Liquid biopsy – a promising and effective method for surveying non-small cell lung cancer minimal residual diseases and anti-cancer drug response after treatment
-
Kaixun Xing
, Xiaoqing Li , Peng Liu , Yinghao Guo , Hetai Teng , Ting Li , Shuyan Dong , Hao Zhu , Shan Yu, Jian Ma
und Hongjiang He
Abstract
Non-small cell lung cancer (NSCLC), which accounts for 85 % of all lung cancers, has been the focal point of cancer research for years and is associated with a high morbidity and mortality worldwide. Minimal residual disease (MRD) and expansion of drug-resistant clones after treatment are the main causes of recurrence in patients with NSCLC. These residual low-level diseases, coupled with mechanisms such as primary drug resistance, immune escape, bypass activation, and tissue-type transformation, have led to difficulties in anticancer therapy over the past decades. Recently, liquid biopsy has emerged as a powerful tool for early cancer detection and monitoring of treatment outcomes. Studies have demonstrated its potential in adjuvant cancer therapy and highlighted its advantages of low risk, high sensitivity, and other merits over other technologies. In this review, we assessed the detection samples and methods used in this technology and discussed the methods for detecting MRD and the latest progress in liquid biopsy for dynamically assessing the response of NSCLC to anticancer drugs, focusing on the application of liquid biopsies in NSCLC. Finally, we provide insights into future directions of liquid biopsies. We found that this technology enables the detection of undetectable MRD, assesses the efficacy of anticancer treatments, and ultimately reduces NSCLC recurrence, highlighting its potential therapeutic application.
Introduction
Non-small cell lung cancer (NSCLC) is the leading cause of cancer-related death worldwide [1]. Notably, the 5-year survival rate of patients with NSCLC has remained as low as 16 % over the past 40 years [2], highlighting the unsatisfactory outcomes of targeted therapy and immunotherapy developed in the last decade. The low survival rates can be attributed to tumor recurrence and drug resistance.
Approximately 30 % of NSCLCs can be cured by surgery, however, the presence of a small number of tumor cells locally or distantly due to minimal residual disease (MRD) eventually leads to cancer relapse [3]. Although MRD is an effective indicator for monitoring recurrence, a widely accepted, accurate method to detect MRD is lacking [4]. Moreover, changes in patients’ condition and drug resistance during treatment warrant that the therapeutic regimen needs to be adjusted continuously. For instance, acquired resistance mechanisms and T790M status (40 % gain or loss mutations) change frequently in patients with NSCLC with epidermal growth factor receptor (EGFR) mutations during therapy [5]. In such cases, a single-site biopsy, which is subject to sampling bias, cannot provide an overall view of tumor genetic mutation due to the complexity and heterogeneity of cancer [6]. These shortcomings highlight the urgent need to develop effective methods to dynamically monitor anti-cancer drug response and sequence the cancer genome in order to institute “personalized medicine” for patients.
In this regard, liquid biopsy has emerged as a non-invasive detection technique, overcoming spatial and temporal heterogeneity, predicated on the analysis of various biologic elements such as circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), exosomes in the blood or other body fluids (e.g., pleural effusion [PE], saliva, urine, and cerebrospinal fluid). Nucleic acid-related substance detection typically employs polymerase chain reaction (PCR)-based multiplexing technology, and identification of CTCs and tumor-associated cells is utilized for the cell phenotyping systems. Liquid biopsy detection techniques based on PCR include the amplification refractory mutation system PCR, the Cobas® Liat PCR system, droplet digital PCR (ddPCR), and next-generation sequencing (NGS) technologies [7], 8]. Meanwhile, liquid biopsy techniques based on phenotypic detection, which rely on immune-affinity, biophysical properties, and microfluidic systems, are also available. However, the only liquid biopsy technique approved for large-scale application is CellSearch® [9].
Extracellular vesicles (EVs), including exosomes, and tumor-educated platelets (TEPs) can be analyzed using dynamic nuclear polarization-nuclear magnetic resonance (DNP-NMR), size-exclusion chromatography, and NGS methods (RNA-Seq) [10], [11], [12]. These latest technologies enable dynamic monitoring of NSCLC, which is conducive to early diagnosis and precise treatment. For instance, Wang et al. developed a quantitative detection method for CTC RNA using liquid biopsy technology. Their model demonstrated a high diagnostic performance for early-stage NSCLC (area under the receiver operating characteristic curve [AUC]=0.93, better than serum carcinoembryonic antigen [CEA] [AUC=0.70]). Similarly, another model based on this detection method can also assess the response of advanced NSCLC to different therapies [13]. Consequently, this information, combined with standard diagnostic methods, can provide detailed information in clinical practice, enabling early intervention for NSCLC and tailored treatment plans for advanced patients based on specific genetic mutations. However, the clinical research in the application of liquid biopsy for early cancer screening, adjusting therapeutic settings, and other aspects is still in the initial stages [12]. Factors such as low detection sensitivity or low accuracy, complicated operation, and high cost limit the development of liquid biopsy [14], 15]. The development of new technologies such as nanotechnology, microfluidics, sensor technology, and spectroscopy, as well as the complementary use of multiple analytes, contributes to improving the acceptance of liquid biopsy [16].
This study aimed to review the application of liquid biopsy technology in NSCLC. First, we introduce the concept of liquid biopsy and review the types of samples and the methods of its application. Subsequently, we discuss the methods for detecting MRD in NSCLC, as well as the latest advancements in liquid biopsy in the dynamic assessment of NSCLC’s response to anti-cancer drugs. Finally, we present our insights into the future directions of this technology.
An overview of liquid biopsy
Liquid biopsy is an emerging, personalized, non-invasive, highly efficient, and sensitive biochemical detection technology. Liquid biopsy samples include a variety of body fluids, including blood, chest and abdominal effusions, cerebrospinal fluid, saliva, urine, stool, and bronchoalveolar lavage fluid (BALF) [6], 17]. Tumor cells constantly die, shed, renew, and release DNA, RNA, proteins, and other metabolites into the peripheral circulation [6]. Based on these dissociated products, liquid biopsy can detect diversified tumor-related substances, including CTCs, ctDNA, circulating tumor RNA (ctRNA, predominantly non-coding RNA: microRNA [miRNA], long non-coding RNA [lncRNA], circular RNA [circRNA]), exosomes, TEPs, tumor endothelial cells (TECs), and circulating genetically abnormal cells (CACs) [18], 19].
Notably, liquid biopsy has significant potential applications in cancer because of its ability to obtain rich substance samples dynamically and non-invasively. Moreover, compared to tissue biopsy, this method enables repeated sampling while avoiding sampling bias due to tumor complexity and heterogeneity [20], [21], [22]. Therefore, the use of liquid biopsy is rapidly being evaluated for differentiation between benign and malignant diseases, early cancer screening, cancer genetic sequencing, determination of prognosis and risk classification, monitoring of therapy efficacy, and detection of drug resistance mechanisms [23], [24], [25]. However, the shortcomings of liquid biopsy, such as low sensitivity, low specificity, complex procedures, and high costs, limit its large-scale clinical application, while tissue biopsy and conventional imaging tests overcome these limitations [26], [27], [28].
Based on PCR, liquid biopsy detection technologies include Amplification Refectory Mutation blocking system PCR, the Cobas® Liat PCR system, ddPCR, and NGS technologies, among which, the NGS technology is most widely used [29], 30]. Cell phenotype analysis is used to detect CTCs and tumor-associated cells, with the CellSearch System currently achieving widespread clinical applications [9]. Technologies, such as DNP-NMR, have been employed for the detection of exosomes and other EVs [31], 32] (Figure 1).

Diverse detection targets used in liquid biopsy and the corresponding detection methods. The source of samples of liquid biopsy includes a variety of body fluids, blood, chest and abdominal effusions, cerebrospinal fluid, saliva, urine, stool and BALF. Diversified tumor-related substances, including CTCs, ctDNA, ctRNA (predominantly non-coding RNA: miRNA, lncRNA, circRNA), exosomes, TEPs, TECs, and CACs were used as detection targets in liquid biopsy. PCR-based multiple techniques were usually used for detecting nucleotide-related substances, and cell phenotype detecting system was used to detect CTCs and tumor-related cells. Note: BALF, bronchoalveolar lavage fluid; CTC, circulating tumor cell; ctDNA, circulating tumor DNA; ctRNA, circulating tumor RNA; miRNA, microRNA; lncRNA, long non-coding RNA; circRNA, circular RNA; TEP, tumor-educated platelet; TEC, tumor endothelial cell; CAC, circulating genetically abnormal cell; PCR, polymerase chain reaction; NGS, next-generation sequencing; DNP-NMR, dynamic nuclear polarization-nuclear magnetic resonance.
Application of liquid biopsy in NSCLC
Liquid biopsy is useful throughout all stages of NSCLC from diagnosis to therapy, including early cancer screening, monitoring of tumor burden and MRD, and detection of molecular resistance. For instance, Liang et al. established a model for high-throughput targeted DNA methylation sequencing using plasma ctDNA and reported a sensitivity of 75.0 % (55.0–90.0 %) and 85.7 % (57.1–100.0 %) in patients with IA and IB lung cancer, respectively. Consequently, ctDNA can aid in the early diagnosis of lung cancer [33]. Another recent clinical study showed that patients with NSCLC in whom CTCs were detectable prior to stereotactic body radiation therapy (SBRT) but undetectable after treatment had a two-year freedom from local and nodal recurrence, whereas patients with a high pre-SBRT and a persistent CTC count after treatment had a regional and distant recurrence [34]. This result highlights the significance of CTCs in local recurrence and distant metastasis.
Notably, the detection of molecular subsets in NSCLC is vital for the development of effective and precise targeted therapies. Patients cannot undergo single or multiple tissue biopsies due to several reasons, such as unbearable physical condition, diffuse small nodules, or deep metastatic lesions [35]. Although liquid biopsy cannot replace tissue biopsy, it is recommended in such cases under the updated College of American Pathologists (CAP)/International Association for the Study of Lung Cancer (IASLC)/Association for Molecular Pathology (AMP) guidelines for molecular testing of patients with NSCLC [36]. Moreover, liquid biopsy helps detect MRD (at the molecular level, not visible on imaging), which has the ability to assess the risk of recurrence [4], and analyze the mechanisms of acquired resistance, one of the biggest hurdles to targeted lung cancer therapy [37]. In the current review, we focused on how liquid biopsies detect MRD and evaluated the efficacy of anticancer drugs in detail. We developed a comprehensive flowchart illustrating liquid biopsy procedures to enhance comprehension. The chart systematically maps sample-test compatibility and correlates specific specimen types with their respective assay methodologies while clearly delineating the target patient populations for each analytical approach [38] (Figure 2).

Process and application of liquid biopsy in NSCLC. Blood was the most commonly collected specimen. The BALF, urine, and PE can also be used to treat MRD. CtDNA can be used for the early screening of NSCLC and the detection of high-risk individuals. This is helpful in predicting tumor and treatment responses. The CellSearch System is helpful for predicting NSCLC and its response to treatment. EVs are typically used in complex situations. It can be combined with other markers to increase the detection rates. Liquid biopsy can be conducted multiple times during the entire course of NSCLC and can dynamically analyze disease status. Note: BALF, bronchoalveolar lavage fluid; PE, pleural effusion; MRD, minimal residual disease; ctDNA, circulating tumor DNA; NSCLC, non-small cell lung cancer; ddPCR, droplet digital polymerase chain reaction; NGS, next-generation sequencing.
Liquid biopsies of peripheral blood detect MRD in NSCLC
The 5-year recurrence rate of patients with NSCLC after surgery ranges from 20 % for stage I to 50 % for stage III [39], in which MRD is not visible on radiological imaging. MRD is the main cause of distant metastasis and “culprit” for tumor distant metastasis and recurrence [40]. In this regard, the development of sophisticated liquid biopsies has increased the prospect of accurately detecting MRD at the molecular scale (Table 1).
Methods of liquid biopsy in NSCLC.
| Biopsies | Sources | Advantage | Challenge | Applicability | Ref |
|---|---|---|---|---|---|
| CTC | Peripheral blood | The extracted CTCs are highly specific. | NSCLC cells undergo epithelial-mesenchymal transition, and some characteristics may change. | Predicting genetic mutations, forecasting lesion dimensions, guiding therapeutic strategies, and anticipating recurrence risks. | [9], 13] |
| BALF | The number of CTCs detected in BALF was significantly higher than that in peripheral blood. They are obtained from exfoliated NSCLC cells and could retain their original characteristics. | Microbial contamination, probably. | Predicting the characteristics of NSCLC. | [41] | |
| ctDNA | Peripheral blood | High sensitivity for the detection of genomic changes such as gene mutation and fusion. | Difficulty distinguishing the source (normal or NSCLC cells). | Predicting genetic mutations, forecasting lesion dimensions, guiding therapeutic strategies, and anticipating recurrence risks. | [33], 42], 43] |
| PE | The total concentration of ctDNA in PE was higher than that of plasma. | Not suitable for patients with no PE and small volume. | Monitoring the advancement of NSCLC has advantages in detecting MRD. | [44] | |
| Urine | The amount of ctDNA extracted is higher because the kidneys filter through more stuff. | Contaminants skew the results and increase the risk of false positives. | Predicting recurrence and reducing protein interference. | [45] | |
| Exosome | Peripheral blood | The double lipid layer protects the internal material from degradation. | Difficulty distinguishing the source (normal or NSCLC cells). | Complementing other detection methods to enhance efficiency. | [46], 47] |
-
CTC, circulating tumor cell; BALF, bronchoalveolar lavage fluid; PE, pleural effusion; NSCLC, non-small cell lung cancer; ctDNA, circulating tumor DNA; MRD, minimal residual disease.
Using peripheral blood CTC to detect MRD
Captured CTCs can be used to analyze protein expression and subcellular localization along with single-cell analysis of tumor heterogeneity [48], 49], and this is because the molecular features of CTCs are correlated with those in primary tumors [50]. For example, EGFR mutations between CTCs and primary tumors are mutable but highly consistent in patients with advanced NSCLC [51]. Additionally, a meta-analysis of 20 trials involving 1,576 patients with NSCLC showed a significant association between the presence of CTCs and reduced disease-free survival (DFS) and overall survival (OS) [52]. Therefore, monitoring CTCs as an indicator of MRD contributes to predicting recurrence and assessing the risk of metastasis.
Surgery is the primary treatment for patients with early-stage (stages I to III) NSCLC, as recommended by the National Comprehensive Cancer Network (NCCN) [53]. During the perioperative period, the CTC count fluctuates in patients with NSCLC who were surgically treated. Additionally, several studies have confirmed that a high pre-/intra-/post-operative CTC count is associated with a poorer prognosis [54], [55], [56]. Consequently, a clinically significant prognostic threshold of ≥5 CTCs per 7.5 mL of blood has been established for NSCLC prognosis using the CellSearch System [57]. Based on this, the CTCs in peripheral blood samples from 56 NSCLC patients who underwent radical surgery were assessed before and one month after the surgery. The presence of CTC at one month following surgery was found to be substantially linked with a shorter DFS (hazard ratio [HR]=5.75; 95 % CI: 1.50–21.946; p=0.010), according to multivariate analysis [58].
The detection of CTCs is a demanding technology due to the small CTC count, as few as one per 106-107 peripheral blood mononuclear cells, in patients with solid tumors [59], 60]. Similarly, previous studies have reported that the detectable CTC levels in the pulmonary vein blood were higher than those in the peripheral blood before surgery, and that the number of CTCs was associated with tumor size [61]. Furthermore, compared to the resected main tumor, early dispersed CTCs exhibited a larger mutational overlap when metastases occurred 10 months later [49]. These results provide novel insights into CTC detection. Consequently, further extensive prospective investigations are required to validate these findings.
In patients with NSCLC receiving chemoradiotherapy, CTCs monitoring serves as an adjunct guide for treatment. A small retrospective study showed that CTCs could be detected before the advent of radiological evidence of recurrence in patients with NSCLC (stage II–III) treated with chemoradiotherapy [62]. Another clinical trial of 101 patients with NSCLC receiving chemotherapy showed that the CTCs count decreased after administration of chemotherapy, which was associated with longer progression-free survival (PFS) (5.4 vs. 1.9 months; p<0.001) and OS (8.3 vs. 3.3 months; p<0.001) [57]. Moreover, while a decrease in CTCs after chemotherapy indicates remission, an increase indicates reactivation, which would lead to treatment adjustment [63].
When at least two CTCs aggregate by physical contact through intercellular junctions, they form clusters, also known as circulating tumor microemboli [64], 65]. CTC clusters have greater metastatic potential than individual CTCs, and may be protected from immune invasion and survive longer because of the shape of the clusters [66], 67]. Accordingly, one study recommended that the percentage of cluster CTCs was significantly correlated with the TNM stage. In addition, 9 out of 36 patients with NSCLC had post-operative recurrence, and at least one CTCs cluster was detected in six of them. Therefore, future studies should investigate the potential of using CTC clusters as biomarkers for monitoring NSCLC recurrence [68].
Using peripheral blood ctDNA to detect MRD
CtDNA is a small fragment of DNA resulting from tumor cells necrosis, apoptosis, or secretion and is released into the peripheral circulation at approximately 90–150 base pairs in length [69]. The DNA fragment can be stably detected in peripheral blood with an abundance of ≥0.02 %, including lung cancer driver genes and other class I/II gene variants. Studies have shown that ctDNA is highly consistent with tumor DNA [70], 71] and demonstrated a linear relationship between ctDNA and tumor volume [72].
Consequently, multiple retrospective analyses have confirmed that the median predictive time from post-operative dynamic monitoring of ctDNA to clinical or radiographic reports of lung cancer recurrence is 70 days to 5.2 months [43], 73], 74]. Tracking NSCLC evolution through therapy (Rx) (TRACERx) [75], by analyzing the blood of 24 patients with NSCLC after surgery, researchers accurately identified more than 90 percent of them as destined to relapse a year before clinical imaging confirmed recurrence. Additionally, >75 % of the tumors exhibited variants of PIK3CA, NF1, and genes involved in chromatin modification and DNA damage repair. Moreover, amplification of CDK4, FOXA1, and BCL11A was associated with an increased risk of death (HR=4.9; p=4.4 × 10−4). Notably, this time gap between relapse prediction and occurrence offers opportunities for clinical intervention and management.
A prospective multicenter cohort study analyzed perioperative ctDNA in 950 plasma samples from 330 patients with stage I–III NSCLC obtained at three different perioperative time points with NGS (before surgery, three days, and one month after surgery). The samples were utilized for ctDNA-based MRD analysis and demonstrated that the presence of MRD (ctDNA positivity at postoperative three days and/or one month) was a substantial predictor for disease relapse (HR=11.1; 95 % CI: 6.5–19.0; p<0.001), and that preoperative ctDNA positivity was linked with poorer recurrence-free survival (RFS; HR=4.2; 95 % CI: 2.6–6.7; p<0.001). The three tumor driver genes with the highest mutation frequencies were EGFR (64 %), TP53 (49 %), and RBM10 (17 %). Similarly, another study showed that the likelihood of recurrence in MRD-positive patients detected using these ctRNAs was 11 times higher than that in MRD-negative patients [42]. Another study analyzed cancer personalized profiling (CAPP)-seq personalized profiled ctDNA from 255 samples (patients with stage I–III lung cancer and healthy individuals) using deep sequencing. Receiver operating characteristic (ROC) analysis revealed an area under the curve of 0.97, with maximal sensitivity and specificity of 93 and 96 % respectively [73]. In another study that assessed MRD using ctDNA from 77 patients with NSCLC with a preoperative ctDNA-positive status demonstrated a significant 3.8–4.0-fold risk of recurrence and death, which was also associated with a lower RFS (HR=3.812; 95 % CI: 1.681–8.855; p=0.0005) and OS (HR=5.004; 95 % CI: 1.731–14.756; p=0.0009). The most common mutations were in TP53 (60 %), EGFR (21 %), and KEAP1 (9 %). Furthermore, the risk of recurrence, metastasis, or death in preoperative ctDNA-positive patients increased by 3.4-fold and 4.0-fold, respectively, compared with that in ctDNA-negative patients [43]. Therefore, detecting MRD using ctDNA is of great value for predicting recurrence and survival rates.
One of the biggest advantages of liquid biopsy over tissue biopsy is that it can be performed repeatedly for dynamic monitoring to further improve MRD surveillance [12]. A prospective study of surgical patients with lung cancer that investigated perioperative dynamic changes in ctDNA demonstrated that ctDNA half-life in patients with MRD was significantly longer than in those without MRD (103.2 min vs. 29.7 min, p=0.01) and determined the appropriate detection time of ctDNA-based surveillance to be the third day after R0 [76]. In another clinical study, patients with NSCLC with ctDNA detected during the post-treatment monitoring period had lower DFS than those with consistently negative ctDNA after surgery (HR=8.5; 95 % CI: 3.7–20; p<0.001). In addition, 79 % of relapsed patients tested positive for at least one ctDNA during disease surveillance. The top three ctDNA-positive findings are EGFR (55.4 %), KRAS mutation (16.1 %), and ALK fusion (8.9 %) [40]. Consequently, the longitudinal ctDNA monitoring is efficient in identifying relapsed patients. Moreover, patients with lung cancer receiving immune checkpoint inhibitors undergo dynamic monitoring of ctDNA and benefit from the detection of MRD for clinical intervention. For instance, ctDNA tracking was performed in 31 patients with NSCLC after a median of 26.7 months of programmed cell death receptor-1/the anti-programmed death 1 ligand (PD-1/PD-L1) blocking therapy. CtDNA was undetectable at the time of monitoring in 27 patients, among whom 93 % (25/27) did not progress. In addition, all four patients with detectable ctDNA experienced disease progression [77]. Moreover, it is difficult to monitor a patient’s condition after immunotherapy, since no clinical symptoms are visible. Therefore, the use of ctDNA provides an opportunity to administer therapy earlier in patients with the potential to relapse.
Using peripheral blood exosomes to detect MRD
The contents of cancer cells carried in exosomes represent critical cancer-specific biomarkers that help detect and analyze the induction of angiogenesis and the suppression of immune responses, migration, and metastasis [78]. A unique advantage of exosomes biopsy is that biological molecules are protected by a lipid bilayer membrane of exosomes that confers a high degree of stability and can be isolated and analyzed [79]. Different RNAs and proteins carried in exosomes have been demonstrated to indicate the poor prognosis of patients with NSCLC, such as miR-17-3p, miR-21, lncRNA RP5-977B1, lncRNA SNHG15, and the eukaryotic translation initiation factor 4G2 [80], [81], [82]. Thus, the detection of such biological information would help integrate MRD detection and risk of recurrence to adjust different clinical treatments. Although ctDNA and CTCs are the mainstream methods for monitoring MRD, the addition of exosomes can achieve complementarity [46]. In addition, combining exosomal RNA (exoRNA) with cell-free DNA (cfDNA) can enhance the identification of EGFR mutations in patients with NSCLC. Therefore, MRD and EGFR mutations can be detected simultaneously [47].
Liquid biopsies of non-blood liquid detect MRD in NSCLC
As some biomarkers may be present in higher amounts in non-blood liquids than in the peripheral blood, liquid biopsy of tumor biomarkers in different bodily fluids is progressively bringing optimism [83]. PE usually occurs in advanced patients with advanced NSCLC, and is currently detected using ctDNA and exosomes. Tong et al. analyzed 30 patients with NSCLC and found that, compared with the control group, the median concentration of ctDNA in the PE supernatant was 278.1 ng/mL, which was significantly greater than the median concentration in the plasma (20.4 ng/mL, p<0.001). Moreover, the detection rate of EGFR-driven mutations was the highest. Other mutations included ALK, BRAF, EGFR, ERBB2, KRAS, KRAS, NF1, PIK3CA, and RET. The results showed that monitoring MRD using ctDNA in PE has the advantage of the abundance of ctDNA [44]. Similarly, Zhong et al. found the number of CTCs detected in BALF was significantly higher than that in peripheral blood CTCs (6.76 ± 0.89 vs. 5.78 ± 0.57, p=0.016) with the same sample size, as well as other biomarkers used to diagnose recurrence, such as NSE, CEA, and CA125 (p<0.05) [41]. CTCs moving from tumors to peripheral blood undergo complex processes such as epithelial-mesenchymal transition, while in BALF, they are directly derived from the primary lesions, retaining original tumor cell characteristics and releasing metabolites. Based on these results, monitoring MRD by detecting CTCs in the BALF is a promising alternative to monitoring peripheral blood. Additionally, liquid urine biopsy is an effective tool for monitoring MRD in patients with NSCLC. One study on urinary ctDNA profiling in EGFR-positive patients with NSCLC used ddPCR sed to detect mutant EGFR in all peel samples collected at various time points. Researchers noticed a notable increase in positive EGFR mutations in urine samples after a 6-month treatment period. Ninety-one percent of individuals with higher urine DNA concentrations experience recurrence [45]. As the kidneys filter out many proteins, it is simpler to extract DNA from urine samples, which may lessen PCR amplification interference [83].
Assessment of anti-cancer drug response after treatment by liquid biopsy
Currently, NSCLC is treated according to different pathological stages and molecular types, such as surgery, chemotherapy, radiotherapy, targeted therapy, and immunotherapy. In this regard, liquid biopsy has been used to non-invasively and dynamically evaluate the efficacy of anti-tumor drugs, drug resistance, and adverse events, and to select the most beneficial patient group suitable for prescribed medicine (Table 2).
The role of different liquid biopsy methods in the treatment of lung cancer.
| Therapies | Biopsies | Influences | Ref |
|---|---|---|---|
| Chemotherapy | CTCs | Subtypes and expression content are associated with PFS. | [84], [85], [86] |
| ctDNA | Detection of post-chemotherapy mutation sites, standing for PFS. | [87], [88], [89] | |
| Exosome/circulating miRNA | Transmit drug resistance and reflect chemosensitivity. | [90], [91], [92] | |
| CECs | The count was correlated with PFS. | [93] | |
| TKIs | ctDNA | Discovery of resistance mechanisms | [94], 95] |
| Monitoring efficacy | [96] | ||
| Exosome | miRNA is linked to PFS and plays a role in treatment resistance. | [97], [98], [99] | |
| Antiangiogenic drugs | CEPCs | Correlated with the histological subtype of lung cancer, solid type can be identified | [100] |
| CECs | The count and PFS showed a correlation. | [101], 102] | |
| CTC, CTEC | Monitoring efficacy | [103] | |
| Immunotherapy | ctDNA | TMB can reflect PFS | [104] |
| Evaluating patients with DCB in synergy with other indicators | [105], 106] | ||
| CTCs | Monitoring the expression of PD-1 dynamically | [107] |
-
CTC, circulating tumor cell; PFS, progression-free survival; ctDNA, circulating tumor DNA; miRNA, microRNA; TKI, tyrosine kinase inhibitor; CEC, circulating endothelial cell; CEPC, circulating endothelial progenitor cell; CTEC, circulating tumor endothelial cell; TMB, tumor mutational burden; DCB, durable clinical benefit; PD-1, programmed cell death receptor-1.
Evaluation of chemotherapy outcome by liquid biopsy
In patients with NSCLC receiving platinum-based therapy, researchers have investigated different subtypes of CTCs using multiple immunofluorescence stains. The results have demonstrated a significant relationship between an increasing ratio of stem cell-like (CD133-positive) to epithelial cells (pan-CK-positive) and mesenchymal N-cadherin-cells (2 vs. 8 months, p=0.003, HR=4.43; 5 vs. 8 months, p=0.03, HR=2.63) [84]. In addition, the expression profiles associated with CTCs provide promising results for the evaluation of platinum-based efficacy and prognosis in patients with NSCLC. For example, the enzyme excision repair cross-complementation group 1 (ERCC1) can remove cisplatin-induced DNA adducts. In a small-scale clinical trial of 17 metastatic patients with NSCLC receiving platinum-based therapy, Das et al. detected the expression of ERCC1 in CTCs and found that increased ERCC1 expression in CTCs correlated with decreased PFS (p<0.04, F-test, linear regression) [108]. Similarly, metastasis and recurrence were linked to higher expression levels of epithelial cell adhesion molecule (EpCAM) on CTCs, in 76 patients with NSCLC receiving postoperative adjuvant chemotherapy, which occurred before CT imaging and diagnosis. Particularly, a statistically significant difference (p=0.008) was observed in the EpCAM expression in CTCs one day prior to the fourth adjuvant treatment [85]. Pt-(GpG) intrastrand crosslinks are one of the major DNA adducts formed by platinum. Previous studies have shown that Pt-(GPG) adducts can be detected in peripheral blood CTCs of patients with NSCLC, and that a substantial difference exists between the effective and ineffective groups [109]. Moreover, platinum resistance mutations can be identified by assessing genetic information from CTCs. Accordingly, CTCs from nine patients with platinum-resistant advanced NSCLC were isolated and subjected to whole-exome sequencing (WES) at the single-cell level. Functional annotation demonstrated that cancer-driver mutations, including EGFR and TP53, and cell cycle-regulated or stem cell-related gene mutations, including SHKBP1, NUMA1, ZNF143, MUC16, ORC1, PON1, and PELP1, were found in CTCs and were to chemotherapy resistance and metastasis [86].
CfDNA has been used to detect mutation sites because it can be conveniently extracted dynamically and noninvasively. For instance, in a study involving 57 patients with NSCLC receiving platinum-based chemotherapy and 13 patients receiving neoadjuvant chemotherapy, WES was used to identify somatic mutations in 225 genes. The platinum response was found to be associated with copy number variations in chromosomes 8q24.3 and 22q11.21, as well as non-synonymous variants in EGFR, TTN, TP53, and KRAS. Additionally, the failure of DNA double-strand breaks and calcium signaling pathways is correlated with the mutational signatures of these variants [87]. In a similar study of cfDNA mutations detected by WES, researchers observed 1,559 point mutations across 98 plasma samples and found that the whole mutation burden decreased after platinum-based chemotherapy (p=0.034). Moreover, fewer non-silent mutations were detected after chemotherapy in patients with a better platinum therapeutic response than in insensitive patients, which reflected the sensitivity of platinum chemotherapy. In addition, some driver mutations, such as TP53 (p.K159X) and EGFR (p.E709 K and p.G719A), were undetectable, while some hotspot mutations, such as EGFR (L858R), KRAS (p.G12C), persisted after platinum chemotherapy [88]. With an in-depth exploration of the longitudinal monitoring of ctDNA, researchers found that ctDNA levels peaked at a median of 7 h after the beginning of chemotherapy (Interquartile Range [IQR]: 2–26 h). CtDNA was detected at baseline in 75 % of patients receiving systemic chemotherapy; CTCs were detected at baseline and had post-treatment genomic alterations in 28 % of patients. The abundance of ctDNA in the plasma increased due to tumor lysis when tested immediately after the initiation of treatment, allowing the detection of genetic changes hidden in baseline testing. Furthermore, experimental data have demonstrated that cfDNA can help predict the prognosis of patients receiving platinum-based chemotherapy. A previous multivariate analysis reported that a higher baseline concentration of cfDNA was an independent prognostic factor, with patients with high cfDNA concentrations at baseline showing poorer DFS and OS than those with lower concentrations (p=0.001) [89].
Exosomes have been confirmed to confer chemoresistance to cancers through the transfer of miRNAs. Exosomal miRNAs are easy to extract and measure from peripheral blood, making them potential biomarkers for predicting chemotherapy response and resistance. Ma et al. observed that individuals tolerant to chemotherapy drugs had higher baseline levels of miR-425-3p in their exosomes. Exosomal miR-425-3p targets AKT1 to promote autophagic activation of recipient cells, ultimately leading to chemoresistance [90]. Additionally, miR-1273a was the most significantly downregulated miRNA in the cisplatin-treated exosomes by microarray analysis. Downregulation of exosomal miR-1273a upregulates the expression of syndecan-binding proteins, eventually increasing cisplatin resistance in NSCLC [91].
Furthermore, an increasing number of studies have suggested that circulating miRNAs are correlated with the prognosis of patients with NSCLC receiving platinum-based chemotherapy. This suggests that they are potential biomarkers for predicting drug response and survival. Accordingly, in an investigation of the expression and clinical significance of different plasma miRNAs in patients who received platinum-based therapy, researchers found that high expression of miR-128 and miR-155 resulted in shorter OS than those with low expression (p<0.05). MiR-128 promotes epithelial-mesenchymal transition and cell migration targeted by Drosha ribonuclease III and Dicer ribonuclease III in NSCLC [110]. The carcinogenic effect of miR-155 is correlated with the inhibition of PTEN and cytokine signaling 1 and 6 [111]. Circulating miRNAs involved in macrophage polarization function were predictive variables in 125 patients with NSCLC treated with first-line platinum-based therapy. Particularly, high miR-202 expression was an independent prognostic factor for shorter PFS (p=0.021) and OS (p=0.024) [92].
Pemetrexed inhibits thymidylate synthase and folate-dependent enzymes, interfering with the biosynthesis of thymidine and purine nucleosides, eventually blocking tumor growth [112]. However, no reliable biomarker is currently available for determining the effectiveness of pemetrexed-based chemotherapy. Therefore, Franchina et al. determined the expression levels of several circulating miRNAs that are possibly involved in the folate pathway in 22 patients with NSCLC treated with pemetrexed-based chemotherapy and 27 healthy controls. Patients with progressing diseases had considerably higher miR-22 expression than those without disease progression (p=0.03). Thus, the correlation between high expression and a lack of response in patients indicates that miR-22 is a predictive biomarker for pemetrexed-based chemotherapy [113]. Accordingly, researchers investigated the relationship between circulating endothelial cells (CECs) counts and the efficacy of platinum plus pemetrexed first-line chemotherapy in 69 patients with NSCLC. Peripheral blood CECs were detected at baseline, and after the second and third cycles. The results showed that the CEC count increased by >50 % between the first and second cycles and was significantly associated with disease progression (p=0.008). Additionally, patients with baseline CEC counts above average (>153 cells/4 mL) had longer PFS and OS (with no statistically significant difference) [93].
The role of liquid biopsy in tyrosine kinase inhibitor treatment
Tyrosine kinase inhibitors (TKIs) are generally prescribed for patients with metastatic NSCLC (stage III or IV) and certain genetic alterations. EGFR mutations are the most common genetic alterations in patients with NSCLC. Liquid biopsy is used in TKI therapy to select the targeted therapy according to the type of molecular mutation, monitor efficacy, and detect drug resistance mechanisms [114].
Deep sequencing of plasma DNA has been used to detect EGFR mutations in ctDNA. When tested in 288 patients with NSCLC, the diagnostic specificity of the exon 19 deletion was 98.0 % and the specificity of L858R was 94.1 %. Thus, owing to their high specificity, EGFR-TKIs can be directly recommended based on positive plasma DNA results [115]. Conversely, tissue biopsies and ctDNA detection yielded false-negative results for EGFR mutations, which may be partly due to the release, distribution, and clearance of ctDNA. For instance, 28/1,017 (2.75 %) patients with NSCLC with EGFR mutation were not detected via tissue biopsy but were detected using ctDNA, which may be correlated with the spatial heterogeneity of the tumor. The PFS of patients who received EGFR TKIs was higher in that patient group than that of those who received chemotherapy, thus identifying that this group had special therapeutic significance. Currently, if ctDNA fails to detect EGFR mutations, tissue biopsy is used as a supplementary test, which may save approximately 30 % of tissue samples [116].
Furthermore, resistance to EGFR-TKIs, driven by various genes and different genomic alterations, has been confirmed using NGS of cfDNA [94]. In addition to EGFR T790 and C797S mutations, rare EGFR mutations or other resistance mechanisms, such as G724S, and L858R/A859S/Y891D triple mutations, FGFR3-TACC3 fusions, and NTRK1 fusion also exist [95], 117], 118]. EGFR mutations coexisting with EML4-ALK gene translocations in ctDNA were first reported in a patient with NSCLC with multiple metastases [119]. Therefore, dynamic monitoring of EGFR-TKI resistance can identify drug resistance sites and predict treatment failure in a timely manner, thus providing a window of opportunity for intervention.
A previous study showed that a decline in ctDNA concentration 48 h after the initiation of EGFR targeted therapy identified good responders to TKI and had longer PFS (14.7 months vs. 8.5 months, p=0.013) than patients with stable or increased ctDNA levels [96]. Thus, the detection of EGFR mutations by ctDNA can predict patient outcomes and assist in the timely adjustment of the appropriate treatment regimen for patients receiving EGFR-TKIs. Moreover, persistent EGFR mutations in ctDNA at weeks 3 and 6 after osimertinib administration predicted shorter PFS in patients who were considered to benefit from additional chemotherapy afterwards [91]. Therefore, it is possible to select appropriate EGFR-TKI therapeutic regimens for patients with NSCLC based on ctDNA levels and identify patients with significant improvements in survival during treatment. Accordingly, in an analysis of 830 plasma samples from 228 patients with stage IV EGFR-positive NSCLC who received first-line osimertinib (3rd generation TKI) or sequential TKI therapy, the OS of the two treatment plans barely differed. Furthermore, the low-risk patients identified receiving sequential TKI therapy at a mutant allele frequency (MAF) <7 % at diagnosis and the high responders who were ctDNA negative after 3 or 6 months of treatment and with an MAF <7 % before treatment had two-thirds lower risk of death than in the opposite case. Consequently, sequential treatment may be advantageous for strong responders and low-risk patients [120].
The First-Line AUtopia of osimertinib in EGFR-mutated NSCLC (FLAURA) study confirmed that EGFR-mutated NSCLC patients treated with osimertinib as first-line treatment had 6.8 months longer OS than patients treated with gefitinib or erlotinib. Unfortunately, resistance is inevitable, and the spectrum of its mechanisms is broadly heterogeneous [121], 122]. Currently, elevated levels of exosome-derived miRNAs, such as miR-210-3p, miR-184, miR-3913-5p, and miR-494-3p, which are involved in epithelial-mesenchymal transition, activation of bypass pathways (RAS-MAPK path anomaly and PI3K pathway activation), and other potentially unknown mechanisms, are capable of participating in osimertinib resistance since their elevated plasma levels corresponded to poor patient outcomes [97], [98], [99].
Liquid biopsy and antiangiogenic drugs
Given the importance of angiogenesis in tumors, CECs and circulating endothelial progenitor cells (CEPCs) have been used to evaluate tumor angiogenic activity and the efficiency of antiangiogenic drugs.
Different lung adenocarcinoma histological subtypes show differences in angiogenic status. The CEPC count and the level of vascular endothelial growth factor (VEGF) were higher in solid adenocarcinomas than in non-solid adenocarcinomas (p<0.001, p=0.005). Therefore, identifying patients with solid adenocarcinoma is crucial because they may benefit from anti-angiogenic therapy [100].
CEC count was confirmed to be correlated with PFS in antiangiogenic therapy. For instance, docetaxel and bevacizumab have been demonstrated to synergistically reduce endothelial cell proliferation and inhibit CECs mobilization. Researchers estimated the level of CECs on days 1 and 8 and found that patients with ≥10 count increase in CECs had longer PFS than patients with <10 count increase in CECs (median PFS of 11.0 vs. 6.90 months, respectively) [101]. Furthermore, 49 patients with NSCLC with anlotinib were stratified according to the ratio of minimal CECs counts to baseline (CECs min/baseline) as <1 or ≥1. Subsequently, CECs were identified using flow cytometry as CD31+ cells and CD105+ cells. The results revealed that patients with CECs (CD31+) min/baseline <1 had longer PFS (p<0.05) than those with CECs (CD31+) min/baseline ≥1. In addition, CD31 showed more responsive alterations than CD105 through the PI3K-AKT pathway, supporting CD31+ CECs as a more sensitive marker for predicting the efficacy of anlotinib treatment [102]. Another study showed that pre-therapeutic patients with vimentin (Vim) + CTECs at baseline showed significantly shorter median progression-free survival (mPFS) than those with Vim-CTECs. Post-therapy patients with epithelial cell adhesion molecule (EpCAM) + CTCs and CTECs, regardless of Vim expression, showed significantly reduced mPFS [103]. Thus, comprehensive co-detection of various subtypes and karyotypic molecular characterization of CTCs and circulating tumor endothelial cells (CTECs) may be useful tools for prognosticating and tracking the effectiveness of treatment in patients with NSCLC receiving bevacizumab with chemotherapy.
Evaluation of immunotherapy by liquid biopsy
Immune checkpoint inhibitors (ICIs), including antibodies targeting PD-1, PD-L1, and cytotoxic T-lymphocyte antigen 4 (anti-CTLA-4), have been approved as first-line treatments for NSCLC or after progression to chemotherapy [123], 124]. Liquid biopsy may help with screening patients who can benefit from ICIs or who are prone to immune-related adverse events (irAEs), in addition to dynamically monitoring the efficacy of ICIs.
Non-synonymous mutations in tumor cells lead to neoantigen production, which is associated with increased tumor immunogenicity [125]. Tumor mutational burden (TMB) is defined as the total number of somatic non-synonymous mutations per megabase in the coding region of a gene, including small insert-deletion variants [126]. Studies have indicated that tumors with a higher TMB and more neoantigens may respond better to immunotherapy [127], 128]. Additionally, clinical trials have suggested that PD-L1 expression is positively correlated with TMB and response to immunotherapy [129]. Moreover, the TMB detection can overcome the dynamic acquisition difficulties and spatial heterogeneity of PD-L1. Accordingly, in June 2020, the U.S. Food and Drug Administration (FDA) recognized the elevated TMB as a requirement for pembrolizumab therapy for several tumor types [130].
Liquid biopsy has been used to evaluate the prognosis of patients with NSCLC receiving immunotherapy. Moreover, TMB, as measured by ctDNA in the blood (bTMB), may be utilized as a biomarker for patients on anti-PD-1/PD-L1 medication. Accordingly, a previous study found that bTMB ≥6 was associated with favorable progression-free survival [104]. Menno et al. detected CTCs in patients with advanced NSCLC with a worse response rate to checkpoint inhibitors and high tumor-derived EVs. CTCs counted ≥18/7.5 mL in EVs were found associated with shorter survival [131]. Similarly, PD-L1 expression was measured in 96 patients with NSCLC treated with nivolumab and was confirmed to be higher in CTCs than in tissues (83 % vs. 41 %), indicating that dynamic monitoring of PD-L1 in CTCs is more convenient and sensitive [107]. Regulatory T (Treg) cells are a subset of T cells that express FoxP3 [132]. Research indicates that FoxP3 may be a favorable prognostic indicator for patients receiving anti-PD-1 treatment. One week after anti-PD-1 therapy, patients with NSCLC had higher response rates, longer progression-free survival, and improved OS in the presence of a greater number of circulating FoxP3+ Tregs (p<0.05). Similarly, low human polymorphonuclear myeloid‐derived suppressor cells (PMN-MDSCs), monocytic myeloid‐derived suppressor cells, and CD39+ CD8+ T cells have the potential to serve as favorable predictors for patients with advanced NSCLC receiving anti-PD-1 therapy [133].
However, most patients with NSCLC treated with ICIs develop early disease progression, making it impossible to identify patients who are likely to have durable clinical benefits (DCB) by imaging examination. In this regard, pre-treatment ctDNA and peripheral CD8+ T-cell levels have recently been demonstrated as independent predictors of DCB. Investigators found that patients with NSCLC with lower baseline circulating CD8+ T cell levels can achieve DCB from ICIs. Additionally, they developed and validated a non-invasive response classifier incorporating pre-treatment ctDNA and immune profiling, which could accurately identify patients with NSCLC with DCB [105]. Similarly, Duchemann et al. calculated the ratio of CD8+ PD-1+ to CD4+ PD-1+ (PD-1-Expressing Ratio on Lymphocytes in a Systemic blood sample, or ‘PERLS’) using cytometry in patients with NSCLC before receiving PD-1/PD-L1 blockers. PERLS+ was suggested to be significantly associated with PFS (9.63 months vs. 2.20 months; p=0.004) and OS (not reached vs. 7.98 months; p=0.02). The PERLS scores of patients with DCB were significantly higher than those of patients without DCB (mean 1.43 vs. 1.06; p=0.002) [106]. The neutrophil-to-lymphocyte ratio represents a proinflammatory diathesis that indicates poor prognosis in patients with NSCLC. Accordingly, in a phase 3 OAK trial (NCT02008227), pre-treatment NLR≥4 was strongly correlated to mortality after atezolizumab (HR=1.64; 95 % CI: 1.35–2.01). Consequently, a low baseline NLR could recognize patients with NSCLC who would benefit more from atezolizumab [134].
Perspectives
Although the results of current clinical trials have provided positive feedback, widespread implementation of liquid biopsy remains a challenge. Liquid biopsy marker detection has low complexity and accuracy; however, owing to the limitations of tumor shedding from different organs and other factors, the sensitivity and specificity of liquid biopsy markers remain insufficient. For example, ctDNA can interfere with the cfDNA background. Additionally, in the early stages of the disease, when the allele frequency (AF) is below 0.1 %, its content may fall below the detection threshold, leading to false negative results [135], [136], [137]. However, tumor heterogeneity leads to complexity in detection, such as inconsistent histological and hematological results and different changes in tumor progression. In addition, specific clinical application scenarios have different requirements, such as higher sensitivity for drug resistance mutation detection and a precise definition of ctDNA clearance for the prediction of drug efficacy.
Currently, a standard method for detecting liquid biopsy markers is lacking. In turn, the test results of different laboratories may be heterogeneous and lack scientific comparability owing to the lack of standardized operating procedures and data analysis methods, which affects their accuracy and reliability to a certain extent. A study comparing two NGS-based cfDNA detection methods reported a concordance rate of 7.5 % [138]. In addition, low concentrations of CTC and ctDNA can reduce the sensitivity. However, CTCs can only survive in blood for 1–2.5 h [139], 140]. Therefore, contamination during sample extraction and processing may lead to false-positive and false-negative results, and complex detection processes and inefficient equipment hinder their wider application [141]. Consequently, a number of standardizations, such as collection container selection, intervals, and protocols between plasma processing and blood drawing, extraction and separation of liquid biopsy components, precise and reliable technology for building libraries, and robust databases and platforms for bioinformatics analysis, must be established to improve its clinical application value.
Finally, a combination of multidimensional and multi-omics techniques is required to enhance the accuracy of liquid biopsies (Figure 3). A few studies have shown that one indicator (mutation or methylation of ctDNA) has obvious limitations in application and that combining multiple analytes can improve sensitivity [142], 143]. In such cases, a multi-marker combination analysis would lead to a comprehensive knowledge of tumor specificity, and ctDNA detection could be considered in combination with methylation, exosomes, circulating miRNAs, CTCs, metabolomics, and histopathology. In addition, based on the complementarity between samples from different sources collected simultaneously, collection at the same time, the precise detection is possible.

Current resources for MRD detection. Lung cancer cells and the substances released after apoptosis enter the blood circulation; therefore, the following substances can be detected in the blood, such as CTCs, ctDNA, exosomes, and circulating miRNA. Apart from blood, liquid biopsies include cerebrospinal fluid, sputum, PE, and urine. Note: MRD, minimal residual disease; CTC, circulating tumor cell; ctDNA, circulating tumor DNA; miRNA, microRNA; PE, pleural effusion.
Funding source: China Primary Health Care Foundation
Award Identifier / Grant number: (2022/2023-001)
Funding source: Science and Technology Program of the Joint Fund of Scientific Research for the Public Hospitals of Inner Mongolia Academy of Medical Sciences
Award Identifier / Grant number: 2024GLLH0864
Funding source: Faculty-Level Educational Science Research Projects of the Third Clinical Medical College, Harbin Medical University
Award Identifier / Grant number: SYY20240012
Funding source: the National Cancer Center Ascending Fund
Award Identifier / Grant number: NCC201808B025
Funding source: the Haiyan Scientific Research Fund of Harbin Medical University Cancer Hospital
Award Identifier / Grant number: JJMS2021-16
Acknowledgments
We thank Zhongli Liang (School of Civil Engineering and Architecture, Harbin Far East Institute of Technology) for providing a detailed explanation of the liquid biopsy methods and techniques.
-
Research ethics: Not applicable.
-
Informed consent: Not applicable.
-
Author contributions: Kaixun Xing and Xiaoqing Li: Data curation, Writing – original draft, Visualization. Peng Liu, Ting Li and Yinghao Guo: Writing – original draft, Writing – review & editing. Hetai Teng and Shuyan Dong: Draft figures. Hao Zhu: Writing – review & editing. Hongjiang He, Shan Yu and Jian Ma: Writing – review & editing, Methodology, Conceptualization, Supervision, Funding acquisition.
-
Use of Large Language Models, AI and Machine Learning Tools: None declared.
-
Conflict of interest: The authors declare no conflict of interest.
-
Research funding: This study was supported by the 2024 Faculty-Level Educational Science Research Projects of the Third Clinical Medical College, Harbin Medical University (No.SYY20240012), China Primary Health Care Foundation (2022/2023-001), the Haiyan Scientific Research Fund of Harbin Medical University Cancer Hospital (No. JJMS2021-16), the National Cancer Center Ascending Fund (NCC201808B025) and Science and Technology Program of the Joint Fund of Scientific Research for the Public Hospitals of Inner Mongolia Academy of Medical Sciences (2024GLLH0864).
-
Data availability: Not applicable.
References
1. Li, Q, Xia, C, Li, H, Yan, X, Yang, F, Cao, M, et al.. Disparities in 36 cancers across 185 countries: secondary analysis of global cancer statistics. Front Med 2024;18:911–20. https://doi.org/10.1007/s11684-024-1058-6.Suche in Google Scholar PubMed
2. Howlader, N, Forjaz, G, Mooradian, MJ, Meza, R, Kong, CY, Cronin, KA, et al.. The effect of advances in lung-cancer treatment on population mortality. New Engl J Med 2020;383:640–9. https://doi.org/10.1056/nejmoa1916623.Suche in Google Scholar PubMed PubMed Central
3. Pantel, K, Alix-Panabières, C. Tumour microenvironment: informing on minimal residual disease in solid tumours. Nat Rev Clin Oncol 2017;14:325–6. https://doi.org/10.1038/nrclinonc.2017.53.Suche in Google Scholar PubMed
4. Gale, D, Heider, K, Ruiz-Valdepenas, A, Hackinger, S, Perry, M, Marsico, G, et al.. Residual ctDNA after treatment predicts early relapse in patients with early-stage non-small cell lung cancer. Ann Oncol 2022;33:500–10. https://doi.org/10.1016/j.annonc.2022.02.007.Suche in Google Scholar PubMed PubMed Central
5. Piotrowska, Z, Niederst, MJ, Mino-Kenudson, M, Morales-Oyarvide, V, Fulton, L, Lockerman, E, et al.. Variation in mechanisms of acquired resistance (AR) among EGFR-mutant NSCLC patients with more than one post-resistant biopsy. J Clin Oncol 2014;32:8053. https://doi.org/10.1200/jco.2014.32.15_suppl.8053.Suche in Google Scholar
6. Siravegna, G, Marsoni, S, Siena, S, Bardelli, A. Integrating liquid biopsies into the management of cancer. Nat Rev Clin Oncol 2017;14:531–48. https://doi.org/10.1038/nrclinonc.2017.14.Suche in Google Scholar PubMed
7. Paweletz, CP, Sacher, AG, Raymond, CK, Alden, RS, O’Connell, A, Mach, SL, et al.. Bias-corrected targeted next-generation sequencing for rapid, multiplexed detection of actionable alterations in cell-free DNA from advanced lung cancer patients. Clin Cancer Res 2016;22:915–22. https://doi.org/10.1158/1078-0432.ccr-15-1627-t.Suche in Google Scholar PubMed PubMed Central
8. Rolfo, C, Mack, P, Scagliotti, GV, Aggarwal, C, Arcila, ME, Barlesi, F, et al.. Liquid biopsy for advanced NSCLC: a consensus statement from the International Association for the Study of Lung Cancer. J Thorac Oncol 2021;16:1647–62. https://doi.org/10.1016/j.jtho.2021.06.017.Suche in Google Scholar PubMed
9. Riethdorf, S, O’Flaherty, L, Hille, C, Pantel, K. Clinical applications of the CellSearch platform in cancer patients. Adv Drug Deliv Rev 2018;125:102–21. https://doi.org/10.1016/j.addr.2018.01.011.Suche in Google Scholar PubMed
10. Guo, J, Wu, C, Lin, X, Zhou, J, Zhang, J, Zheng, W, et al.. Establishment of a simplified dichotomic size-exclusion chromatography for isolating extracellular vesicles toward clinical applications. J Extracell Vesicles 2021;10:e12145. https://doi.org/10.1002/jev2.12145.Suche in Google Scholar PubMed PubMed Central
11. Raez, LE, Brice, K, Dumais, K, Lopez-Cohen, A, Wietecha, D, Izquierdo, PA, et al.. Liquid biopsy versus tissue biopsy to determine front line therapy in metastatic non-small cell lung cancer (NSCLC). Clin Lung Cancer 2023;24:120–9. https://doi.org/10.1016/j.cllc.2022.11.007.Suche in Google Scholar PubMed
12. Coppola, CA, De Summa, S, Matera, G, Pilato, B, Traversa, D, Tommasi, S. Liquid biopsy: the challenges of a revolutionary approach in oncology. Int J Mol Sci 2025;26:5013. https://doi.org/10.3390/ijms26115013.Suche in Google Scholar PubMed PubMed Central
13. Wang, X, Ding, P, Xu, W, Qiu, L, Ren, J, Fei, Y, et al.. Distinct CTC specific RNA profile enables NSCLC early detection and dynamic monitoring of advanced NSCLC. Adv Sci (Weinh) 2025;12:e2417849. https://doi.org/10.1002/advs.202417849.Suche in Google Scholar PubMed PubMed Central
14. Salleh, EA, Lee, YY, Zakaria, AD, Jalil, NAC, Musa, M. Cancer-associated fibroblasts of colorectal cancer: translational prospects in liquid biopsy and targeted therapy. Biocell 2023;47:2233–44. https://doi.org/10.32604/biocell.2023.030541.Suche in Google Scholar
15. Zhuri, D, Yalcintepe, S. Liquid biopsy as a new era in endometrial cancer. Oncologie 2024;26:483–6. https://doi.org/10.1515/oncologie-2024-0036.Suche in Google Scholar
16. Guo, Q, Shan, L, Luo, J, Huang, Y, Bao, Y, Wang, X, et al.. Novel strategies in liquid biopsy. Clin Chim Acta 2025;576:120385. https://doi.org/10.1016/j.cca.2025.120385.Suche in Google Scholar PubMed
17. Rolfo, C, Malapelle, U, Russo, A. Expanding the full potential of liquid biopsies for lung cancer patients. Cancer Res 2022;82:2826–8. https://doi.org/10.1158/0008-5472.can-22-2109.Suche in Google Scholar
18. Katz, RL, He, W, Khanna, A, Fernandez, RL, Zaidi, TM, Krebs, M, et al.. Genetically abnormal circulating cells in lung cancer patients: an antigen-independent fluorescence in situ hybridization-based case-control study. Clin Cancer Res 2010;16:3976–87. https://doi.org/10.1158/1078-0432.ccr-09-3358.Suche in Google Scholar PubMed PubMed Central
19. Li, W, Liu, JB, Hou, LK, Yu, F, Zhang, J, Wu, W, et al.. Liquid biopsy in lung cancer: significance in diagnostics, prediction, and treatment monitoring. Mol Cancer 2022;21:25. https://doi.org/10.1186/s12943-022-01505-z.Suche in Google Scholar PubMed PubMed Central
20. Kwizera, EA, Ou, W, Lee, S, Stewart, S, Shamul, JG, Xu, J, et al.. Correction to greatly enhanced CTC culture enabled by capturing CTC heterogeneity using a PEGylated PDMS-Titanium-Gold electromicrofluidic device with glutathione-controlled gentle cell release. ACS Nano 2022;16:19606. https://doi.org/10.1021/acsnano.2c10382.Suche in Google Scholar PubMed
21. Lu, L, Wang, Y, Ding, Y, Wang, Y, Zhu, Z, Lu, J, et al.. Profiling phenotypic heterogeneity of circulating tumor cells through spatially resolved immunocapture on nanoporous micropillar arrays. ACS Nano 2024;18:31135–47. https://doi.org/10.1021/acsnano.4c08893.Suche in Google Scholar PubMed
22. Moro, G, Fratte, CD, Normanno, N, Polo, F, Cinti, S. Point-of-Care testing for the detection of microRNAs: towards liquid biopsy on a chip. Angew Chem Int Ed Engl 2023;62:e202309135. https://doi.org/10.1002/ange.202309135.Suche in Google Scholar
23. Dvorská, D, Škovierová, H, Braný, D, Halašová, E, Danková, Z. Liquid biopsy as a tool for differentiation of leiomyomas and sarcomas of Corpus Uteri. Int J Mol Sci 2019;20:3825. https://doi.org/10.3390/ijms20153825.Suche in Google Scholar PubMed PubMed Central
24. Bradley, SH, Barclay, ME. “Liquid biopsy” for cancer screening. BMJ Clin Res Ed 2021;372:m4933. https://doi.org/10.1136/bmj.m4933.Suche in Google Scholar PubMed
25. Murtaza, M, Dawson, SJ, Tsui, DW, Gale, D, Forshew, T, Piskorz, AM, et al.. Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA. Nature 2013;497:108–12. https://doi.org/10.1038/nature12065.Suche in Google Scholar PubMed
26. Nikanjam, M, Kato, S, Kurzrock, R. Liquid biopsy: current technology and clinical applications. J Hematol Oncol 2022;15:131. https://doi.org/10.1186/s13045-022-01351-y.Suche in Google Scholar PubMed PubMed Central
27. Lin, D, Shen, L, Luo, M, Zhang, K, Li, J, Yang, Q, et al.. Circulating tumor cells: biology and clinical significance. Signal Transduct Targeted Ther 2021;6:404. https://doi.org/10.1038/s41392-021-00817-8.Suche in Google Scholar PubMed PubMed Central
28. Mosele, MF, Westphalen, CB, Stenzinger, A, Barlesi, F, Bayle, A, Bièche, I, et al.. Recommendations for the use of next-generation sequencing (NGS) for patients with advanced cancer in 2024: a report from the ESMO Precision Medicine Working Group. Ann Oncol 2024;35:588–606. https://doi.org/10.1016/j.annonc.2024.04.005.Suche in Google Scholar PubMed
29. Pott, C, Kotrova, M, Darzentas, N, Brüggemann, M, Khouja, M, cfDNA-Based NGS IG analysis in lymphoma. Methods Mol Biol 2022;2453:101–17. https://doi.org/10.1007/978-1-0716-2115-8_7.Suche in Google Scholar PubMed PubMed Central
30. Zhao, Y, O’Keefe, CM, Hsieh, K, Cope, L, Joyce, SC, Pisanic, TR, et al.. Multiplex digital methylation-specific PCR for noninvasive screening of lung cancer. Adv Sci (Weinh) 2023;10:e2206518. https://doi.org/10.1002/advs.202206518.Suche in Google Scholar PubMed PubMed Central
31. Kimiz-Gebologlu, I, Oncel, SS. Exosomes: large-scale production, isolation, drug loading efficiency, and biodistribution and uptake. J Control Release 2022;347:533–43. https://doi.org/10.1016/j.jconrel.2022.05.027.Suche in Google Scholar PubMed
32. Zhu, L, Sun, HT, Wang, S, Huang, SL, Zheng, Y, Wang, CQ, et al.. Isolation and characterization of exosomes for cancer research. J Hematol Oncol 2020;13:152. https://doi.org/10.1186/s13045-020-00987-y.Suche in Google Scholar PubMed PubMed Central
33. Liang, W, Zhao, Y, Huang, W, Gao, Y, Xu, W, Tao, J, et al.. Non-invasive diagnosis of early-stage lung cancer using high-throughput targeted DNA methylation sequencing of circulating tumor DNA (ctDNA). Theranostics 2019;9:2056–70. https://doi.org/10.7150/thno.28119.Suche in Google Scholar PubMed PubMed Central
34. Frick, MA, Feigenberg, SJ, Jean-Baptiste, SR, Aguarin, LA, Mendes, A, Chinniah, C, et al.. Circulating tumor cells are associated with recurrent disease in patients with early-stage non-small cell lung cancer treated with stereotactic body radiotherapy. Clin Cancer Res 2020;26:2372–80. https://doi.org/10.1158/1078-0432.ccr-19-2158.Suche in Google Scholar PubMed PubMed Central
35. Riely, GJ, Wood, DE, Ettinger, DS, Aisner, DL, Akerley, W, Bauman, JR, et al.. Non-small cell lung cancer, version 4.2024, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Cancer Netw 2024;22:249–74. https://doi.org/10.6004/jnccn.2204.0023.Suche in Google Scholar PubMed
36. Lindeman, NI, Cagle, PT, Aisner, DL, Arcila, ME, Beasley, MB, Bernicker, EH, et al.. Updated molecular testing guideline for the selection of lung cancer patients for treatment with targeted tyrosine kinase inhibitors: guideline from the college of American pathologists, the International Association for the Study of Lung Cancer, and the Association for Molecular Pathology. J Thorac Oncol 2018;13:323–58. https://doi.org/10.1016/j.jtho.2017.12.001.Suche in Google Scholar PubMed
37. Lim, ZF, Ma, PC. Emerging insights of tumor heterogeneity and drug resistance mechanisms in lung cancer targeted therapy. J Hematol Oncol 2019;12:134. https://doi.org/10.1186/s13045-019-0818-2.Suche in Google Scholar PubMed PubMed Central
38. Liu, Y, Zhang, B, Wu, X, Wang, F, Yang, Z, Li, M, et al.. A facile liquid biopsy assay for highly efficient CTCs capture and reagent-less monitoring of immune checkpoint PD-L1 expression on CTCs with non-small cell lung cancer patients. Biosens Bioelectron 2025;275:117236. https://doi.org/10.1016/j.bios.2025.117236.Suche in Google Scholar PubMed
39. Abbosh, C, Birkbak, NJ, Swanton, C. Early stage NSCLC – challenges to implementing ctDNA-based screening and MRD detection. Nat Rev Clin Oncol 2018;15:577–86. https://doi.org/10.1038/s41571-018-0058-3.Suche in Google Scholar PubMed
40. Qiu, B, Guo, W, Zhang, F, Lv, F, Ji, Y, Peng, Y, et al.. Dynamic recurrence risk and adjuvant chemotherapy benefit prediction by ctDNA in resected NSCLC. Nat Commun 2021;12:6770. https://doi.org/10.1038/s41467-021-27022-z.Suche in Google Scholar PubMed PubMed Central
41. Zhong, CH, Tong, D, Zhou, ZQ, Su, ZQ, Luo, YL, Xing, J, et al.. Performance evaluation of detecting circulating tumor cells and tumor cells in bronchoalveolar lavage fluid in diagnosis of peripheral lung cancer. J Thorac Dis 2018;10:S830–s7. https://doi.org/10.21037/jtd.2017.12.125.Suche in Google Scholar PubMed PubMed Central
42. Xia, L, Mei, J, Kang, R, Deng, S, Chen, Y, Yang, 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;28:3308–17. https://doi.org/10.1158/1078-0432.ccr-21-3044.Suche in Google Scholar
43. Peng, M, Huang, Q, Yin, W, Tan, S, Chen, C, Liu, W, et al.. Circulating tumor DNA as a prognostic biomarker in localized non-small cell lung cancer. Front Oncol 2020;10:561598. https://doi.org/10.3389/fonc.2020.561598.Suche in Google Scholar PubMed PubMed Central
44. Tong, L, Ding, N, Tong, X, Li, J, Zhang, Y, Wang, X, et al.. Tumor-derived DNA from pleural effusion supernatant as a promising alternative to tumor tissue in genomic profiling of advanced lung cancer. Theranostics 2019;9:5532–41. https://doi.org/10.7150/thno.34070.Suche in Google Scholar PubMed PubMed Central
45. Hu, T, Shen, H, Huang, H, Song, M, Yang, Z, Zhou, Y, et al.. Urinary circulating DNA profiling in non-small cell lung cancer patients following treatment shows prognostic potential. J Thorac Dis 2018;10:4137–46. https://doi.org/10.21037/jtd.2018.06.50.Suche in Google Scholar PubMed PubMed Central
46. Yu, W, Hurley, J, Roberts, D, Chakrabortty, SK, Enderle, D, Noerholm, M, et al.. Exosome-based liquid biopsies in cancer: opportunities and challenges. Ann Oncol 2021;32:466–77. https://doi.org/10.1016/j.annonc.2021.01.074.Suche in Google Scholar PubMed PubMed Central
47. Krug, AK, Enderle, D, Karlovich, C, Priewasser, T, Bentink, S, Spiel, A, et al.. Improved EGFR mutation detection using combined exosomal RNA and circulating tumor DNA in NSCLC patient plasma. Ann Oncol 2018;29:700–6. https://doi.org/10.1093/annonc/mdx765.Suche in Google Scholar PubMed PubMed Central
48. Carter, L, Rothwell, DG, Mesquita, B, Smowton, C, Leong, HS, Fernandez-Gutierrez, F, et al.. Molecular analysis of circulating tumor cells identifies distinct copy-number profiles in patients with chemosensitive and chemorefractory small-cell lung cancer. Nat Med 2017;23:114–9. https://doi.org/10.1038/nm.4239.Suche in Google Scholar PubMed
49. Chemi, F, Rothwell, DG, McGranahan, N, Gulati, S, Abbosh, C, Pearce, SP, et al.. Publisher correction: pulmonary venous circulating tumor cell dissemination before tumor resection and disease relapse. Nat Med 2020;26:1147. https://doi.org/10.1038/s41591-020-0865-9.Suche in Google Scholar PubMed
50. Zhang, C, Guan, Y, Sun, Y, Ai, D, Guo, Q. Tumor heterogeneity and circulating tumor cells. Cancer Lett 2016;374:216–23. https://doi.org/10.1016/j.canlet.2016.02.024.Suche in Google Scholar PubMed
51. Sawada, T, Watanabe, M, Fujimura, Y, Yagishita, S, Shimoyama, T, Maeda, Y, et al.. Sensitive cytometry based system for enumeration, capture and analysis of gene mutations of circulating tumor cells. Cancer Sci 2016;107:307–14. https://doi.org/10.1111/cas.12868.Suche in Google Scholar PubMed PubMed Central
52. Wang, J, Huang, J, Wang, K, Xu, J, Huang, J, Zhang, T. Prognostic significance of circulating tumor cells in non-small-cell lung cancer patients: a meta-analysis. PLoS One 2013;8:e78070. https://doi.org/10.1371/journal.pone.0078070.Suche in Google Scholar PubMed PubMed Central
53. Ettinger, DS, Wood, DE, Aisner, DL, Akerley, W, Bauman, JR, Bharat, A, et al.. NCCN guidelines insights: non-small cell lung cancer, version 2.2021. J Natl Compr Cancer Netw 2021;19:254–66. https://doi.org/10.6004/jnccn.2021.0013.Suche in Google Scholar PubMed
54. Hofman, V, Bonnetaud, C, Ilie, MI, Vielh, P, Vignaud, JM, Fléjou, JF, et al.. Preoperative circulating tumor cell detection using the isolation by size of epithelial tumor cell method for patients with lung cancer is a new prognostic biomarker. Clin Cancer Res 2011;17:827–35. https://doi.org/10.1158/1078-0432.ccr-10-0445.Suche in Google Scholar
55. Sher, YP, Shih, JY, Yang, PC, Roffler, SR, Chu, YW, Wu, CW, et al.. Prognosis of non-small cell lung cancer patients by detecting circulating cancer cells in the peripheral blood with multiple marker genes. Clin Cancer Res 2005;11:173–9. https://doi.org/10.1158/1078-0432.173.11.1.Suche in Google Scholar
56. Wu, C, Hao, H, Li, L, Zhou, X, Guo, Z, Zhang, L, et al.. Preliminary investigation of the clinical significance of detecting circulating tumor cells enriched from lung cancer patients. J Thorac Oncol 2009;4:30–6. https://doi.org/10.1097/jto.0b013e3181914125.Suche in Google Scholar
57. Krebs, MG, Sloane, R, Priest, L, Lancashire, L, Hou, JM, Greystoke, A, et al.. Evaluation and prognostic significance of circulating tumor cells in patients with non-small-cell lung cancer. J Clin Oncol 2011;29:1556–63. https://doi.org/10.1200/jco.2010.28.7045.Suche in Google Scholar PubMed
58. Bayarri-Lara, C, Ortega, FG, Cueto Ladrón de Guevara, A, Puche, JL, Ruiz Zafra, J, de Miguel-Pérez, D, et al.. Circulating tumor cells identify early recurrence in patients with non-small cell lung cancer undergoing radical resection. PLoS One 2016;11:e0148659. https://doi.org/10.1371/journal.pone.0148659.Suche in Google Scholar PubMed PubMed Central
59. Lyu, M, Zhou, J, Ning, K, Ying, B. The diagnostic value of circulating tumor cells and ctDNA for gene mutations in lung cancer. Onco Targets Ther 2019;12:2539–52. https://doi.org/10.2147/ott.s195342.Suche in Google Scholar PubMed PubMed Central
60. O’Flaherty, L, Wikman, H, Pantel, K. Biology and clinical significance of circulating tumor cell subpopulations in lung cancer. Transl Lung Cancer Res 2017;6:431–43. https://doi.org/10.21037/tlcr.2017.07.03.Suche in Google Scholar PubMed PubMed Central
61. Reddy, RM, Murlidhar, V, Zhao, L, Grabauskiene, S, Zhang, Z, Ramnath, N, et al.. Pulmonary venous blood sampling significantly increases the yield of circulating tumor cells in early-stage lung cancer. J Thorac Cardiovasc Surg 2016;151:852–8. https://doi.org/10.1016/j.jtcvs.2015.09.126.Suche in Google Scholar PubMed
62. Chinniah, C, Aguarin, L, Cheng, P, Decesaris, C, Cutillo, A, Berman, AT, et al.. Early detection of recurrence in patients with locally advanced non-small-cell lung cancer via circulating tumor cell analysis. Clin Lung Cancer 2019;20:384–90.e2. https://doi.org/10.1016/j.cllc.2019.04.011.Suche in Google Scholar PubMed PubMed Central
63. Maly, V, Maly, O, Kolostova, K, Bobek, V. Circulating tumor cells in diagnosis and treatment of lung cancer. In Vivo 2019;33:1027–37. https://doi.org/10.21873/invivo.11571.Suche in Google Scholar PubMed PubMed Central
64. Carlsson, A, Nair, VS, Luttgen, MS, Keu, KV, Horng, G, Vasanawala, M, et al.. Circulating tumor microemboli diagnostics for patients with non-small-cell lung cancer. J Thorac Oncol 2014;9:1111–9. https://doi.org/10.1097/jto.0000000000000235.Suche in Google Scholar
65. Aceto, N. Bring along your friends: homotypic and heterotypic circulating tumor cell clustering to accelerate metastasis. Biomed J 2020;43:18–23. https://doi.org/10.1016/j.bj.2019.11.002.Suche in Google Scholar PubMed PubMed Central
66. Aceto, N, Bardia, A, Miyamoto, DT, Donaldson, MC, Wittner, BS, Spencer, JA, et al.. Circulating tumor cell clusters are oligoclonal precursors of breast cancer metastasis. Cell 2014;158:1110–22. https://doi.org/10.1016/j.cell.2014.07.013.Suche in Google Scholar PubMed PubMed Central
67. Liotta, LA, Saidel, MG, Kleinerman, J. The significance of hematogenous tumor cell clumps in the metastatic process. Cancer Res 1976;36:889–94.Suche in Google Scholar
68. Murlidhar, V, Reddy, RM, Fouladdel, S, Zhao, L, Ishikawa, MK, Grabauskiene, S, et al.. Poor prognosis indicated by venous circulating tumor cell clusters in early-stage lung cancers. Cancer Res 2017;77:5194–206. https://doi.org/10.1158/0008-5472.can-16-2072.Suche in Google Scholar
69. Mouliere, F, Chandrananda, D, Piskorz, AM, Moore, EK, Morris, J, Ahlborn, LB, et al.. Enhanced detection of circulating tumor DNA by fragment size analysis. Sci Transl Med 2018;10:eaat4921. https://doi.org/10.1126/scitranslmed.aat4921.Suche in Google Scholar PubMed PubMed Central
70. Guo, N, Lou, F, Ma, Y, Li, J, Yang, B, Chen, W, et al.. Circulating tumor DNA detection in lung cancer patients before and after surgery. Sci Rep 2016;6:33519. https://doi.org/10.1038/srep33519.Suche in Google Scholar PubMed PubMed Central
71. Chen, K, Zhang, J, Guan, T, Yang, F, Lou, F, Chen, W, et al.. Comparison of plasma to tissue DNA mutations in surgical patients with non-small cell lung cancer. J thorac cardiov sur 2017;154:1123–31.e2. https://doi.org/10.1016/j.jtcvs.2017.04.073.Suche in Google Scholar PubMed
72. Newman, AM, Bratman, SV, To, J, Wynne, JF, Eclov, NC, Modlin, LA, et al.. An ultrasensitive method for quantitating circulating tumor DNA with broad patient coverage. Nat Med 2014;20:548–54. https://doi.org/10.1038/nm.3519.Suche in Google Scholar PubMed PubMed Central
73. Chaudhuri, AA, Chabon, JJ, Lovejoy, AF, Newman, AM, Stehr, H, Azad, TD, et al.. Early detection of molecular residual disease in localized lung cancer by circulating tumor DNA profiling. Cancer Discov 2017;7:1394–403. https://doi.org/10.1158/2159-8290.cd-17-0716.Suche in Google Scholar PubMed PubMed Central
74. Abbosh, C, Birkbak, NJ, Wilson, GA, Jamal-Hanjani, M, Constantin, T, Salari, R, et al.. Phylogenetic ctDNA analysis depicts early-stage lung cancer evolution. Nature 2017;545:446–51. https://doi.org/10.1038/nature22364.Suche in Google Scholar PubMed PubMed Central
75. Jamal-Hanjani, M, Wilson, GA, McGranahan, N, Birkbak, NJ, Watkins, TBK, Veeriah, S, et al.. Tracking the evolution of non-small-cell lung cancer. New Engl J Med 2017;376:2109–21. https://doi.org/10.1056/nejmoa1616288.Suche in Google Scholar PubMed
76. Chen, K, Zhao, H, Shi, Y, Yang, F, Wang, LT, Kang, G, et al.. Perioperative dynamic changes in circulating tumor DNA in patients with lung cancer (DYNAMIC). Clin Cancer Res 2019;25:7058–67. https://doi.org/10.1158/1078-0432.ccr-19-1213.Suche in Google Scholar PubMed
77. Hellmann, MD, Nabet, BY, Rizvi, H, Chaudhuri, AA, Wells, DK, Dunphy, MPS, et al.. Circulating tumor DNA analysis to assess risk of progression after long-term response to PD-(L)1 blockade in NSCLC. Clin Cancer Res 2020;26:2849–58. https://doi.org/10.1158/1078-0432.ccr-19-3418.Suche in Google Scholar PubMed PubMed Central
78. Xu, R, Rai, A, Chen, M, Suwakulsiri, W, Greening, DW, Simpson, RJ. Extracellular vesicles in cancer – implications for future improvements in cancer care. Nat Rev Clin Oncol 2018;15:617–38. https://doi.org/10.1038/s41571-018-0036-9.Suche in Google Scholar PubMed
79. Trombetta, D, Sparaneo, A, Fabrizio, FP, Muscarella, LA. Liquid biopsy and NSCLC. Lung Cancer Manag 2016;5:91–104. https://doi.org/10.2217/lmt-2016-0006.Suche in Google Scholar PubMed PubMed Central
80. Han, P, Zhao, J, Gao, L. Increased serum exosomal long non-coding RNA SNHG15 expression predicts poor prognosis in non-small cell lung cancer. J Clin Lab Anal 2021;35:e23979. https://doi.org/10.1002/jcla.23979.Suche in Google Scholar PubMed PubMed Central
81. Dong, Q, Dong, L, Liu, S, Kong, Y, Zhang, M, Wang, X. Tumor-derived exosomal eIF4E as a biomarker for survival prediction in patients with non-small cell lung cancer. Med Sci Monit 2020;26:e923210. https://doi.org/10.12659/msm.923210.Suche in Google Scholar PubMed PubMed Central
82. Santarpia, M, Liguori, A, D’Aveni, A, Karachaliou, N, Gonzalez-Cao, M, Daffinà, MG, et al.. Liquid biopsy for lung cancer early detection. J Thorac Dis 2018;10:S882–s97. https://doi.org/10.21037/jtd.2018.03.81.Suche in Google Scholar PubMed PubMed Central
83. Yan, X, Liu, C. Application of non-blood-derived fluid biopsy in monitoring minimal residual diseases of lung cancer. Front Surg 2022;9:865040. https://doi.org/10.3389/fsurg.2022.865040.Suche in Google Scholar PubMed PubMed Central
84. Nel, I, Jehn, U, Gauler, T, Hoffmann, AC. Individual profiling of circulating tumor cell composition in patients with non-small cell lung cancer receiving platinum based treatment. Transl Lung Cancer Res 2014;3:100–6. https://doi.org/10.3978/j.issn.2218-6751.2014.03.05.Suche in Google Scholar PubMed PubMed Central
85. Zhen, Z, Li, YS, Su, CY, Cheng, X, Zhou, SJ, Han, Y, et al.. Expression level of epithelial cell adhesion molecule (EpCAM) of circulating tumor cells (CTCs) of patients with NSCLC as an early indicator to monitor the effects of postoperative adjuvant chemotherapy. Transl Cancer Res 2021;10:3299–305. https://doi.org/10.21037/tcr-21-205.Suche in Google Scholar PubMed PubMed Central
86. Chang, Y, Wang, Y, Li, B, Lu, X, Wang, R, Li, H, et al.. Whole-exome sequencing on circulating tumor cells explores platinum-drug resistance mutations in advanced non-small cell lung cancer. Front Genet 2021;12:722078. https://doi.org/10.3389/fgene.2021.722078.Suche in Google Scholar PubMed PubMed Central
87. Guo, AX, Xiao, F, Shao, WH, Zhan, Y, Zhang, L, Xiong, J, et al.. Sequential whole exome sequencing reveals somatic mutations associated with platinum response in NSCLC. Onco Targets Ther 2020;13:6485–96. https://doi.org/10.2147/ott.s254747.Suche in Google Scholar PubMed PubMed Central
88. Han, X, Han, Y, Tan, Q, Huang, Y, Yang, J, Yang, S, et al.. Tracking longitudinal genetic changes of circulating tumor DNA (ctDNA) in advanced lung adenocarcinoma treated with chemotherapy. J Transl Med 2019;17:339. https://doi.org/10.1186/s12967-019-2087-9.Suche in Google Scholar PubMed PubMed Central
89. Breadner, DA, Vincent, MD, Correa, R, Black, M, Warner, A, Sanatani, M, et al.. Exploitation of treatment induced tumor lysis to enhance the sensitivity of ctDNA analysis: a first-in-human pilot study. Lung Cancer 2022;165:145–51. https://doi.org/10.1016/j.lungcan.2022.01.013.Suche in Google Scholar PubMed
90. Ma, Y, Yuwen, D, Chen, J, Zheng, B, Gao, J, Fan, M, et al.. Exosomal transfer of cisplatin-induced miR-425-3p confers cisplatin resistance in NSCLC through activating autophagy. Int J Nanomed 2019;14:8121–32. https://doi.org/10.2147/ijn.s221383.Suche in Google Scholar
91. Zhao, X, Li, M, Dai, X, Yang, Y, Peng, Y, Xu, C, et al.. Downregulation of exosomal miR-1273a increases cisplatin resistance of non-small cell lung cancer by upregulating the expression of syndecan binding protein. Oncol Rep 2020;44:2165–73. https://doi.org/10.3892/or.2020.7753.Suche in Google Scholar PubMed PubMed Central
92. Monastirioti, A, Papadaki, C, Rounis, K, Kalapanida, D, Mavroudis, D, Agelaki, S. A prognostic role for circulating microRNAs involved in macrophage polarization in advanced non-small cell lung cancer. Cells 2021;10:1988. https://doi.org/10.3390/cells10081988.Suche in Google Scholar PubMed PubMed Central
93. Sánchez Hernández, A, José, JO, Vidal Martínez, J, Blanco, R, Maciá, S, Esquerdo, GG, et al.. Quantification of circulating endothelial cells as a predictor of response to chemotherapy with platinum and pemetrexed in patients with advanced non-squamous non-small cell lung carcinoma. Clin Transl Oncol 2015;17:281–8. https://doi.org/10.1007/s12094-014-1223-5.Suche in Google Scholar PubMed
94. Ku, BM, Kim, YJ, Park, D, Lee, SH, Ahn, JS, Park, K, et al.. Role of circulating tumor DNA profiling in patients with non-small cell lung cancer treated with EGFR inhibitor. Oncol Basel 2022;100:228–37. https://doi.org/10.1159/000516813.Suche in Google Scholar PubMed
95. Yang, Y, Zhang, X, Wang, R, Qin, J, Wang, J, Li, Z, et al.. Osimertinib resistance with a novel EGFR L858R/A859S/Y891D triple mutation in a patient with non-small cell lung cancer: a case report. Front Oncol 2020;10:542277. https://doi.org/10.3389/fonc.2020.542277.Suche in Google Scholar PubMed PubMed Central
96. Moiseyenko, FV, Kuligina, ES, Zhabina, AS, Belukhin, SA, Laidus, TA, Martianov, AS, et al.. Changes in the concentration of EGFR-mutated plasma DNA in the first hours of targeted therapy allow the prediction of tumor response in patients with EGFR-driven lung cancer. Int J Clin Oncol 2022;27:850–62. https://doi.org/10.1007/s10147-022-02128-6.Suche in Google Scholar PubMed PubMed Central
97. Hisakane, K, Seike, M, Sugano, T, Yoshikawa, A, Matsuda, K, Takano, N, et al.. Exosome-derived miR-210 involved in resistance to osimertinib and epithelial-mesenchymal transition in EGFR mutant non-small cell lung cancer cells. Thorac Cancer 2021;12:1690–8. https://doi.org/10.1111/1759-7714.13943.Suche in Google Scholar PubMed PubMed Central
98. Li, X, Chen, C, Wang, Z, Liu, J, Sun, W, Shen, K, et al.. Elevated exosome-derived miRNAs predict osimertinib resistance in non-small cell lung cancer. Cancer Cell Int 2021;21:428. https://doi.org/10.1186/s12935-021-02075-8.Suche in Google Scholar PubMed PubMed Central
99. Kaźmierczak, D, Eide, IJZ, Gencheva, R, Lai, Y, Lewensohn, R, Tsakonas, G, et al.. Elevated expression of miR-494-3p is associated with resistance to osimertinib in EGFR T790M-positive non-small cell lung cancer. Transl Lung Cancer Res 2022;11:722–34. https://doi.org/10.21037/tlcr-21-955.Suche in Google Scholar PubMed PubMed Central
100. Maeda, R, Ishii, G, Ito, M, Hishida, T, Yoshida, J, Nishimura, M, et al.. Number of circulating endothelial progenitor cells and intratumoral microvessel density in non-small cell lung cancer patients: differences in angiogenic status between adenocarcinoma histologic subtypes. J Thorac Oncol 2012;7:503–11. https://doi.org/10.1097/jto.0b013e318241780e.Suche in Google Scholar
101. Ikeda, S, Kato, T, Ogura, T, Sekine, A, Oda, T, Masuda, N, et al.. Phase II study of bevacizumab, cisplatin, and docetaxel plus maintenance bevacizumab as first-line treatment for patients with advanced non-squamous non-small-cell lung cancer combined with exploratory analysis of circulating endothelial cells: Thoracic Oncology Research Group (TORG)1016. BMC Cancer 2018;18:241. https://doi.org/10.1186/s12885-018-4150-y.Suche in Google Scholar PubMed PubMed Central
102. Liu, Z, Wang, J, Meng, Z, Wang, X, Zhang, C, Qin, T, et al.. CD31-labeled circulating endothelial cells as predictor in anlotinib-treated non-small-cell lung cancer: analysis on ALTER-0303 study. Cancer Med 2018;7:3011–21. https://doi.org/10.1002/cam4.1584.Suche in Google Scholar PubMed PubMed Central
103. Zhang, T, Zhang, L, Gao, Y, Wang, Y, Liu, Y, Zhang, H, et al.. Role of aneuploid circulating tumor cells and CD31+ circulating tumor endothelial cells in predicting and monitoring anti-angiogenic therapy efficacy in advanced NSCLC. Mol Oncol 2021;15:2891–909. https://doi.org/10.1002/1878-0261.13092.Suche in Google Scholar PubMed PubMed Central
104. Wang, Z, Duan, J, Cai, S, Han, M, Dong, H, Zhao, J, et al.. Assessment of blood tumor mutational burden as a potential biomarker for immunotherapy in patients with non-small cell lung cancer with use of a next-generation sequencing cancer gene panel. Jama Oncol 2019;5:696–702. https://doi.org/10.1001/jamaoncol.2018.7098.Suche in Google Scholar PubMed PubMed Central
105. Nabet, BY, Esfahani, MS, Moding, EJ, Hamilton, EG, Chabon, JJ, Rizvi, H, et al.. Noninvasive early identification of therapeutic benefit from immune checkpoint inhibition. Cell 2020;183:363–76.e13. https://doi.org/10.1016/j.cell.2020.09.001.Suche in Google Scholar PubMed PubMed Central
106. Duchemann, B, Naigeon, M, Auclin, E, Ferrara, R, Cassard, L, Jouniaux, JM, et al.. CD8+PD-1+ to CD4+PD-1+ ratio (PERLS) is associated with prognosis of patients with advanced NSCLC treated with PD-(L)1 blockers. J Immunother Cancer 2022;10:e004012. https://doi.org/10.1136/jitc-2021-004012.Suche in Google Scholar PubMed PubMed Central
107. Guibert, N, Delaunay, M, Lusque, A, Boubekeur, N, Rouquette, I, Clermont, E, et al.. PD-L1 expression in circulating tumor cells of advanced non-small cell lung cancer patients treated with nivolumab. Lung cancer 2018;120:108–12. https://doi.org/10.1016/j.lungcan.2018.04.001.Suche in Google Scholar PubMed
108. Das, M, Riess, JW, Frankel, P, Schwartz, E, Bennis, R, Hsieh, HB, et al.. ERCC1 expression in circulating tumor cells (CTCs) using a novel detection platform correlates with progression-free survival (PFS) in patients with metastatic non-small-cell lung cancer (NSCLC) receiving platinum chemotherapy. Lung Cancer 2012;77:421–6. https://doi.org/10.1016/j.lungcan.2012.04.005.Suche in Google Scholar PubMed
109. Nel, I, Gauler, TC, Eberhardt, WE, Nickel, AC, Schuler, M, Thomale, J, et al.. Formation and repair kinetics of Pt-(GpG) DNA adducts in extracted circulating tumour cells and response to platinum treatment. Br J Cancer 2013;109:1223–9. https://doi.org/10.1038/bjc.2013.419.Suche in Google Scholar PubMed PubMed Central
110. Frixa, T, Sacconi, A, Cioce, M, Roscilli, G, Ferrara, FF, Aurisicchio, L, et al.. MicroRNA-128-3p-mediated depletion of Drosha promotes lung cancer cell migration. Carcinogenesis 2018;39:293–304. https://doi.org/10.1093/carcin/bgx134.Suche in Google Scholar PubMed
111. Xue, X, Liu, Y, Wang, Y, Meng, M, Wang, K, Zang, X, et al.. MiR-21 and MiR-155 promote non-small cell lung cancer progression by downregulating SOCS1, SOCS6, and PTEN. Oncotarget 2016;7:84508–19. https://doi.org/10.18632/oncotarget.13022.Suche in Google Scholar PubMed PubMed Central
112. Curtin, NJ, Hughes, AN. Pemetrexed disodium, a novel antifolate with multiple targets. Lancet Oncol 2001;2:298–306. https://doi.org/10.1016/s1470-2045(00)00325-9.Suche in Google Scholar PubMed
113. Franchina, T, Amodeo, V, Bronte, G, Savio, G, Ricciardi, GR, Picciotto, M, et al.. Circulating miR-22, miR-24 and miR-34a as novel predictive biomarkers to pemetrexed-based chemotherapy in advanced non-small cell lung cancer. J Cell Physiol 2014;229:97–9. https://doi.org/10.1002/jcp.24422.Suche in Google Scholar PubMed
114. Buszka, K, Ntzifa, A, Owecka, B, Kamińska, P, Kolecka-Bednarczyk, A, Zabel, M, et al.. Liquid biopsy analysis as a tool for TKI-based treatment in non-small cell lung cancer. Cells 2022;11:2871. https://doi.org/10.3390/cells11182871.Suche in Google Scholar PubMed PubMed Central
115. Uchida, J, Kato, K, Kukita, Y, Kumagai, T, Nishino, K, Daga, H, et al.. Diagnostic accuracy of noninvasive genotyping of EGFR in lung cancer patients by deep sequencing of plasma cell-free DNA. Clin Chem 2015;61:1191–6. https://doi.org/10.1373/clinchem.2015.241414.Suche in Google Scholar PubMed
116. Wan, R, Wang, Z, Lee, JJ, Wang, S, Li, Q, Tang, F, et al.. Comprehensive analysis of the discordance of EGFR mutation status between tumor tissues and matched circulating tumor DNA in advanced non-small cell lung cancer. J Thorac Oncol 2017;12:1376–87. https://doi.org/10.1016/j.jtho.2017.05.011.Suche in Google Scholar PubMed
117. Ou, SI, Horn, L, Cruz, M, Vafai, D, Lovly, CM, Spradlin, A, et al.. Emergence of FGFR3-TACC3 fusions as a potential by-pass resistance mechanism to EGFR tyrosine kinase inhibitors in EGFR mutated NSCLC patients. Lung Cancer 2017;111:61–4. https://doi.org/10.1016/j.lungcan.2017.07.006.Suche in Google Scholar PubMed PubMed Central
118. Oztan, A, Fischer, S, Schrock, AB, Erlich, RL, Lovly, CM, Stephens, PJ, et al.. Emergence of EGFR G724S mutation in EGFR-mutant lung adenocarcinoma post progression on osimertinib. Lung Cancer 2017;111:84–7. https://doi.org/10.1016/j.lungcan.2017.07.002.Suche in Google Scholar PubMed
119. Liang, W, He, Q, Chen, Y, Chuai, S, Yin, W, Wang, W, et al.. Metastatic EML4-ALK fusion detected by circulating DNA genotyping in an EGFR-mutated NSCLC patient and successful management by adding ALK inhibitors: a case report. BMC Cancer 2016;16:62. https://doi.org/10.1186/s12885-016-2088-5.Suche in Google Scholar PubMed PubMed Central
120. Provencio, M, Serna-Blasco, R, Franco, F, Calvo, V, Royuela, A, Auglytė, M, et al.. Analysis of circulating tumour DNA to identify patients with epidermal growth factor receptor-positive non-small cell lung cancer who might benefit from sequential tyrosine kinase inhibitor treatment. Eur J Cancer 2021;149:61–72. https://doi.org/10.1016/j.ejca.2021.02.031.Suche in Google Scholar PubMed
121. Ramalingam, SS, Vansteenkiste, J, Planchard, D, Cho, BC, Gray, JE, Ohe, Y, et al.. Overall survival with osimertinib in untreated, EGFR-mutated advanced NSCLC. New Engl J Med 2020;382:41–50. https://doi.org/10.1056/nejmoa1913662.Suche in Google Scholar PubMed
122. Leonetti, A, Sharma, S, Minari, R, Perego, P, Giovannetti, E, Tiseo, M. Resistance mechanisms to osimertinib in EGFR-mutated non-small cell lung cancer. Br J Cancer 2019;121:725–37. https://doi.org/10.1038/s41416-019-0573-8.Suche in Google Scholar PubMed PubMed Central
123. Prelaj, A, Ferrara, R, Rebuzzi, SE, Proto, C, Signorelli, D, Galli, G, et al.. EPSILoN: a prognostic score for immunotherapy in advanced non-small-cell lung cancer: a validation cohort. Cancers (Basel) 2019;11:1954. https://doi.org/10.3390/cancers11121954.Suche in Google Scholar PubMed PubMed Central
124. Dal Bello, MG, Alama, A, Coco, S, Vanni, I, Grossi, F. Understanding the checkpoint blockade in lung cancer immunotherapy. Drug Discov Today 2017;22:1266–73. https://doi.org/10.1016/j.drudis.2017.05.016.Suche in Google Scholar PubMed
125. Teng, F, Meng, X, Kong, L, Yu, J. Progress and challenges of predictive biomarkers of anti PD-1/PD-L1 immunotherapy: a systematic review. Cancer Lett 2018;414:166–73. https://doi.org/10.1016/j.canlet.2017.11.014.Suche in Google Scholar PubMed
126. Früh, M, Peters, S. Genomic features of response to combination immunotherapy in lung cancer. Cancer Cell 2018;33:791–3. https://doi.org/10.1016/j.ccell.2018.04.005.Suche in Google Scholar PubMed
127. Alborelli, I, Leonards, K, Rothschild, SI, Leuenberger, LP, Savic Prince, S, Mertz, KD, et al.. Tumor mutational burden assessed by targeted NGS predicts clinical benefit from immune checkpoint inhibitors in non-small cell lung cancer. J Pathol 2020;250:19–29. https://doi.org/10.1002/path.5344.Suche in Google Scholar PubMed PubMed Central
128. Rizvi, NA, Hellmann, MD, Snyder, A, Kvistborg, P, Makarov, V, Havel, JJ, et al.. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 2015;348:124–8. https://doi.org/10.1126/science.aaa1348.Suche in Google Scholar PubMed PubMed Central
129. Frelaut, M, Le Tourneau, C, Borcoman, E. Hyperprogression under immunotherapy. Int J Mol Sci 2019;20:2674. https://doi.org/10.3390/ijms20112674.Suche in Google Scholar PubMed PubMed Central
130. Indini, A, Rijavec, E, Grossi, F. Circulating biomarkers of response and toxicity of immunotherapy in advanced non-small cell lung cancer (NSCLC): a comprehensive review. Cancers (Basel) 2021;13:1794. https://doi.org/10.3390/cancers13081794.Suche in Google Scholar PubMed PubMed Central
131. Tamminga, M, de Wit, S, Hiltermann, TJN, Timens, W, Schuuring, E, Terstappen, L, et al.. Circulating tumor cells in advanced non-small cell lung cancer patients are associated with worse tumor response to checkpoint inhibitors. J Immunother Cancer 2019;7:173. https://doi.org/10.1186/s40425-019-0649-2.Suche in Google Scholar PubMed PubMed Central
132. Cases, M, Ritter, N, Rincon-Arevalo, H, Kroh, S, Adam, A, Kirchner, M, et al.. Novel non-coding FOXP3 transcript isoform associated to potential transcriptional interference in human regulatory T cells. RNA Biol 2025;22:1–20. https://doi.org/10.1080/15476286.2025.2502719.Suche in Google Scholar PubMed PubMed Central
133. Koh, J, Hur, JY, Lee, KY, Kim, MS, Heo, JY, Ku, BM, et al.. Regulatory (FoxP3+) T cells and TGF-β predict the response to anti-PD-1 immunotherapy in patients with non-small cell lung cancer. Sci Rep 2020;10:18994. https://doi.org/10.1038/s41598-020-76130-1.Suche in Google Scholar PubMed PubMed Central
134. Cortellini, A, Ricciuti, B, Borghaei, H, Naqash, AR, D’Alessio, A, Fulgenzi, CAM, et al.. Differential prognostic effect of systemic inflammation in patients with non-small cell lung cancer treated with immunotherapy or chemotherapy: a post hoc analysis of the phase 3 OAK trial. Cancer Am Cancer Soc 2022;128:3067–79. https://doi.org/10.1002/cncr.34348.Suche in Google Scholar PubMed PubMed Central
135. Li, Z, Yi, L, Gao, P, Zhang, R, Li, J. The cornerstone of integrating circulating tumor DNA into cancer management. Biochim Biophys Acta Rev Cancer 2019;1871:1–11. https://doi.org/10.1016/j.bbcan.2018.11.002.Suche in Google Scholar PubMed
136. Bettegowda, C, Sausen, M, Leary, RJ, Kinde, I, Wang, Y, Agrawal, N, et al.. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci Transl Med 2014;6:224ra24. https://doi.org/10.1126/scitranslmed.3007094.Suche in Google Scholar PubMed PubMed Central
137. Wan, JCM, Massie, C, Garcia-Corbacho, J, Mouliere, F, Brenton, JD, Caldas, C, et al.. Liquid biopsies come of age: towards implementation of circulating tumour DNA. Nat Rev Cancer 2017;17:223–38. https://doi.org/10.1038/nrc.2017.7.Suche in Google Scholar PubMed
138. Torga, G, Pienta, KJ. Patient-paired sample congruence between 2 commercial liquid biopsy tests. JAMA Oncol 2018;4:868–70. https://doi.org/10.1001/jamaoncol.2017.4027.Suche in Google Scholar PubMed PubMed Central
139. Ye, Q, Ling, S, Zheng, S, Xu, X. Liquid biopsy in hepatocellular carcinoma: circulating tumor cells and circulating tumor DNA. Mol Cancer 2019;18:114. https://doi.org/10.1186/s12943-019-1043-x.Suche in Google Scholar PubMed PubMed Central
140. Ignatiadis, M, Sledge, GW, Jeffrey, SS. Liquid biopsy enters the clinic - implementation issues and future challenges. Nat Rev Clin Oncol 2021;18:297–312. https://doi.org/10.1038/s41571-020-00457-x.Suche in Google Scholar PubMed
141. Feng, Z, Wu, J, Lu, Y, Chan, YT, Zhang, C, Wang, D, et al.. Circulating tumor cells in the early detection of human cancers. Int J Biol Sci 2022;18:3251–65. https://doi.org/10.7150/ijbs.71768.Suche in Google Scholar PubMed PubMed Central
142. Malapelle, U, Leighl, N, Addeo, A, Hershkovitz, D, Hochmair, MJ, Khorshid, O, et al.. Recommendations for reporting tissue and circulating tumour (ct)DNA next-generation sequencing results in non-small cell lung cancer. Br J Cancer 2024;131:212–9. https://doi.org/10.1038/s41416-024-02709-4.Suche in Google Scholar PubMed PubMed Central
143. Hofmann, L, Sallinger, K, Haudum, C, Smolle, M, Heitzer, E, Moser, T, et al.. A multi-analyte approach for improved sensitivity of liquid biopsies in prostate cancer. Cancers (Basel) 2020;12:2247. https://doi.org/10.3390/cancers12082247.Suche in Google Scholar PubMed PubMed Central
© 2025 the author(s), published by De Gruyter on behalf of Tech Science Press (TSP)
This work is licensed under the Creative Commons Attribution 4.0 International License.
Artikel in diesem Heft
- Frontmatter
- Review Articles
- Liquid biopsy – a promising and effective method for surveying non-small cell lung cancer minimal residual diseases and anti-cancer drug response after treatment
- Current application status of proton beam therapy for gastrointestinal tumors
- Research progress on the regulation of cuproptosis-related genes by non-coding RNAs in tumors
- Deep learning in hepatic oncology imaging: a narrative review of computed tomography applications
- Synergistic approaches: a narrative mini-review of radiotherapy and immunotherapy in the treatment of lung cancer
- Research Articles
- Intravesical prostatic protrusion as a predictor of acute urinary retention following stereotactic body radiation therapy for localised prostate cancer: a retrospective study
- The differential effect of glutamine supplementation on the orthotopic and subcutaneous growth of two syngeneic murine models of glioma
- Intermittent afatinib treatment suppresses the growth of resistant T790M-H1975 cells in non-small cell lung cancer (NSCLC) co-culture
- Prognostic stratification of colorectal cancer by immune profiling reveals SPP1 as a key indicator for tumor immune status
- The activity of base excision repair is positively correlated with the infiltration of CD4+ T cells in melanoma
- Integrated analysis of immunity and ferroptosis related tumor microenvironment in a novel risk score model for lung adenocarcinoma prognosis
- Retrospective analysis of risk factors for early recurrence after hepatocellular carcinoma resection
- The ENST00000539930 transcript predicts sensitivity to PARP inhibitors and clinical prognosis in cancers
- VTA1 and breast cancer: a potential indicator for diagnostic and prognostic evaluation
- USP24 stabilizes VDAC2 via deubiquitination to promote apoptosis and ferroptosis in clear cell renal cell carcinoma (ccRCC)
- Clinicopathological characteristics, prognosis, and therapeutic implications in breast cancer with pathologically confirmed bone marrow metastases: an observational retrospective study
- Short Commentaries
- Cancer cell mitochondria: the missing puzzle in predicting response to PD-1/PD-L1 inhibitors
- From mitochondrial cristae pathobiology to metabolic reprogramming in cancer: the α and ω of Malignancies?
- Article Commentary
- Stopping SOAT1 sparks an immune attack on liver cancer: a metabolic-immune axis in hepatocellular carcinoma
Artikel in diesem Heft
- Frontmatter
- Review Articles
- Liquid biopsy – a promising and effective method for surveying non-small cell lung cancer minimal residual diseases and anti-cancer drug response after treatment
- Current application status of proton beam therapy for gastrointestinal tumors
- Research progress on the regulation of cuproptosis-related genes by non-coding RNAs in tumors
- Deep learning in hepatic oncology imaging: a narrative review of computed tomography applications
- Synergistic approaches: a narrative mini-review of radiotherapy and immunotherapy in the treatment of lung cancer
- Research Articles
- Intravesical prostatic protrusion as a predictor of acute urinary retention following stereotactic body radiation therapy for localised prostate cancer: a retrospective study
- The differential effect of glutamine supplementation on the orthotopic and subcutaneous growth of two syngeneic murine models of glioma
- Intermittent afatinib treatment suppresses the growth of resistant T790M-H1975 cells in non-small cell lung cancer (NSCLC) co-culture
- Prognostic stratification of colorectal cancer by immune profiling reveals SPP1 as a key indicator for tumor immune status
- The activity of base excision repair is positively correlated with the infiltration of CD4+ T cells in melanoma
- Integrated analysis of immunity and ferroptosis related tumor microenvironment in a novel risk score model for lung adenocarcinoma prognosis
- Retrospective analysis of risk factors for early recurrence after hepatocellular carcinoma resection
- The ENST00000539930 transcript predicts sensitivity to PARP inhibitors and clinical prognosis in cancers
- VTA1 and breast cancer: a potential indicator for diagnostic and prognostic evaluation
- USP24 stabilizes VDAC2 via deubiquitination to promote apoptosis and ferroptosis in clear cell renal cell carcinoma (ccRCC)
- Clinicopathological characteristics, prognosis, and therapeutic implications in breast cancer with pathologically confirmed bone marrow metastases: an observational retrospective study
- Short Commentaries
- Cancer cell mitochondria: the missing puzzle in predicting response to PD-1/PD-L1 inhibitors
- From mitochondrial cristae pathobiology to metabolic reprogramming in cancer: the α and ω of Malignancies?
- Article Commentary
- Stopping SOAT1 sparks an immune attack on liver cancer: a metabolic-immune axis in hepatocellular carcinoma