]
  252. Keup, C.; Kimmig, R.; Kasimir-Bauer, S. Liquid Biopsies to Evaluate Immunogenicity of Gynecological/Breast Tumors: On the Way to Blood-Based Biomarkers for Immunotherapies. Breast Care 2020, 15, 470–480. [Google Scholar] [CrossRef]
  253. Roweth, H.G.; Battinelli, E.M. Lessons to Learn from Tumor-Educated Platelets. Blood 2021, 137, 3174–3180. [Google Scholar] [CrossRef]
  254. Liu, L.; Lin, F.; Ma, X.; Chen, Z.; Yu, J. Tumor-Educated Platelet as Liquid Biopsy in Lung Cancer Patients. Crit. Rev. Oncol. Hematol. 2020, 146, 102863. [Google Scholar] [CrossRef]
  255. Calverley, D.C.; Phang, T.L.; Choudhury, Q.G.; Gao, B.; Oton, A.B.; Weyant, M.J.; Geraci, M.W. Significant Downregulation of Platelet Gene Expression in Metastatic Lung Cancer. Clin. Transl. Sci. 2010, 3, 227–232. [Google Scholar] [CrossRef] [PubMed]
  256. Sheng, M.; Dong, Z.; Xie, Y. Identification of Tumor-Educated Platelet Biomarkers of Non-Small-Cell Lung Cancer. OncoTargets Ther. 2018, 11, 8143–8151. [Google Scholar] [CrossRef] [PubMed]
  257. Best, M.G.; Sol, N.; Kooi, I.; Tannous, J.; Westerman, B.A.; Rustenburg, F.; Schellen, P.; Verschueren, H.; Post, E.; Koster, J.; et al. RNA-Seq of Tumor-Educated Platelets Enables Blood-Based Pan-Cancer, Multiclass, and Molecular Pathway Cancer Diagnostics. Cancer Cell 2015, 28, 666–676. [Google Scholar] [CrossRef]
  258. Best, M.G.; In’t Veld, S.G.J.G.; Sol, N.; Wurdinger, T. RNA Sequencing and Swarm Intelligence-Enhanced Classification Algorithm Development for Blood-Based Disease Diagnostics Using Spliced Blood Platelet RNA. Nat. Protoc. 2019, 14, 1206–1234. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The metastatic cascade and associated biomarkers. (1) During cancer cell proliferation, DNA and extracellular vesicles (EVs) may be released from necrotic or apoptotic cells. (2) Tumor-derived EVs promote tumor angiogenesis. Proteins such as matrix metalloproteinases (MMPs) and vascular endothelial growth factor (VEGF) contribute to angiogenesis by mechanisms such as degrading basement membranes and other extracellular matrix (ECM) components, allowing endothelial cells to detach and migrate into new tissue, and by releasing ECM-bound proangiogenic factors (bFGF, VEGF, and TGFβ). (3) EVs released by cancer cells contribute to endothelial-to-mesenchymal transition. MMPs and VEGF promote intravasation. (4) Platelets and neutrophils can aid circulating tumor cells (CTCs) in evading the immune system and surviving in the bloodstream by forming clusters or aggregates with CTCs. (5) Tumor-educated platelets (TEP) can facilitate the extravasation of CTCs by releasing EVs that can modulate the dynamics of endothelial cell junctions and increase the permeability of endothelial cells. (6) Tumor-derived exosomes contribute to the formation of the pre-metastatic niche and influence the site of metastatic colonization. VEGF is involved in the formation of pre-metastatic niches through direct and indirect mechanisms. Tumor- or host-cell-derived VEGF assists disseminated tumor cell colonization by neovascularization and generating a suppressive immune microenvironment. Moreover, in some cases, VEGF triggers the phenotypic switch from dormancy to proliferative status and restarts the colonization process.
Figure 1. The metastatic cascade and associated biomarkers. (1) During cancer cell proliferation, DNA and extracellular vesicles (EVs) may be released from necrotic or apoptotic cells. (2) Tumor-derived EVs promote tumor angiogenesis. Proteins such as matrix metalloproteinases (MMPs) and vascular endothelial growth factor (VEGF) contribute to angiogenesis by mechanisms such as degrading basement membranes and other extracellular matrix (ECM) components, allowing endothelial cells to detach and migrate into new tissue, and by releasing ECM-bound proangiogenic factors (bFGF, VEGF, and TGFβ). (3) EVs released by cancer cells contribute to endothelial-to-mesenchymal transition. MMPs and VEGF promote intravasation. (4) Platelets and neutrophils can aid circulating tumor cells (CTCs) in evading the immune system and surviving in the bloodstream by forming clusters or aggregates with CTCs. (5) Tumor-educated platelets (TEP) can facilitate the extravasation of CTCs by releasing EVs that can modulate the dynamics of endothelial cell junctions and increase the permeability of endothelial cells. (6) Tumor-derived exosomes contribute to the formation of the pre-metastatic niche and influence the site of metastatic colonization. VEGF is involved in the formation of pre-metastatic niches through direct and indirect mechanisms. Tumor- or host-cell-derived VEGF assists disseminated tumor cell colonization by neovascularization and generating a suppressive immune microenvironment. Moreover, in some cases, VEGF triggers the phenotypic switch from dormancy to proliferative status and restarts the colonization process.
Ijms 24 08894 g001
Table 1. Clinical utility of liquid biopsy biomarkers in cancer.
Table 1. Clinical utility of liquid biopsy biomarkers in cancer.
Body FluidBiomarkers DetectedExamples of Applications
PlasmactDNA to determine resistance mechanisms in patients with advanced NSCLC [50]
Prognostic biomarkers: miR-10b-5p, miR-23b-3p, and miR-21-5p [51]
Gene mutations including ALK, EGFR, KRAS with droplet digital PCR [52]
Plasma protein signatures reflect tumor biology [53]
Increased RP11-438N5.3 lncRNA levels for patients’ prognosis [54]
EGFR-status-related miRNA panel derived from exosomes isolated from metastatic disease [55]
SerumLevels of metabolites in disease progression: lactic acid, benzoic acid, and fumaric acid [56]
Prolactin, an early preventive factor in metastatic NSCLC for poor clinical outcomes [57]
High exosomal microRNAs in advanced disease and poor survival: miR-378 and miR-214 [58]
CA125 levels are an indicator of metastasis to the liver [59]
CYFRA 21-1 can be correlated with worsened PFS and OS in metastatic patients [60]
SputumChromosomal aneusomy by FISH predicts lung cancer incidence [61]
Sputum supernatant has comparable mutation profiling to plasma samples of NSCLC, with advantage of convenience in collection [62]
Use of nomograms to demonstrate the utility of sputum samples for genome profiling [63]
Sputum as an alternative source for somatic variation profiling with NGS [64]
BALCytologic examination of BAL has a comparable diagnostic yield to other endoscopic techniques for metastasis [65]BAL better reflect the cancer proteome than serum samples [66]
Pleural effusionMMP-9, cathepsin-B, C-reactive protein, chondroitin sulfate marker panel, and CA19-9, CA15-3, kallikrein-12 panel, are highly discriminative for malignant vs. tuberculous effusion, or lung adenocarcinoma vs. mesothelioma [67,68]Detection of extracellular vesicles and cfDNA in pleural effusions enhances EGFR genotyping of adenocarcinoma patients [69]
UrineEGFR mutations [70]ctDNA EGFR mutation testing detects T790M mutations overlooked in tissue biopsies due to sample quality or tumor heterogeneity [71]
ddPCR and Illumina MiSeq to monitor EGFR alterations during treatment [72]
CSFEGFR, ROS1, ALK, BRAF, and/or EGFR T790M mutations [35]Profiling for actionable mutation rate (EGFR, ROS1, ALK, BRAF) and resistance mutation rates (EGFR T790M mutation) [35]
Higher detection sensitivity for leptomeningeal metastasis by CTCs than with MRI [72]
Abbreviations: cfDNA: cell-free DNA; ctDNA: circulating tumor DNA; CTCs: circulating tumor cells; NSCLC: non-small-cell lung cancer; NGS: next-generation sequencing; lncRNA: long non-coding RNA; MS: mass spectrometry; BAL: bronchoalveolar fluid; CSF: cerebrospinal fluid; ddPCR: droplet digital PCR; FISH: fluorescence in situ hybridization; PFS: progression-free survival; OS: overall survival; MRI: magnetic resonance imaging.
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

Souza, V.G.P.; Forder, A.; Brockley, L.J.; Pewarchuk, M.E.; Telkar, N.; de Araújo, R.P.; Trejo, J.; Benard, K.; Seneda, A.L.; Minutentag, I.W.; et al. Liquid Biopsy in Lung Cancer: Biomarkers for the Management of Recurrence and Metastasis. Int. J. Mol. Sci. 2023, 24, 8894. https://doi.org/10.3390/ijms24108894

AMA Style

Souza VGP, Forder A, Brockley LJ, Pewarchuk ME, Telkar N, de Araújo RP, Trejo J, Benard K, Seneda AL, Minutentag IW, et al. Liquid Biopsy in Lung Cancer: Biomarkers for the Management of Recurrence and Metastasis. International Journal of Molecular Sciences. 2023; 24(10):8894. https://doi.org/10.3390/ijms24108894

Chicago/Turabian Style

Souza, Vanessa G. P., Aisling Forder, Liam J. Brockley, Michelle E. Pewarchuk, Nikita Telkar, Rachel Paes de Araújo, Jessica Trejo, Katya Benard, Ana Laura Seneda, Iael W. Minutentag, and et al. 2023. "Liquid Biopsy in Lung Cancer: Biomarkers for the Management of Recurrence and Metastasis" International Journal of Molecular Sciences 24, no. 10: 8894. https://doi.org/10.3390/ijms24108894

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