You are currently viewing a new version of our website. To view the old version click .
International Journal of Molecular Sciences
  • Review
  • Open Access

14 December 2025

Blood-Based miRNA Panels for Timely Detection of Non-Small-Cell Lung Cancer: From Biomarker Discovery to Clinical Translation

,
,
,
,
,
,
and
1
Institute of Chemical Biology and Fundamental Medicine Siberian Branch Russian Academy of Sciences ICBFM SB RAS, Academician Lavrentyev Avenue 8, Novosibirsk 630090, Russia
2
Faculty of Natural Sciences, Novosibirsk State University, Novosibirsk 630090, Russia
3
Research Institute of Pulmonology of the Federal Medical-Biological Agency of the Russian Federation, Orekhoviy Boulevard 28, Moscow 115682, Russia
*
Authors to whom correspondence should be addressed.
This article belongs to the Special Issue Molecular Perspectives in Lung Diseases: Pathogenesis, Diagnosis, and Treatment

Abstract

Lung cancer (LC) remains a leading cause of global cancer mortality, driving the need for novel timely detection strategies, i.e., stages I–II detection when tumor curation is efficient. Circulating microRNA (miRNAs), with their unique stability in biofluids, offer a powerful approach for non-invasive detection. This review compiles validated miRNAs implicated in the early stages of non-small-cell lung cancer (NSCLC), elucidating their roles in key oncogenic pathways such as epithelial-mesenchymal transition (EMT), PI3K/AKT/mTOR, and JAK-STAT, which regulate proliferation, apoptosis, and metastasis. Furthermore, we critically evaluate developed miRNA panels with a specific focus on advanced quantification and normalization strategies, including exogenous spike-in controls and data-driven methods like pairwise normalization, to enhance diagnostic accuracy. Consequently, we identify and rank the most viable miRNA candidates according to key analytical and clinical metrics, providing a clear roadmap for translating these biomarkers into effective panels for the timely detection of NSCLC.

1. Introduction

Since lung cancer (LC) is considered one of the most common and deadliest types of cancer, great attention is paid to its diagnosis and treatment [1]. The vast gas exchange surface of the lungs, while facilitating exposure to carcinogens like tobacco smoke, also renders early-stage lesions exceptionally difficult to detect with conventional methods [2,3]. The risk of lung cancer development is 20–40 times higher in lifelong smokers compared to non-smokers. Moreover, second-hand smoke exposure increases the risk of lung cancer by more than 20% among never-smokers [4]. Of course, heredity, somatic mutations and immune system failures contribute to the development of lung cancer, whether related to the contact of lung cells with chemical or physical carcinogens or not [5]. With all this, lung cancer usually develops asymptomatically, and 60–70% of patients are at advanced stage IV at the time of diagnosis [6]. The 5-year survival rate for LC patients is 80–85% at stage I [7], and 63.1% in pN0, 51.9% in pN1, and 33.5% in pN2 of stage II [8], but at the later stages it tends to be 16% [6], demonstrating an urgent need for early lung cancer screening. No less than 80% of all LC cases are non-small-cell LC (NSCLC, including adenocarcinoma (AD) and squamous cell carcinoma (SCC)), with the remaining 10–15% being small-cell lung cancer (according to the American Cancer Society [cancer.org |1.800.227.2345]). In this review, we will concentrate on early-stage NSCLC diagnostics using promising molecular markers from the circulating blood.
A number of methods are used for NSCLC diagnostics, including CT, MRI, PET, IA (immunoassay), IGH (immunohistochemistry), FISH, mass spectroscopy assay of volatile organic compounds (VOCs), and liquid biopsy methods [9,10]. Computed tomography (CT) has been shown to be a very convenient method for early NSCLC screening of smokers, and it demonstrates a 20% reduction in lung cancer mortality with annual CT screening [11]. Nevertheless, limited access to modern imaging equipment and qualified personnel, cost and time of the procedure, and high false-positive rates (up to almost 50%) [12] do not allow this procedure to be considered a screening test. MRI and PET have similar limitations (except for the specificity of PET). A separate problem is the spread of lung cancer among non-smokers. In Asia, for example, in recent years, 45% to 70% of all patients with lung cancer in this population group have been non-smokers [13]. Most of the listed methods used to diagnose NSCLC cannot be considered screening methods, and at present, only immunoassay of tumor markers, analysis of tumor-related nucleic acids using Polymerase Chain Reaction (PCR) as well as other areas of liquid biopsy and, possibly, mass spectrometry analysis of volatile metabolites can be applied for LC screening. Immunoassay is inexpensive and allows for the analysis of a large number of samples, but the analysis of NSCLC-specific markers, such as CEA (carcinoembryonic antigen), NSE (neuron-specific enolase), CYFRA 21-1 (a soluble fragment of cytokeratin 19 associated with epithelial cell tumors), and SCCA (squamous cell carcinoma antigen), does not have sufficient sensitivity and specificity for early LC detection [14]. The analysis of volatile organic compounds (VOCs) using mass spectroscopy could serve as a screening tool for lung cancer due to its rapidity and technical simplicity [15]. However, there is still not a set of VOC biomarkers that are generally accepted and suitable for diagnosing LC [16], not to mention the possible limitations in terms of the specificity of such tests [17]. Liquid biopsy involves the examination of extracellular fluids, primarily blood, for tumor-specific markers such as cell-free DNA and RNAs, circulating tumor cells, and extracellular vesicles (EVs), as well as other circulating molecular and cellular analytes, which offer promising opportunities for cancer diagnosis [18]. Nucleic acids studied by different variants of PCR and next-generation sequencing (NGS) represent the main diagnostic material of liquid biopsy. Moreover, few tests like the Epi proLung® BL Reflex Assay (Epigenomics AG, Berlin, Germany) [19] or Lepu Medical Methylation Kit (Lepu Medical Technology Co., Ltd., Beijing, China) have a CE-IVD and China NMPA approval (but not FDA). There are also few officially approved NGS panels for the detection of mutations and rearrangements in genes involved in LC development and crucial for the selection of LC-targeted therapy, but they are not intended for use for the early diagnosis of LC [20,21]. The limited number of diagnostic kits and the lack of their widespread use make the search for early LC diagnosis markers and new early LC diagnostic tests an urgent task. Such LC screening tests must be robust and inexpensive, such as liquid biopsy. The tests should not lead to overdiagnosis, detection of cancer at the precancerous stage, or cancer in situ but should reliably detect cancer at the early stages of development, when current protocols of LC curation are to be applied and patients can be cured of LC. In one breakthrough retrospective study, the authors were able to predict the incidence of cancer up to four years before its clinical manifestation [22]. However, it is not yet clear whether such data would improve outcomes or whether the negative consequences of such data would outweigh the positive ones.
In this regard, the issue of early markers involves several key considerations. First, markers found exclusively or predominantly at the early stages are strong candidates for early-stage detection. In contrast, markers specific to later stages can be used in conjugation for accurate tumor staging. Furthermore, markers that appear early and persist throughout cancer progression are prime candidates for early diagnostics as their expression may be independent of tumor stage, remaining consistent as the disease advances.
Other markers may be linked to tumor type or heterogeneity yet still serve as good indicators of early-stage cancer. It is obvious that the markers involved in the main pathways of tumor maintenance, especially those frequently encountered across different phenotypes and heterogeneities, are also strong candidates for the timely diagnosis of NSCLC.
In other words, various markers can be used for timely NSCLC diagnosis; however, markers that occur in the early stages may be more promising since they do not require additional preliminary studies involving groups of patients in the early stages, mainly before verification. Additionally, screening potential markers based on their involvement in non-overlapping regulatory pathways, clustering, frequency of occurrence, and features of analytical systems (specificity and multiplication in one probe) is highly valuable for selecting the most promising markers for a diagnostic panel.
For the timely diagnostics of NSCLC, cell-free microRNA (cf-miRNA) presents a promising tool. These nucleic acids are released by tumor cells into the circulation, making them highly accessible biomarkers in blood plasma and a core component of liquid biopsy for NSCLC detection that can easily be isolated and quantified [23,24]. Tumor-specific microRNAs were found at the early and late stages of NSCLC in numerous studies [25]. While early NSCLC diagnostics relied on a single microRNA [26], later research proposed diagnostic panels comprising dozens of different miRNAs [27]. Significant heterogeneity in miRNA selection criteria and experimental methodologies has led to a lack of consensus among studies, with few miRNAs being independently validated.
This review addresses the following question: “Which circulating cell-free miRNAs show the strongest diagnostic potential for early-stage NSCLC?”. To answer this, we provide a novel integrative analysis of validated studies. We analyze data on miRNA occurrence across stages, functional roles in tumorigenesis, normalization methods, and diagnostic performance, based on multiple panels, to critically evaluate and identify the most promising miRNA sets for early detection panels.

2. Cell-Free miRNAs as Promising Biomarkers for Early-Stage NSCLC

A distinct spectrum of cell-free miRNAs (cf-miRNA) has been characterized in the circulating blood of patients with early-stage NSCLC. A subset of these cf-miRNAs was initially discovered through large-scale screening methods like microarray platforms or NGS. However, the presence of the majority of candidate cf-miRNAs has been confirmed using more targeted methods such as PCR. The latter were selected from large-scale data as well as data on miRNA expression in different cell types or synthetic data obtained by bioinformatic methods or via looking through a database [28]. In principle, different analytical methods should generate consistent data on microRNA identity and abundance. However, the results of different analytical platforms often converge. A major contributing factor is the absence of universal protocols, which leads to significant variability in the efficacy and reproducibility of miRNA isolation, analysis, and data normalization.
While miRNAs can be isolated from various biological sources like sputum and bronchoalveolar lavage (BAL), this review focuses exclusively on blood-based samples due to their universality and minimally invasive nature, which are critical for widespread early-stage screening [29]. Here, we will discuss the list of miRNAs found in early NSCLC patient’s blood. microRNAs will be ranked by frequency of occurrence in different studies (Table 1), and the key features of the study, including sample type (plasma, serum or microvesicles), the analytical method used, and the normalization method, will be mentioned for subsequent discussion of the applicability of the marker in further sections of the review (Table 2).
Table 1. Most frequently reported miRNAs in early-stage NSCLC detection studies.
The microRNAs miR-145 and miR-21 are among the most frequently identified biomarkers in studies of NSCLC. In particular, miR-21 has been consistently implicated in the carcinogenic process, showing elevated levels especially at early disease stages [30,31,32,33]. Multiple studies employing quantitative RT-PCR with different normalization controls (miR-16, RNU66, miR-197) have demonstrated that plasma and tissue levels of miR-21 tend to increase in NSCLC vs. healthy donors [30,31,32]. However, contradictory results exist regarding whether miR-21 expression correlates precisely with NSCLC stage. For example, it was found that patients with advanced stages (III–IV) exhibit significantly higher plasma miR-21 expression than those in early stages (I–II), and elevated tissue expression correlates with disease development even within stage I patients across diverse cohorts. Conversely, another study showed higher miR-21 serum levels in stage I patients compared to the controls; it found no significant distinction between early and later stages, indicating that miR-21 expression stabilizes after initial increase [32]. Overall, these converging findings suggest that miR-21 is a promising biomarker for early NSCLC detection, particularly effective in identifying squamous cell carcinoma [33].
miR-145 is a well-established regulatory molecule and a validated biomarker for its early detection in NSCLC. Findings using TaqMan miRNA assays with RNU48, miR-191, and cel-miR-39 as normalized controls demonstrated that miR-145 expression levels were significantly downregulated in the plasma of NSCLC patients compared to healthy controls [34,35,36]. However, the correlation between miR-145 expression and disease stage presents inconsistent results. While several studies reported no significant difference in the expression levels across the NSCLC stages [34,36,37], others observed a progressive decrease in miR-145 with advancing disease [38]. Despite this discrepancy regarding stages, the consistent downregulation in patients supports the potential of miR-145 as a valuable tool for the early detection of NSCLC.
Researchers frequently studied miR-126 level as well for its diagnostic potential in early-stage NSCLC. In studies utilizing TaqMan miRNA assays normalized to references such as U6 and cel-miR-39, the results indicate that miR-126 is generally downregulated in NSCLC patients compared to healthy controls, with this downregulation becoming more pronounced in advanced stages [39]. Additionally, miR-126-3p was evaluated in the serum of NSCLC patients in the early stages and demonstrated an independent prognostic role in SCC by significantly associating with disease-free survival (DFS); however, no such significant association was observed in the AD cohort, highlighting a histology-specific function for this miRNA [40].
Several reports have investigated the roles of miR-223, miR-20a, and miR-155 as potential diagnostic biomarkers for early-stage NSCLC. They used quantitative real-time PCR (RT-qPCR) with U6 snRNA for normalization. The results consistently showed that the expression levels of these miRNAs were elevated in patients with early-stage NSCLC compared to healthy controls [33,41,42]. Furthermore, the level of miR-155 was found to increase progressively with advancing disease stage, suggesting its potential utility not only in diagnostics but also in monitoring disease progression [43].
Numerous miRNAs have been reported in two or more independent studies as promising diagnostic biomarkers for detecting early-stage NSCLC. The downregulation of let-7 in NSCLC is a hallmark event linked to tumor initiation, progression, and poor prognosis, making it a compelling candidate for diagnostic and prognostic biomarker development [44]. One study reported the decreased expression levels of let-7a in stage I–II NSCLC patients [45]. In a separate study, Let-7b expression levels varied significantly between NSCLC patients with stage IA/B and the controls by measuring serum and plasma levels with Taq-man miRNA assays using cel-miR-54 and cel-miR-238 for normalization [46]. A study on the miR-200 family in NSCLC demonstrated that miR-200a, miR-200b, miR-200c, and miR-429 were significantly downregulated, while miR-141 was upregulated in NSCLC tissue samples in comparison with the control [47]. Separately, miR-125b was found to be elevated in stage I AD patient samples [48]. In contrast, miR-146a was downregulated in cancer tissues overall but was expressed at significantly higher levels in stage I–II samples compared to stage III–IV samples [49]. A comprehensive summary of these and additional findings is provided in Table 2.
Table 2. List of miRNAs validated for detection of early-stage NSCLC.
Table 2. List of miRNAs validated for detection of early-stage NSCLC.
miRNAStudy ModelSample TypeAnalytical TechniqueNormalizationRef.RegulationBiological Processes RegulatedTargetExpression ChangesRef.
miR-2163 NSCLC patients.
Stages: I–IV
Mouse model
PlasmaRT-qPCRmiR-16[31]Cell cycle
DNA repair
Apoptosis
Angiogenesis
Proteolysis
Cell adhesion
MAPK/ERK pathway
TGFβ pathway
G-protein pathway
cell growth and resistance to apoptosis
STAG2, KIF6
MSH2, FANCC, CHD7
PDCD4, APAF1, STAT3, MALT1, SGK3
SOS2, JAG1, MAP3K1, STAT3
WWP1
CCL1, MATN2, TGFBI, VCL
MAP3K1, STAT3, SOS2, NKIRAS1, SPRY1, SPRY2
BMPR2, SMAD7
SOS2, TIAM2, GPR64, KRIT1
PTEN
↑ in stage I compared to control group.
↑ as NSCLC progressed.
[31,32,43,50,51]
Retrospective analysis of three cohorts included 317 NSCLC patients.
Stages: I–III
Tissue samplesTaqMan miRNA assayRNU66[30]
miR-14580 NSCLC patients.
Stages: I–IV
Plasma,
Tissue samples
TaqMan miRNA assayRNU48[34]Proliferation, EMT, Migration, Invasion
Metastasis, Immune evasion
Cell cycle
Apoptosis
Tumor growth
GOLM1, RTKN, SOX9
CXCL3
c-Myc
Caspase-3/-9, MTDH
EGFR, NUDT1
Study A: ↓ in all patients, regardless of clinical stage [36].
Study B: ↓ as NSCLC progressed [38].
[34,52,53,54,55,56]
70 paired normal and NSCLC tissues.
Stages: I–III
Tissue samplesTaqMan miRNA assaymiR-191[35]
miR-12645 NSCLC patients.
Stages: I–IV
Serum,
Exosomal,
Exosomal-free
TaqMan miRNA assay
and RT-qPCR
U6
cel miR-39
[39]Angiogenesis
VEGF-A/VEGFR-2/ERK
PI3K/AKT signaling
mTOR signaling
EMT Invasion
EGFL7, IRS-1, Crk, VEGFA
PIK3R2
LAT1
SOX11, PLOD2
↓ in advanced stage patients compared to early-stage patients [39].[57,58,59]
miR-126-3p182 NSCLC patients.
Stages: IA–IIIA
SerumRT-qPCRcel-miR-39[40]
miR-15550 NSCLC patients and 50 healthy volunteers.
Early stages
SerumRT-qPCRMean of CT of all healthy controls[42]Tumor growth Metastasis
Migration Invasion
PTEN, SOCS1, SOCS6
Smad2
↑ as NSCLC progressed.[43,60]
80 paired normal and NSCLC tissues.
Stages: I–III
Tissue samplesRT-qPCRU6[43]
miR-205265 NSCLC patients.
Stage: I
Tissue samplesTaqMan miRNA assayRNU6B[61]Growth
metastasis
EMT
Apoptosis
NF-κB signaling
Proliferation
PTEN, TP53INP1
Cripto-1
APBB2
EGFR
↑ as NSCLC progressed.[49,61,62,63,64]
miR-150171 NSCLC patients.
Stages: I–II
SerumStem-loop array reverse transcription PCR (SLA-RT-PCR)U6 snRNA[65]Proliferation
Metastasis
SRCIN1, P-STAT3, ROS, EPG5
FOXO4
↑ as NSCLC progressed.[65,66,67,68,69]
miR-141155 NSCLC patients.
Stages: I–III
Tissue samplesTaqMan miRNA assayRNU44 RNU48[47]AngiogenesisKLF6, VEGFAStudy A: no correlations between the expression level and TNM stage [70].
Study B: ↑ as NSCLC progressed [71].
[47]
miR-146a101 NSCLC patients.
Stages: I–IV
Tissue samplesRT-qPCRRNU6B [49]Proliferation
EMT
Migration
Survivability
EGFR, TNF-α, NF-κB and MEK-1/2, and JNK-1/2.
Notch2
TRAF6
↓ in advanced stage patients compared to early-stage patients.[49,72,73]
miR-22375 NSCLC patients and 111 tumor-free controls.
Stages: I–II
Serumdroplet digital PCR (ddPCR)UniSp6/cel-miR-39-3p [74]PI3K/AKT pathway
Proliferation and invasion
Migration
EGFR
IGF-1R
NLRP3, E2F8
↓ in advanced stage patients compared to early-stage patients.[74,75,76,77,78]
31 NSCLC patients.
Stages: I–IV
3 NSCLC cell lines.
Mouse model.
SerumReal-Time PCR
U6 snRNA[78]
miR-34b/c140 AC patients.
Stages: I–II
15 human lung AC cell lines.
Tissue samplesRT-qPCRRNU48[79]Tumor growth
Cell cycle
Apoptosis
EMT
TP53
CDK4, CDK6, CCND1
BCL2, SIRT1
Zeb1
N/A[80]
miR-18333 AD patients.
Stage: I
2 NSCLC cell lines.
Tissue samplesRT-qPCRU6 snRNA[81]mTOR regulation
Proliferation, migration and cell cycle
Metastasis
SESN1
PTEN, FOXO1
LOXL4
N/A[81,82,83,84]
miR-1246105 NSCLC patients, 50 patients with NMRD and 50 healthy volunteers.
Stages: I–IV
SerumRT-qPCR
cel-miR-39[85]Stemness
Metastasis, Wnt/β-Catenin Pathway
Radioresistance
TRIM17
CPEB4, GSK-3β
DR5
↑ in advanced stage patients compared to early-stage patients.[85,86,87,88,89]
miR-328-5P86 NSCLC patients and 24 healthy donors.
Stages: I–IV.
Peripheral blood cellsTaqMan miRNA assayRNU38B, RNU58A[90]MigrationPRKCA, IL-1beta, c-Raf1, LOXL4N/A[90,91]
miR-328-3PGenomic stabilityH2AX↓ as NSCLC progressed.[92]
miR-200168 NSCLC patients and 128 patients with benign lung nodules.
Stages: I–II
Plasma
(EV-derived)
RT-qPCRcel-miR-39-3p[93]EMT through Notch signalingNotch ligand Jagged1 and Jagged2, Flt1
PTEN, ABCA1
Stage II patients exhibited the highest expression level compared to the other stages.[94,95,96,97,98,99,100,101,102]
Let-7b220 NSCLC patients and 220 healthy controls. Stages: IA–IIBPlasmaTaqMan miRNA assayCel-miR-54 Cel-miR-238[46]MAPK/ERK pathway
Immune response
BRF2
PD-L1
N/A[103,104,105]
Serum
miR-4732-5p18 AD patients and 18 BPN patients. Stage: I.Serum
(EV-derived)
RT-qPCRmiR-20a[106]Migration
EMT
TSPAN13
XPR1, PI3K/Akt/GSK3β/Snail pathway
↑ as NSCLC progressed.[107,108]
miR-374a38 NSCLC patients and 27 Heathy controls Stages: IA–IIBTissue samplesTaqMan miRNA assay RNU48, miR-16 and miR-26b[109]Metastasis
Proliferation
γ-adducin
NCK1
N/A[110,111]
↑ upregulation/higher expression of miRNA. ↓ downregulation/lower expression of miRNA.

3. Blood-Based microRNA Biomarkers: Functional Implications in NSCLC

As was mentioned, miR-145, functioning as a key tumor suppressor, is frequently downregulated in the plasma of early-stage NSCLC patients as it acts as a pivotal regulator of proliferation, apoptosis, and migration of tumor cells. miR-145 directly targets EGFR (Epidermal Growth Factor Receptor) and NUDT1 (Nudix Hydrolase 1), thereby inhibiting downstream oncogenic signaling pathways, mainly EGFR/PI3K/AKT, and suppressing tumor growth [52]. Moreover, miR-145 suppresses the cell cycle through inhibiting c-Myc and CLR4, which leads to the deactivation of the c-Myc/eIF4E pathway, downregulation of cell cycle regulators such as Cyclin D1, and arresting the cell cycle [53]. Additionally, the metastatic potential of NSCLC is suppressed by miR-145 through its inhibition of the Oct4-mediated Wnt/β-catenin signaling pathway and EMT [112]. miR-145 also promotes the apoptosis process in NSCLC cells by activating the caspase cascade primarily through targeting caspase-3 and by modulating the Bcl-2 family [113]. This regulation of multiple critical pathways highlights the significance of miR-145 as a potential timely biomarker in early-stage NSCLC.
miR-21 plays a central oncogenic role in the development of NSCLC, particularly from the early stages of the disease. Its regulation of key targets such as PTEN and PDCD4 leads to activation of the PI3K/AKT/mTOR signaling pathway, enhancing tumor cell proliferation and disrupting normal differentiation [50,114]. Additionally, it was suggested that miR-21 influences the tumor microenvironment by modulating antioxidant enzymes like SOD2 and GPx1, as well as signaling suppressors SOCS1 and SOCS6, thereby promoting conditions favorable for tumor growth [43,115]. A critical aspect of NSCLC pathogenesis is the reciprocal regulation between miR-21 and the hypoxia-inducible factor HIF-1α, which drives metabolic adaptation by increasing glycolysis through HIF-1α-induced enzymes and simultaneously supporting angiogenesis via the upregulation of vascular factors such as VEGF, as shown in vitro [116,117,118].
This coordinated regulation of metabolism and blood vessel formation facilitates tumor progression and contributes to therapy resistance. Elevated HIF-1α levels correlate with advanced tumor stage and poor clinical outcomes, emphasizing the clinical importance of miR-21’s interaction with hypoxia pathways in NSCLC growth. Overall, these findings highlight miR-21 as a multifaceted regulator that orchestrates the key signaling and metabolic pathways driving NSCLC progression, making it a promising target for early intervention and therapeutic strategies.
miR-126 is a critical tumor suppressor and master regulator of angiogenesis in lung cancer. Several studies proposed that miR-126 functions by directly inhibiting key pro-angiogenic targets, including EGFL7, IRS-1, Crk, VEGF-A, and, most importantly, the LAT1-mediated mTOR signaling pathway [57,58]. Additionally, miR-126 is responsible for inducing the programmed cell death pathway and inhibiting the metastasis of tumor cells by targeting the VEGF-A/VEGFR-2/ERK and PI3K/AKT pathways. These cascades promote essential steps of angiogenesis, including endothelial cell proliferation and tube formation, which results in increased tumor microvessel density. Overall, miR-126 represents a central node in the angiogenic switch and a potential therapeutic and diagnostic target in early-stage NSCLC [57,119].
In early-stage NSCLC, miR-155 plays opposing roles depending on lung cancer cell type. In AD, one study reported that it acts as an oncogene by suppressing SOCS1 and PTEN tumor suppressors, leading to activated JAK/STAT and PI3K signaling pathways that drive tumor growth and correlate with poor patient outcomes [43]. Conversely, in SCC, miR-155 can function as a tumor suppressor by inhibiting SMAD2 and reducing cancer metastasis, associated with better prognosis in certain patients. This tissue-specific duality complicates its use as a biomarker but highlights its importance in NSCLC biology [60].
miR-205 acts as an oncogene in NSCLC by promoting tumor growth, metastasis, and chemoresistance of tumor cells through targeting PTEN [62]. Additionally, miR-205-3p promotes progression and inhibits apoptosis of the NSCLC cells by targeting APBB2 (Amyloid β Precursor Protein-Binding Family B Member 2), which was associated with worse prognosis in AD patients [63]. Conversely, it was shown that low expression of miR-205 was a negative prognostic indicator in early-stage disease. It identified an inverse correlation between miR-205 and the oncoprotein Cripto-1 (CR-1), which, in turn, activates the pro-tumorigenic pathways (SRC and SMAD) and promotes the EMT pathways, thereby driving metastasis and resistance to targeted therapies such as EGFR-TKIs [61].
The antitumor activity of miR-146a is characterized by suppressing proliferation and promoting apoptosis of NSCLC cells by inhibiting EGFR and the downstream pathways (ERK-1/-2, AKT, and STAT). By targeting the IRAK-1 and NF-kB signaling pathways, miR-146a reduces tumor cell motility and metastasis [49].
The let-7 family of microRNAs, mainly let-7a, -7b, -7c, and -7g, is one of the most studied tumor suppressor miRNA families in lung cancer, particularly in the context of cancer initiation. Its loss removes a critical brake on cell proliferation (MAPK/PI3K), stemness (Wnt/β-catenin), and pro-tumorigenic inflammation (STAT3/NF-κB) [120]. Therefore, the expression level of let-7 miRNAs serves as a powerful biomarker: high let-7 activity suppresses these pathways and correlates with a favorable prognosis, while low let-7 activity allows for pathway hyperactivation and predicts aggressive disease and poor survival [121]. The profound regulatory function of the let-7 family makes its members significant targets for future NSCLC early detection.
The miR-34b/c cluster is located on chromosome 11q23 and regulated by p53. It exerts significant tumor suppressor activity in early-stage NSCLC [80]. miR-34 family members induce multiple tumor-suppressive effects, including the inhibition of cell migration, cell cycle arrest, and promotion of apoptosis by directly targeting key oncogenes such as CCNE1, EGFR, and SIRT6 [122]. Furthermore, it inhibits tumor growth and metastasis through the upregulation of PTEN and repression of YY1. In parallel, miR-34a, a related family member, significantly reduces cellular proliferation, induces cell cycle arrest, and promotes senescence in lung cancer cells via direct modulation of the E2F transcription factors E2F1 and E2F3 [123].
Analysis of miRNAs targets in early-stage NSCLC has identified key signaling pathways, mainly PI3K/Akt/mTOR, NF-kB, EGFR, JAK-STAT, and Notch, as being frequently implicated in carcinogenesis. These pathways primarily drive tumor development by regulating critical processes such as cell cycle progression, proliferation, EMT, migration, invasion, angiogenesis, stemness, and apoptosis, as described in Figure 1. Considering their regulatory functions, the aforementioned miRNAs could be used as additional biomarkers to guide targeted therapy. For instance, the mTOR pathway is directly inhibited by Rapamycin and its analogs [124]. Other specific low-molecular-weight kinase inhibitors include Erlotinib targeting EGFR [125] and Ruxolitinib targeting JAK1/2 [126]. Furthermore, high-molecular-weight monoclonal antibodies are generally employed, such as Bevacizumab targeting the VEGF ligand to inhibit angiogenesis [127] and Atezolizumab targeting the PD-L1 ligand on tumor cells to enhance immune response [128]. A synergetic approach combining such targeted agents should be further investigated to potentially limit cancer progression.
Figure 1. Main genes targeted by miRNAs in early-stage NSCLC and main biological processes involved in the carcinogenesis highlighted in purple. Subtypes of the main genes are highlighted in grey.

4. Multi-miRNA Panels: Improving Early NSCLC Detection

The data described demonstrates that microRNAs are emerging as powerful tools in the management of early-stage NSCLC. Considering diagnostic efficacy as well as the variability in miRNA expression depending on tumor type and individual patient features, specific miRNA panels (rather than single miRNAs) are required for reliable cancer diagnostics [25]. The selection of miRNAs for these panels is a precise process rooted in high-throughput sequencing and microarray studies that compare miRNA expression profiles between NSCLC tumor tissue and matched healthy tissue. The criteria for selecting miRNA panels emphasize differential expression, stability, functional relevance, and robust performance metrics. The development process involves discovery, validation, panel construction, clinical evaluation, and blinded testing, supported by bioinformatic analysis. These steps ensure that miRNA panels are reliable, reproducible, and clinically useful for non-invasive diagnostics [129]. miRNAs are selected based on their significant dysregulation (up- or downregulation) in disease states compared to healthy controls. Moreover, they must be stable and reliably detectable in liquid biopsies (e.g., serum, plasma). Furthermore, the chosen miRNAs should be involved in several key cancer pathways (e.g., proliferation, apoptosis, metastasis). In addition, it is desirable that there be no correlation between the expression of microRNA biomarkers and the patient’s gender and age, as well as physiological state (hormonal status, dietary pattern, etc.) [130]. Therefore, the miRNA panels should be evaluated based on multiple factors such as sensitivity, specificity, area under the curve (AUC), positive likelihood ratio (PLR), negative likelihood ratio (NLR), and diagnostic odds ratio (DOR) [131].
Despite their promising results, the panels reviewed here share common limitations that affect their real-world reliability. Some studies, like the one performed by Zhong et al. (2021) [132], used relevant control groups (including benign nodules) but were limited by small or non-diverse cohorts. Others, like the one performed by Ying et al. (2020) [133], enrolled large, multi-ethnic populations but did not test specifically against benign conditions, since distinguishing cancer from benign nodules is the main challenge in screening. Blinded validation was also inconsistently applied. To build panels that are both reproducible and clinically useful, future work must prioritize diverse patient groups, include benign disease controls, and use blinded evaluation. Without these steps, the findings will remain difficult to compare or translate into practice.
A key challenge for miRNA panels in NSCLC detection is ensuring high specificity as many individual miRNAs (e.g., miR-21) are dysregulated across various cancer types, potentially leading to false positives. While miRNA panels show strong sensitivity for lung cancer, they must be carefully validated to differ between NSCLC and other malignancies, resulting in an accurate disease-specific detection [134]. However, this does not imply that different cancer types share an identical global cf-miRNA expression profile. A promising solution to this problem is paired normalization within a miRNA panel. This approach involves analyzing the ratios between specific miRNA pairs rather than relying solely on the absolute levels of individual miRNAs. This can significantly improve the accuracy of disease-specific detection and differentiate NSCLC from other malignancies [135].
The growing body of evidence demonstrates that miRNA panels hold great promise as non-invasive diagnostic tools for early-stage NSCLC. The rigorous selection process, which integrates high-throughput sequencing, functional pathway relevance, and advanced bioinformatic analyses, ensures that candidate miRNAs exhibit both biological significance and clinical utility. Multiple studies, some mentioned in Table 3, have validated distinct miRNA signatures with high diagnostic accuracy, achieving impressive AUC values often exceeding 0.85 to 0.97, along with strong sensitivity and specificity across diverse patient cohorts and NSCLC subtypes. The composite score for each diagnostic panel was calculated using the following formula: AUC + (specificity/100) + (sensitivity/100). This metric provides an integrated assessment of the panel’s overall performance. For instance, the panel proposed by Zhong et al. (2021) achieved a score of 2.385 [132], which is lower than the score of 2.867 [135] attained by the panel from Wang, Y. et al.’s study (2016) [136].
Notably, subtype-specific panels targeting AD and SCC have improved diagnostic granularity by capturing unique molecular profiles within heterogeneous NSCLC populations.
The use of liquid biopsies such as serum and plasma samples for miRNA detection emphasizes the clinical feasibility of these panels for routine screening and early diagnosis. However, variability in performance metrics across studies highlights the urgent need for standardization of sample processing, normalization controls, and diagnostic thresholds. While miRNA panels have shown superior diagnostic performance compared to single miRNA markers, challenges remain in establishing universally accepted panels suitable for clinical application. Large-scale, prospective, multicenter studies are essential to refine panel composition and validate them. The validation of pre-analytical steps (blood processing, isolation of microvesicles, isolation and processing of miRNAs) and standardization of the analytical system, including universal spike-in control and internal control of few stably expressed cf-miRNAs, are demanded for the successful development of NCLC miRNA-based diagnostics.
Table 3. List of blood-based miRNA panels validated for early-stage NSCLC diagnostics.
Table 3. List of blood-based miRNA panels validated for early-stage NSCLC diagnostics.
PanelsmiRNAsStudy CohortSample TypeAUCSensitivitySpecificityPanel ScoreRef.
Zhong, Y. et al. (2021)miR-520c-3p, miR-1274b207 NSCLC patients, 168 healthy controls, 31 benign nodule patientsSerum and plasma0.82382.373.92.385[132]
Wang, P. et al. (2015)miR-125a-5p, miR-25, miR-12694 NSCLC patients and 48 stage III–IV NSCLC patients and 111 healthy controlsSerum0.93687.587.52.686[137]
Zheng, D. et al. (2011)miR-155, miR-197, miR-18274 NSCLC patients and 68 healthy controlsPlasma0.901281.3386.762.5821[138]
Lv, S. et al. (2017)miR-146a, miR-222, miR-223180 AD patients and 180 healthy controlsSerum0.95184.3590.832.7028[139]
Peng, H. et al. (2016)miR-1254, miR-485-5p, miR-574-5pTraining set: 36 NSCLCs vs. 36 controls
validation set: 120 NSCLCs and 71 controls
Serum0.84493.373.22.509[140]
Aiso, T. et al. (2018)miR-145-5p, miR-20a-5p, miR-21-5p56 NSCLC patients and 26 healthy controlsSerum0.89385.7802.55[36]
Ying, L. et al. (2020)let-7a-5p, miR-1-3p, miR-1291, miR-214-3p, miR-375744 NSCLC patients and 944 healthy controls Serum0.93582.990.72.671[133]
Yang, X. et al. (2019)miR-146b, miR-205, miR-29c and miR-30b128 NSCLC patients and 30 healthy controlsSerum0.9695.3182.982.7429[141]
Poh, K.C. et al. (2025)miR-196a-5p, miR-1268, miR-130b-5p, miR-1290, miR-106b-5p, miR-124682 NSCLC patients and 123 healthy controlsSerum0.98992.197.52.885[13]
Wang, Y. et al. (2016)miR-532, miR-628-3p, and miR-425-3p201 early-stage and 25 late-stage AD patients and 43 patients with lung benign disease and 178 healthy controlsPlasma0.97690.298.92.867[136]
Leng, Q. et al. (2017)miR-21, 210, and 486-5p92 LC patients and 88 cancer-free smokersPlasma0.8575.585.32.458[142]
miR-126, miR-145, miR-210, and miR-205-5p0.9691.596.22.837
Abdipourbozorgbaghi, M. et al. (2024)miR-9-3p, miR-96-5p, miR-147b-3p, miR-196a-5p, miR-708-3p, miR-708-5p, miR-4652-5p78 NSCLC patients and 44 healthy controlsPlasma0.8583782.46[143]
miR-130b-3p, miR-269-3p, miR-301a-5p, miR-301b-5p, miR-744-3p, miR-760, miR-767-5p, miR-4652-5p, miR-6499-3p0.8892732.53

5. Conclusions and Perspectives

Comparing measurements of circulating miRNAs from different studies can be challenging as they can be affected by pre-analytical and biological factors. For instance, serum preparation involves coagulation, which induces the lysis of red blood cells, resulting in the release of intracellular RNA, thereby altering the profile of circulating extracellular miRNAs. Consequently, plasma is often favored over serum for miRNA studies as it avoids coagulation and reflects normal circulating miRNAs. Moreover, biological differences such as gender-related variations in circulating miRNA levels should be considered [144]. Besides blood-based samples, respiratory fluids such as sputum and BAL fluid offer a significant advantage for lung cancer-specific miRNA profiling. These samples provide a more localized snapshot of the tumor microenvironment, potentially enriching for lung-derived miRNAs and reducing background noise from systemic sources, but they are less universal and more difficult to standardize [145].
The search for diagnostic microRNAs for detecting lung cancer in the early stages and the assembly of microRNA panels is complicated by the limited number of patients with reliably diagnosed early-stage lung cancer. According to the data in Table 2, the majority of early-stage cancer biomarkers are also present in its late stages. This means that the search for markers can be carried out using plasma samples from patients with late-stage cancer, followed by verification of the most promising diagnostic microRNAs on cf-miRNA samples from patients at stages I and II.
Simultaneously, when selecting miRNA biomarkers for diagnostic or prognostic purposes, it is necessary to verify that their expression is specific to the physiological state and not confounded by other patient variables. Evidence shows that global miRNA expression in blood can be influenced by age [146,147] and hormonal status [148]. Moreover, the presence of the disease itself can shift the physiological miRNA profile [147]. Therefore, it is essential to ensure that the expression of the selected miRNA biomarkers does not correlate with patient characteristics.
The normalization step is critical when quantifying circulating miRNA expression levels as it is essential for differentiating the true biological signals from non-biological variations. Typical normalization methods use endogenous small non-coding RNAs such as miR-16, U6, or SNORD RNAs, which can be poorly suited for fluid-based samples and not suitable for normalization circulating miRNA data [144,149]. The introduction of exogenous spike-in controls for normalization is also used. This involves adding synthetic miRNAs (e.g., from C. elegans) to each sample at the start of RNA extraction and helps control the technical variations in RNA extraction and amplification efficiency [142,150]. For further refinement, data-driven approaches like global mean normalization (using the average of all detected miRNAs) or the mean-endo method (using the geometric mean of a pre-validated panel of stable endogenous miRNAs) can be applied to minimize bias and improve accuracy [151]. However, better normalization methods have been developed, such as pairwise normalization. This advanced strategy abandons the concept of a single reference gene. Instead, it uses the ratio between two miRNAs as a stable, unit-less measurement which cancels out the majority of the technical and non-biological variability that affects both miRNAs in a pair [152].
So far, adapting a universal reference miRNA for blood-based studies is not feasible due to significant biological and technical variability. This variability arises from differences in how miRNAs are packaged and released into circulation, changes in miRNA expression related to disease states, and inconsistencies in sample preparation methods. Therefore, the focus should shift from searching for a single universal normalizer to adopting a standardized method where each study validates its own set of stable reference miRNAs specific to its experimental conditions and cohort. This approach prioritizes reliable and comparable results within a study over broad generalization [153].
The performance variations among panels can be attributed to several factors, including the biological samples used (serum, plasma, or sputum), the selection method for miRNA candidates, the population characteristics (ethnicity, smoking status, cancer stage, physiological state), and the analytical techniques employed for miRNA quantification. For instance, the integrated approach using both sputum and plasma miRNAs demonstrated great performance (87% sensitivity and 89% specificity) compared to panels using single biofluid sources. This enhancement highlights the complementary nature of different biomarker sources and the potential advantage of multi-source sampling strategies [154]. Furthermore, the number of miRNAs in each panel appears to influence diagnostic performance, though not always linearly. While some studies found that smaller panels (3–5 miRNAs) achieved excellent performance [139], others developed more comprehensive signatures (6–9 miRNAs) to maximize diagnostic accuracy [13]. Therefore, the variation suggests that beyond mere quantity, the biological relevance and functional coordination of selected miRNAs within pathways crucial to lung carcinogenesis are paramount considerations for developing effective diagnostic panels [155].
The integration of miRNA biomarkers with current low-dose computed tomography (LD-CT) screening protocols represents the most promising application for improving early NSCLC detection. Research demonstrates that combining miRNA signatures with radiologic imaging significantly enhances diagnostic performance. For instance, a study incorporating six serum miRNA biomarkers with nodule size achieved remarkable AUC values between 0.96 and 0.99, with sensitivities of 92–98% and specificities of 93–98% [13]. This represents a substantial improvement over either approach alone, highlighting the synergistic effect of combining molecular and imaging biomarkers.
While miRNAs such as miR-20a, miR-21, miR-145, and miR-223 are frequently validated individually for the early detection of NSCLC, multi-miRNA panels often demonstrate better diagnostic value. Our analysis of 14 independent panels, listed in Table 3, aimed to identify the most robust biomarkers. To achieve this, for each miRNA, we calculated its mean composite score by averaging the panel scores of all panels in which this miRNA appeared. This provided a metric to rank the miRNAs by their average diagnostic performance across studies. This approach identified miR-1246, miR-1290, miR-130b-5p, miR-532, miR-205, miR-29c, miR-126b, and miR-222 as top-performing candidates (Figure 2) from the chosen panels. These miRNAs can be recommended for inclusion in future panels for the timely detection of NSCLC as they contributed to ensuring high accuracy across the chosen studies.
Figure 2. Ranking of miRNAs by their average performance score in the selected diagnostic panels.
Despite the promising diagnostic performance of multi-miRNA panels in research studies, their translation into clinical diagnostics faces significant challenges, with no panel currently holding FDA or CE approval. This can be due to lack of standardization in pre-analytical and analytical methods, including sample collection, processing, and data normalization, which impairs reproducibility and comparability across studies [156,157]. As mentioned earlier in the article, many candidate panels lack robust validation in large-scale, prospective, and ethnically diverse multicenter cohorts, which is essential to prove generalizability and clinical utility. Finally, the technical complexity and high cost of implementing advanced discovery platforms in routine clinical laboratories present a significant practical challenge [158]. To overcome these challenges, we recommend shifting the focus from individual miRNA biomarkers to the validation of multi-miRNA signatures within prospective, multicenter studies. This requires prospective, blinded validation studies to be performed across multiple centers and diverse populations using standardized methodologies. Such an approach represents a critical first step toward translating these panels into clinical diagnostics.

Author Contributions

P.P.L. designed the framework and supervised the research. Y.Z. performed the literature search, wrote the entire manuscript, and prepared the figures and the tables. M.Y.K. and O.E.B. provided critical review, assisted in manuscript editing, and helped with the figures. A.A.I., Y.M.D., S.D.G., and K.A.Z. provided substantive support and helped with the project administration. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by the Russian State-funded project for the FBSI “Research Institute of Pulmonology” of the FMBA of Russia (grant number 388-03-2024-136) and supported by the Russian State-funded project for ICBFM SB RAS (grant number 125012900932-4).

Institutional Review Board Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare that they have no competing interest.

Abbreviations

ADAdenocarcinoma
BALBronchoalveolar lavage
Cf-miRNACell-free microRNA
LCLung Cancer
miRNAMicroRNA
NSCLCNon-Small-Cell Lung Cancer
RT-qPCRQuantitative real-time Polymerase Chain Reaction
SSCSquamous Cell Carcinoma

References

  1. Siegel, R.L.; Kratzer, T.B.; Giaquinto, A.N.; Sung, H.; Jemal, A. Cancer statistics, 2025. CA Cancer J. Clin. 2025, 75, 10–45. [Google Scholar] [CrossRef]
  2. Verbraecken, J.; Van de Heyning, P.; De Backer, W.; Van Gaal, L. Body surface area in normal-weight, overweight, and obese adults. A comparison study. Metabolism 2006, 55, 515–524. [Google Scholar] [CrossRef]
  3. Ozlu, T.; Bulbul, Y. Smoking and lung cancer. Tuberk. Toraks 2005, 53, 200–209. [Google Scholar] [PubMed]
  4. Possenti, I.; Romelli, M.; Carreras, G.; Biffi, A.; Bagnardi, V.; Specchia, C.; Gallus, S.; Lugo, A. Association between second-hand smoke exposure and lung cancer risk in never-smokers: A systematic review and meta-analysis. Eur. Respir. Rev. 2024, 33, 240077. [Google Scholar] [CrossRef]
  5. Saller, J.J.; Boyle, T.A. Molecular Pathology of Lung Cancer. Cold Spring Harb. Perspect. Med. 2022, 12, a037812. [Google Scholar] [CrossRef]
  6. Guo, H.; Li, H.; Zhu, L.; Feng, J.; Huang, X.; Baak, J.P.A. “How Long Have I Got?” in Stage IV NSCLC Patients with at Least 3 Months Up to 10 Years Survival, Accuracy of Long-, Intermediate-, and Short-Term Survival Prediction Is Not Good Enough to Answer This Question. Front. Oncol. 2021, 11, 761042. [Google Scholar] [CrossRef] [PubMed]
  7. Jovanoski, N.; Bowes, K.; Brown, A.; Belleli, R.; Di Maio, D.; Chadda, S.; Abogunrin, S. Survival and quality-of-life outcomes in early-stage NSCLC patients: A literature review of real-world evidence. Lung Cancer Manag. 2023, 12, LMT60. [Google Scholar] [CrossRef] [PubMed]
  8. Lee, D.H.; Kim, J.B.; Keum, D.Y.; Hwang, I.; Park, C.K. Long term survival of patients with unsuspected n2 disease in non-small cell lung cancer. Korean J. Thorac. Cardiovasc. Surg. 2013, 46, 49–55. [Google Scholar] [CrossRef]
  9. Park, H.J.; Lee, S.H.; Chang, Y.S. Recent advances in diagnostic technologies in lung cancer. Korean J. Intern. Med. 2020, 35, 257–268. [Google Scholar] [CrossRef]
  10. Jeon, H.; Wang, S.; Song, J.; Gill, H.; Cheng, H. Update 2025: Management of Non-Small-Cell Lung Cancer. Lung 2025, 203, 53. [Google Scholar] [CrossRef]
  11. National Lung Screening Trial Research Team; Aberle, D.R.; Adams, A.M.; Berg, C.D.; Black, W.C.; Clapp, J.D.; Fagerstrom, R.M.; Gareen, I.F.; Gatsonis, C.; Marcus, P.M.; et al. Reduced lung-cancer mortality with low-dose computed tomographic screening. N. Engl. J. Med. 2011, 365, 395–409. [Google Scholar] [CrossRef]
  12. Force, U.S.P.S.T.; Krist, A.H.; Davidson, K.W.; Mangione, C.M.; Barry, M.J.; Cabana, M.; Caughey, A.B.; Davis, E.M.; Donahue, K.E.; Doubeni, C.A.; et al. Screening for Lung Cancer: US Preventive Services Task Force Recommendation Statement. JAMA 2021, 325, 962–970. [Google Scholar] [CrossRef]
  13. Poh, K.C.; Ren, T.M.; Ling, G.L.; Goh, J.S.Y.; Rose, S.; Wong, A.; Mehta, S.S.; Goh, A.; Chong, P.Y.; Cheng, S.W.; et al. Development of a miRNA-Based Model for Lung Cancer Detection. Cancers 2025, 17, 942. [Google Scholar] [CrossRef]
  14. Wadowska, K.; Bil-Lula, I.; Trembecki, L.; Sliwinska-Mosson, M. Genetic Markers in Lung Cancer Diagnosis: A Review. Int. J. Mol. Sci. 2020, 21, 4569. [Google Scholar] [CrossRef]
  15. Zou, Y.; Wang, Y.; Jiang, Z.; Zhou, Y.; Chen, Y.; Hu, Y.; Jiang, G.; Xie, D. Breath profile as composite biomarkers for lung cancer diagnosis. Lung Cancer 2021, 154, 206–213. [Google Scholar] [CrossRef]
  16. Lv, W.; Shi, W.; Zhang, Z.; Ru, L.; Feng, W.; Tang, H.; Wang, X. Identification of volatile biomarkers for lung cancer from different histological sources: A comprehensive study. Anal. Biochem. 2024, 690, 115527. [Google Scholar] [CrossRef]
  17. Janssens, E.; van Meerbeeck, J.P.; Lamote, K. Volatile organic compounds in human matrices as lung cancer biomarkers: A systematic review. Crit. Rev. Oncol. Hematol. 2020, 153, 103037. [Google Scholar] [CrossRef]
  18. Bazan Russo, T.D.; Pepe, F.; Gristina, V.; Gottardo, A.; Russo, G.; Scimone, C.; Palumbo, L.; Busuito, G.; Incorvaia, L.; Guerry, J.A.; et al. Recent advances in liquid biopsy for precision oncology: Emerging biomarkers and clinical applications in lung cancer. Future Oncol. 2025, 21, 2803–2821. [Google Scholar] [CrossRef] [PubMed]
  19. Farooq, M.; Herman, J.G. Noninvasive Diagnostics for Early Detection of Lung Cancer: Challenges and Potential with a Focus on Changes in DNA Methylation. Cancer Epidemiol. Biomark. Prev. 2020, 29, 2416–2422. [Google Scholar] [CrossRef] [PubMed]
  20. Chow, Y.P.; Zainul Abidin, N.; Kow, K.S.; Tho, L.M.; Wong, C.L. Analytical and clinical validation of a custom 15-gene next-generation sequencing panel for the evaluation of circulating tumor DNA mutations in patients with advanced non-small-cell lung cancer. PLoS ONE 2022, 17, e0276161. [Google Scholar] [CrossRef] [PubMed]
  21. Kumbrink, J.; Demes, M.C.; Jeroch, J.; Brauninger, A.; Hartung, K.; Gerstenmaier, U.; Marienfeld, R.; Hillmer, A.; Bohn, N.; Lehning, C.; et al. Development, testing and validation of a targeted NGS-panel for the detection of actionable mutations in lung cancer (NSCLC) using anchored multiplex PCR technology in a multicentric setting. Pathol. Oncol. Res. 2024, 30, 1611590. [Google Scholar] [CrossRef] [PubMed]
  22. Chen, X.; Gole, J.; Gore, A.; He, Q.; Lu, M.; Min, J.; Yuan, Z.; Yang, X.; Jiang, Y.; Zhang, T.; et al. Non-invasive early detection of cancer four years before conventional diagnosis using a blood test. Nat. Commun. 2020, 11, 3475. [Google Scholar] [CrossRef] [PubMed]
  23. Bao, Y.; Zhang, D.; Guo, H.; Ma, W. Beyond blood: Advancing the frontiers of liquid biopsy in oncology and personalized medicine. Cancer Sci. 2024, 115, 1060–1072. [Google Scholar] [CrossRef]
  24. Bryzgunova, O.; Konoshenko, M.; Zaporozhchenko, I.; Yakovlev, A.; Laktionov, P. Isolation of Cell-Free miRNA from Biological Fluids: Influencing Factors and Methods. Diagnostics 2021, 11, 865. [Google Scholar] [CrossRef]
  25. Liao, J.; Shen, J.; Leng, Q.; Qin, M.; Zhan, M.; Jiang, F. MicroRNA-based biomarkers for diagnosis of non-small cell lung cancer (NSCLC). Thorac. Cancer 2020, 11, 762–768. [Google Scholar] [CrossRef]
  26. Li, W.; Wang, Y.; Zhang, Q.; Tang, L.; Liu, X.; Dai, Y.; Xiao, L.; Huang, S.; Chen, L.; Guo, Z.; et al. MicroRNA-486 as a Biomarker for Early Diagnosis and Recurrence of Non-Small Cell Lung Cancer. PLoS ONE 2015, 10, e0134220, Correction in PLoS ONE 2016, 11, e0148589. https://doi.org/10.1371/journal.pone.0148589. [Google Scholar] [CrossRef]
  27. Yi, M.; Liao, Z.; Deng, L.; Xu, L.; Tan, Y.; Liu, K.; Chen, Z.; Zhang, Y. High diagnostic value of miRNAs for NSCLC: Quantitative analysis for both single and combined miRNAs in lung cancer. Ann. Med. 2021, 53, 2178–2193. [Google Scholar] [CrossRef] [PubMed]
  28. Jin, X.; Guan, Y.; Zhang, Z.; Wang, H. Microarray data analysis on gene and miRNA expression to identify biomarkers in non-small cell lung cancer. BMC Cancer 2020, 20, 329. [Google Scholar] [CrossRef]
  29. Schwarzenbach, H.; da Silva, A.M.; Calin, G.; Pantel, K. Data Normalization Strategies for MicroRNA Quantification. Clin. Chem. 2015, 61, 1333–1342. [Google Scholar] [CrossRef]
  30. Saito, M.; Schetter, A.J.; Mollerup, S.; Kohno, T.; Skaug, V.; Bowman, E.D.; Mathe, E.A.; Takenoshita, S.; Yokota, J.; Haugen, A.; et al. The association of microRNA expression with prognosis and progression in early-stage, non-small cell lung adenocarcinoma: A retrospective analysis of three cohorts. Clin. Cancer Res. 2011, 17, 1875–1882. [Google Scholar] [CrossRef]
  31. Wei, J.; Gao, W.; Zhu, C.J.; Liu, Y.Q.; Mei, Z.; Cheng, T.; Shu, Y.Q. Identification of plasma microRNA-21 as a biomarker for early detection and chemosensitivity of non-small cell lung cancer. Chin. J. Cancer 2011, 30, 407–414. [Google Scholar] [CrossRef]
  32. Qi, Z.; Yang, D.Y.; Cao, J. Increased micro-RNA 17, 21, and 192 gene expressions improve early diagnosis in non-small cell lung cancer. Med. Oncol. 2014, 31, 195. [Google Scholar] [CrossRef]
  33. Geng, Q.; Fan, T.; Zhang, B.; Wang, W.; Xu, Y.; Hu, H. Five microRNAs in plasma as novel biomarkers for screening of early-stage non-small cell lung cancer. Respir. Res. 2014, 15, 149. [Google Scholar] [CrossRef]
  34. Cho, W.C.; Wong, C.F.; Li, K.P.; Fong, A.H.; Fung, K.Y.; Au, J.S. miR-145 as a Potential Biomarker and Therapeutic Target in Patients with Non-Small Cell Lung Cancer. Int. J. Mol. Sci. 2023, 24, 10022. [Google Scholar] [CrossRef] [PubMed]
  35. Campayo, M.; Navarro, A.; Vinolas, N.; Diaz, T.; Tejero, R.; Gimferrer, J.M.; Molins, L.; Cabanas, M.L.; Ramirez, J.; Monzo, M.; et al. Low miR-145 and high miR-367 are associated with unfavourable prognosis in resected nonsmall cell lung cancer. Eur. Respir. J. 2013, 41, 1172–1178. [Google Scholar] [CrossRef] [PubMed]
  36. Aiso, T.; Ohtsuka, K.; Ueda, M.; Karita, S.; Yokoyama, T.; Takata, S.; Matsuki, N.; Kondo, H.; Takizawa, H.; Okada, A.A.; et al. Serum levels of candidate microRNA diagnostic markers differ among the stages of non-small-cell lung cancer. Oncol. Lett. 2018, 16, 6643–6651. [Google Scholar] [CrossRef] [PubMed]
  37. Mataki, H.; Seki, N.; Mizuno, K.; Nohata, N.; Kamikawaji, K.; Kumamoto, T.; Koshizuka, K.; Goto, Y.; Inoue, H. Dual-strand tumor-suppressor microRNA-145 (miR-145-5p and miR-145-3p) coordinately targeted MTDH in lung squamous cell carcinoma. Oncotarget 2016, 7, 72084–72098. [Google Scholar] [CrossRef]
  38. Shen, H.; Shen, J.; Wang, L.; Shi, Z.; Wang, M.; Jiang, B.H.; Shu, Y. Low miR-145 expression level is associated with poor pathological differentiation and poor prognosis in non-small cell lung cancer. Biomed. Pharmacother. 2015, 69, 301–305. [Google Scholar] [CrossRef]
  39. Grimolizzi, F.; Monaco, F.; Leoni, F.; Bracci, M.; Staffolani, S.; Bersaglieri, C.; Gaetani, S.; Valentino, M.; Amati, M.; Rubini, C.; et al. Exosomal miR-126 as a circulating biomarker in non-small-cell lung cancer regulating cancer progression. Sci. Rep. 2017, 7, 15277. [Google Scholar] [CrossRef]
  40. Ulivi, P.; Petracci, E.; Marisi, G.; Baglivo, S.; Chiari, R.; Billi, M.; Canale, M.; Pasini, L.; Racanicchi, S.; Vagheggini, A.; et al. Prognostic Role of Circulating miRNAs in Early-Stage Non-Small Cell Lung Cancer. J. Clin. Med. 2019, 8, 131. [Google Scholar] [CrossRef]
  41. Zhang, H.; Mao, F.; Shen, T.; Luo, Q.; Ding, Z.; Qian, L.; Huang, J. Plasma miR-145, miR-20a, miR-21 and miR-223 as novel biomarkers for screening early-stage non-small cell lung cancer. Oncol. Lett. 2017, 13, 669–676. [Google Scholar] [CrossRef]
  42. Azimi, S.A.; Sadegh Nia, H.R.; Bahrami, N.; Dargahi, H.; Jamaati, H.; Pasdar, A.; Kazempour Dizaji, M.; Shirian, S.; Zerehsaz, B.; Mohamadnia, A. Expression of miR-155 and CEA and VEGF Proteins as Diagnostic Markers in Early Stages of Non-Small Cell Lung Cancer in Peripheral Blood. Tanaffos 2024, 23, 58–64. [Google Scholar]
  43. Xue, X.; Liu, Y.; Wang, Y.; Meng, M.; Wang, K.; Zang, X.; Zhao, S.; Sun, X.; Cui, L.; Pan, L.; et al. MiR-21 and MiR-155 promote non-small cell lung cancer progression by downregulating SOCS1, SOCS6, and PTEN. Oncotarget 2016, 7, 84508–84519. [Google Scholar] [CrossRef]
  44. Zhang, W.T.; Zhang, G.X.; Gao, S.S. The Potential Diagnostic Accuracy of Let-7 Family for Cancer: A Meta-Analysis. Technol. Cancer Res. Treat. 2021, 20, 15330338211033061. [Google Scholar] [CrossRef]
  45. Tulinsky, L.; Dzian, A.; Matakova, T.; Ihnat, P. Overexpression of the miR-143/145 and reduced expression of the let-7 and miR-126 for early lung cancer diagnosis. J. Appl. Biomed. 2022, 20, 1–6. [Google Scholar] [CrossRef]
  46. Heegaard, N.H.; Schetter, A.J.; Welsh, J.A.; Yoneda, M.; Bowman, E.D.; Harris, C.C. Circulating micro-RNA expression profiles in early stage nonsmall cell lung cancer. Int. J. Cancer 2012, 130, 1378–1386. [Google Scholar] [CrossRef] [PubMed]
  47. Tejero, R.; Navarro, A.; Campayo, M.; Vinolas, N.; Marrades, R.M.; Cordeiro, A.; Ruiz-Martinez, M.; Santasusagna, S.; Molins, L.; Ramirez, J.; et al. miR-141 and miR-200c as markers of overall survival in early stage non-small cell lung cancer adenocarcinoma. PLoS ONE 2014, 9, e101899. [Google Scholar] [CrossRef]
  48. Zeybek, A.; Oz, N.; Kalemci, S.; Edgunlu, T.; Kiziltug, M.T.; Tosun, K.; Tunc, M.; Tekin, L.; Erdal, M.E. Diagnostic Value of MiR-125b as a Potential Biomarker for Stage I Lung Adenocarcinoma. Curr. Mol. Med. 2019, 19, 216–227. [Google Scholar] [CrossRef] [PubMed]
  49. Chen, G.; Umelo, I.A.; Lv, S.; Teugels, E.; Fostier, K.; Kronenberger, P.; Dewaele, A.; Sadones, J.; Geers, C.; De Greve, J. miR-146a inhibits cell growth, cell migration and induces apoptosis in non-small cell lung cancer cells. PLoS ONE 2013, 8, e60317. [Google Scholar] [CrossRef]
  50. Capodanno, A.; Boldrini, L.; Proietti, A.; Ali, G.; Pelliccioni, S.; Niccoli, C.; D’Incecco, A.; Cappuzzo, F.; Chella, A.; Lucchi, M.; et al. Let-7g and miR-21 expression in non-small cell lung cancer: Correlation with clinicopathological and molecular features. Int. J. Oncol. 2013, 43, 765–774. [Google Scholar] [CrossRef] [PubMed]
  51. Meng, F.; Henson, R.; Wehbe-Janek, H.; Ghoshal, K.; Jacob, S.T.; Patel, T. MicroRNA-21 regulates expression of the PTEN tumor suppressor gene in human hepatocellular cancer. Gastroenterology 2007, 133, 647–658. [Google Scholar] [CrossRef]
  52. Cho, W.C.; Chow, A.S.; Au, J.S. MiR-145 inhibits cell proliferation of human lung adenocarcinoma by targeting EGFR and NUDT1. RNA Biol. 2011, 8, 125–131. [Google Scholar] [CrossRef]
  53. Chen, Z.; Zeng, H.; Guo, Y.; Liu, P.; Pan, H.; Deng, A.; Hu, J. miRNA-145 inhibits non-small cell lung cancer cell proliferation by targeting c-Myc. J. Exp. Clin. Cancer Res. 2010, 29, 151. [Google Scholar] [CrossRef]
  54. Zhang, G.; Lei, S.; Zhang, K. The effect of microRNA-145 on proliferation and apoptosis of cutaneous squamous cell carcinoma cells. Discov. Oncol. 2025, 16, 1201. [Google Scholar] [CrossRef]
  55. Pei, X.; Chen, S.W.; Long, X.; Zhu, S.Q.; Qiu, B.Q.; Lin, K.; Lu, F.; Xu, J.J.; Zhang, P.F.; Wu, Y.B. circMET promotes NSCLC cell proliferation, metastasis, and immune evasion by regulating the miR-145-5p/CXCL3 axis. Aging 2020, 12, 13038–13058. [Google Scholar] [CrossRef]
  56. Alinejad, T.; Hao, Z.; Zhou, W.; Zareh, D.; Farajtabrizi, E.; Mossahebi-Mohammadi, M.; Chen, C.S. Knockdown of long noncoding RNA MALAT1 enhances the anti-cancer effects of polysaccharides Glehnia littoralis in lung cancer cells possibly via the regulation of miR-145/SOX9 axis. Int. Immunopharmacol. 2025, 164, 115323. [Google Scholar] [CrossRef]
  57. Chen, Q.; Chen, S.; Zhao, J.; Zhou, Y.; Xu, L. MicroRNA-126: A new and promising player in lung cancer. Oncol. Lett. 2021, 21, 35. [Google Scholar] [CrossRef] [PubMed]
  58. Cao, D.; Mikosz, A.M.; Ringsby, A.J.; Anderson, K.C.; Beatman, E.L.; Koike, K.; Petrache, I. MicroRNA-126-3p Inhibits Angiogenic Function of Human Lung Microvascular Endothelial Cells via LAT1 (L-Type Amino Acid Transporter 1)-Mediated mTOR (Mammalian Target of Rapamycin) Signaling. Arter. Thromb. Vasc. Biol. 2020, 40, 1195–1206. [Google Scholar] [CrossRef] [PubMed]
  59. Chen, P.; Gu, Y.Y.; Ma, F.C.; He, R.Q.; Li, Z.Y.; Zhai, G.Q.; Lin, X.; Hu, X.H.; Pan, L.J.; Chen, G. Expression levels and co-targets of miRNA-126-3p and miRNA-126-5p in lung adenocarcinoma tissues: Alphan exploration with RT-qPCR, microarray and bioinformatic analyses. Oncol. Rep. 2019, 41, 939–953. [Google Scholar] [CrossRef]
  60. Lin, J.; Chen, Y.; Liu, L.; Shen, A.; Zheng, W. MicroRNA-155-5p suppresses the migration and invasion of lung adenocarcinoma A549 cells by targeting Smad2. Oncol. Lett. 2018, 16, 2444–2452. [Google Scholar] [CrossRef] [PubMed]
  61. Park, K.S.; Moon, Y.W.; Raffeld, M.; Lee, D.H.; Wang, Y.; Giaccone, G. High cripto-1 and low miR-205 expression levels as prognostic markers in early stage non-small cell lung cancer. Lung Cancer 2018, 116, 38–45. [Google Scholar] [CrossRef]
  62. Lei, L.; Huang, Y.; Gong, W. miR-205 promotes the growth, metastasis and chemoresistance of NSCLC cells by targeting PTEN. Oncol. Rep. 2013, 30, 2897–2902. [Google Scholar] [CrossRef] [PubMed]
  63. Xu, L.B.; Xiong, J.; Zhang, Y.H.; Dai, Y.; Ren, X.P.; Ren, Y.J.; Han, D.; Wei, S.H.; Qi, M. miR-205-3p promotes lung cancer progression by targeting APBB2. Mol. Med. Rep. 2021, 24, 588. [Google Scholar] [CrossRef]
  64. Zhao, Y.L.; Zhang, J.X.; Yang, J.J.; Wei, Y.B.; Peng, J.F.; Fu, C.J.; Huang, M.H.; Wang, R.; Wang, P.Y.; Sun, G.B.; et al. MiR-205-5p promotes lung cancer progression and is valuable for the diagnosis of lung cancer. Thorac. Cancer 2022, 13, 832–843. [Google Scholar] [CrossRef] [PubMed]
  65. Zhang, L.; Lin, J.; Ye, Y.; Oba, T.; Gentile, E.; Lian, J.; Wang, J.; Zhao, Y.; Gu, J.; Wistuba, I.I.; et al. Serum MicroRNA-150 Predicts Prognosis for Early-Stage Non-Small Cell Lung Cancer and Promotes Tumor Cell Proliferation by Targeting Tumor Suppressor Gene SRCIN1. Clin. Pharmacol. Ther. 2018, 103, 1061–1073. [Google Scholar] [CrossRef]
  66. Qin, A.; Chen, H.; Xu, F.; Li, W.; Guo, S.; Zhang, G.; Zhang, A.; Zheng, A.; Tian, F.; Zheng, Q. MiR-150 deletion promotes lung tumor growth by upregulating P-STAT3 and ROS in MDSCs. Sci. Rep. 2025, 15, 12988. [Google Scholar] [CrossRef]
  67. Li, H.; Liu, J.; Cao, W.; Xiao, X.; Liang, L.; Liu-Smith, F.; Wang, W.; Liu, H.; Zhou, P.; Ouyang, R.; et al. C-myc/miR-150/EPG5 axis mediated dysfunction of autophagy promotes development of non-small cell lung cancer. Theranostics 2019, 9, 5134–5148. [Google Scholar] [CrossRef] [PubMed]
  68. Li, H.; Ouyang, R.; Wang, Z.; Zhou, W.; Chen, H.; Jiang, Y.; Zhang, Y.; Li, H.; Liao, M.; Wang, W.; et al. MiR-150 promotes cellular metastasis in non-small cell lung cancer by targeting FOXO4. Sci. Rep. 2016, 6, 39001. [Google Scholar] [CrossRef]
  69. Yin, Q.W.; Sun, X.F.; Yang, G.T.; Li, X.B.; Wu, M.S.; Zhao, J. Increased expression of microRNA-150 is associated with poor prognosis in non-small cell lung cancer. Int. J. Clin. Exp. Pathol. 2015, 8, 842–846. [Google Scholar]
  70. Zhao, Y. The diagnostic and prognostic role of circulating miR-141 expression in non-small-cell lung cancer patients. Int. J. Clin. Exp. Pathol. 2018, 11, 2597–2604. [Google Scholar]
  71. Zhang, X.; Li, P.; Rong, M.; He, R.; Hou, X.; Xie, Y.; Chen, G. MicroRNA-141 is a biomarker for progression of squamous cell carcinoma and adenocarcinoma of the lung: Clinical analysis of 125 patients. Tohoku J. Exp. Med. 2015, 235, 161–169. [Google Scholar] [CrossRef]
  72. Wang, C.; Zhang, W.; Zhang, L.; Chen, X.; Liu, F.; Zhang, J.; Guan, S.; Sun, Y.; Chen, P.; Wang, D.; et al. miR-146a-5p mediates epithelial-mesenchymal transition of oesophageal squamous cell carcinoma via targeting Notch2. Br. J. Cancer 2016, 115, 1548–1554, Correction in Br. J. Cancer 2018, 118, e12. https://doi.org/10.1038/bjc.2017.471. [Google Scholar] [CrossRef] [PubMed]
  73. Liu, X.; Liu, B.; Li, R.; Wang, F.; Wang, N.; Zhang, M.; Bai, Y.; Wu, J.; Liu, L.; Han, D.; et al. miR-146a-5p Plays an Oncogenic Role in NSCLC via Suppression of TRAF6. Front. Cell Dev. Biol. 2020, 8, 847. [Google Scholar] [CrossRef] [PubMed]
  74. D’Antona, P.; Cattoni, M.; Dominioni, L.; Poli, A.; Moretti, F.; Cinquetti, R.; Gini, E.; Daffre, E.; Noonan, D.M.; Imperatori, A.; et al. Serum miR-223: A Validated Biomarker for Detection of Early-Stage Non-Small Cell Lung Cancer. Cancer Epidemiol. Biomark. Prev. 2019, 28, 1926–1933. [Google Scholar] [CrossRef] [PubMed]
  75. Ma, H.P.; Kong, W.X.; Li, X.Y.; Li, W.; Zhang, Y.; Wu, Y. miRNA-223 is an anticancer gene in human non-small cell lung cancer through the PI3K/AKT pathway by targeting EGFR. Oncol. Rep. 2019, 41, 1549–1559. [Google Scholar] [CrossRef] [PubMed]
  76. Nian, W.; Ao, X.; Wu, Y.; Huang, Y.; Shao, J.; Wang, Y.; Chen, Z.; Chen, F.; Wang, D. miR-223 functions as a potent tumor suppressor of the Lewis lung carcinoma cell line by targeting insulin-like growth factor-1 receptor and cyclin-dependent kinase 2. Oncol. Lett. 2013, 6, 359–366. [Google Scholar] [CrossRef]
  77. Zhu, S.; Kong, X.; Song, M.; Chi, M.; Liu, Y.; Zhang, P.; Zhang, Q.; Shang, P.; Feng, F. MiR-223-3p attenuates the migration and invasion of NSCLC cells by regulating NLRP3. Front. Oncol. 2022, 12, 985962. [Google Scholar] [CrossRef]
  78. Dou, L.; Han, K.; Xiao, M.; Lv, F. miR-223-5p Suppresses Tumor Growth and Metastasis in Non-Small Cell Lung Cancer by Targeting E2F8. Oncol. Res. 2019, 27, 261–268. [Google Scholar] [CrossRef]
  79. Nadal, E.; Chen, G.; Gallegos, M.; Lin, L.; Ferrer-Torres, D.; Truini, A.; Wang, Z.; Lin, J.; Reddy, R.M.; Llatjos, R.; et al. Epigenetic inactivation of microRNA-34b/c predicts poor disease-free survival in early-stage lung adenocarcinoma. Clin. Cancer Res. 2013, 19, 6842–6852. [Google Scholar] [CrossRef]
  80. Kim, J.S.; Kim, E.J.; Lee, S.; Tan, X.; Liu, X.; Park, S.; Kang, K.; Yoon, J.S.; Ko, Y.H.; Kurie, J.M.; et al. MiR-34a and miR-34b/c have distinct effects on the suppression of lung adenocarcinomas. Exp. Mol. Med. 2019, 51, 1–10. [Google Scholar] [CrossRef]
  81. Huang, G.; Liu, Y.; Li, L.; Li, B.; Jiang, T.; Cao, Y.; Yang, X.; Liu, X.; Qu, H.; Li, S.; et al. Integration analysis of microRNAs as potential biomarkers in early-stage lung adenocarcinoma: The diagnostic and therapeutic significance of miR-183-3p. Front. Oncol. 2024, 14, 1508715. [Google Scholar] [CrossRef]
  82. Zhang, T.; Li, W.; Gu, M.; Wang, Z.; Zhou, S.; Hao, X.; Li, W.; Xu, S. Clinical Significance of miR-183-3p and miR-182-5p in NSCLC and Their Correlation. Cancer Manag. Res. 2021, 13, 3539–3550. [Google Scholar] [CrossRef]
  83. Chen, J.; Zhou, D.; Liao, H.; Li, Y. miR-183-5p regulates ECM and EMT to promote non-small cell lung cancer progression by targeting LOXL4. J. Thorac. Dis. 2023, 15, 1734–1748. [Google Scholar] [CrossRef]
  84. Zhang, L.; Quan, H.; Wang, S.; Li, X.; Che, X. MiR-183 promotes growth of non-small cell lung cancer cells through FoxO1 inhibition. Tumour Biol. 2015, 36, 8121–8126. [Google Scholar] [CrossRef]
  85. Huang, D.; Qu, D. Early diagnostic and prognostic value of serum exosomal miR-1246 in non-small cell lung cancer. Int. J. Clin. Exp. Pathol. 2020, 13, 1601–1607. [Google Scholar] [PubMed]
  86. Liu, Z.; Zhao, W.; Yang, R. MiR-1246 is responsible for lung cancer cells-derived exosomes-mediated promoting effects on lung cancer stemness via targeting TRIM17. Environ. Toxicol. 2022, 37, 2651–2659. [Google Scholar] [CrossRef] [PubMed]
  87. Zeng, M.; Li, F.; Wang, L.; Chen, C.; Huang, X.; Wu, X.; She, W.; Zhou, L.; Tao, Z. Downregulated cytoplasmic polyadenylation element-binding protein-4 is associated with the carcinogenesis of head and neck squamous cell carcinoma. Oncol. Lett. 2018, 15, 3226–3232. [Google Scholar] [CrossRef] [PubMed]
  88. Yang, F.; Xiong, H.; Duan, L.; Li, Q.; Li, X.; Zhou, Y. MiR-1246 Promotes Metastasis and Invasion of A549 cells by Targeting GSK-3beta-Mediated Wnt/beta-Catenin Pathway. Cancer Res. Treat. 2019, 51, 1420–1429. [Google Scholar] [CrossRef]
  89. Yuan, D.; Xu, J.; Wang, J.; Pan, Y.; Fu, J.; Bai, Y.; Zhang, J.; Shao, C. Extracellular miR-1246 promotes lung cancer cell proliferation and enhances radioresistance by directly targeting DR5. Oncotarget 2016, 7, 32707–32722. [Google Scholar] [CrossRef]
  90. Ulivi, P.; Foschi, G.; Mengozzi, M.; Scarpi, E.; Silvestrini, R.; Amadori, D.; Zoli, W. Peripheral blood miR-328 expression as a potential biomarker for the early diagnosis of NSCLC. Int. J. Mol. Sci. 2013, 14, 10332–10342. [Google Scholar] [CrossRef]
  91. Ji, Y.; You, Y.; Wu, Y.; Wang, M.; He, Q.; Zhou, X.; Chen, L.; Sun, X.; Liu, Y.; Fu, X.; et al. Overexpression of miR-328-5p influences cell growth and migration to promote NSCLC progression by targeting LOXL4. Ann. Transl. Med. 2022, 10, 301. [Google Scholar] [CrossRef]
  92. Ma, W.; Ma, C.N.; Zhou, N.N.; Li, X.D.; Zhang, Y.J. Up-regulation of miR-328-3p sensitizes non-small cell lung cancer to radiotherapy. Sci. Rep. 2016, 6, 31651. [Google Scholar] [CrossRef]
  93. Liu, L.; Zhang, F.; Niu, D.; Guo, X.; Lei, T.; Liu, H. Diagnostic value of microRNA-200 expression in peripheral blood-derived extracellular vesicles in early-stage non-small cell lung cancer. Clin. Exp. Med. 2024, 24, 214. [Google Scholar] [CrossRef] [PubMed]
  94. Huang, Y.; Bao, T.; Li, Z.; Ji, G.; Zhang, L. Function of miR-200a in proliferation and apoptosis of non-small cell lung cancer cells. Oncol. Lett. 2020, 20, 1256–1262. [Google Scholar] [CrossRef] [PubMed]
  95. Yu, J.; He, X.; Fang, C.; Wu, H.; Hu, L.; Xue, Y. MicroRNA-200a-3p and GATA6 are abnormally expressed in patients with non-small cell lung cancer and exhibit high clinical diagnostic efficacy. Exp. Ther. Med. 2022, 23, 281. [Google Scholar] [CrossRef]
  96. Li, W.; Jia, M.X.; Deng, J.; Wang, J.H.; Lin, Q.L.; Tang, J.X.; Zeng, X.X.; Cai, F.; Ma, L.; Su, W.; et al. Down-regulation of microRNA-200b is a potential prognostic marker of lung cancer in southern-central Chinese population. Saudi J. Biol. Sci. 2019, 26, 173–177. [Google Scholar] [CrossRef] [PubMed]
  97. Liu, K.; Zhang, W.; Tan, J.; Ma, J.; Zhao, J. MiR-200b-3p Functions as an Oncogene by Targeting ABCA1 in Lung Adenocarcinoma. Technol. Cancer Res. Treat. 2019, 18, 1533033819892590. [Google Scholar] [CrossRef]
  98. Chen, S.; Tu, Y.; Yuan, H.; Shi, Z.; Guo, Y.; Gong, W.; Tu, S. Regulatory functions of miR-200b-3p in tumor development (Review). Oncol. Rep. 2022, 47, 96. [Google Scholar] [CrossRef]
  99. Guo, H.; Zhang, N.; Huang, T.; Shen, N. MicroRNA-200c in Cancer Generation, Invasion, and Metastasis. Int. J. Mol. Sci. 2025, 26, 710. [Google Scholar] [CrossRef]
  100. Zhang, Q.; Pan, J.; Xiong, D.; Zheng, J.; McPherson, K.N.; Lee, S.; Huang, M.; Xu, Y.; Chen, S.H.; Wang, Y.; et al. Aerosolized miR-138-5p and miR-200c targets PD-L1 for lung cancer prevention. Front. Immunol. 2023, 14, 1166951. [Google Scholar] [CrossRef]
  101. Shen, Y.; Zhu, Y.; Rong, F. miR-200c-3p regulates the proliferation and apoptosis of human trabecular meshwork cells by targeting PTEN. Mol. Med. Rep. 2020, 22, 1605–1612. [Google Scholar] [CrossRef]
  102. Liu, Y.; Zhang, Y.; Li, Q.; Xu, R.; Huang, N. MiR-200c-3p and miR-485-5p overexpression elevates cisplatin sensitivity and suppresses the malignant phenotypes of non-small cell lung cancer cells through targeting RRM2. Thorac. Cancer 2022, 13, 1974–1985. [Google Scholar] [CrossRef] [PubMed]
  103. Pop-Bica, C.; Pintea, S.; Magdo, L.; Cojocneanu, R.; Gulei, D.; Ferracin, M.; Berindan-Neagoe, I. The Clinical Utility of miR-21 and let-7 in Non-small Cell Lung Cancer (NSCLC). A Systematic Review and Meta-Analysis. Front. Oncol. 2020, 10, 516850. [Google Scholar] [CrossRef] [PubMed]
  104. Li, Y.; Dong, R.; Lu, M.; Cheng, C.; Feng, Z.; Zhao, R.; Liang, J.; Han, J.; Jiang, J.; Xu-Welliver, M.; et al. Let-7b-3p inhibits tumor growth and metastasis by targeting the BRF2-mediated MAPK/ERK pathway in human lung adenocarcinoma. Transl. Lung Cancer Res. 2021, 10, 1841–1856. [Google Scholar] [CrossRef]
  105. Zhang, Q.; Pan, J.; Xiong, D.; Wang, Y.; Miller, M.S.; Sei, S.; Shoemaker, R.H.; Izzotti, A.; You, M. Pulmonary Aerosol Delivery of Let-7b microRNA Confers a Striking Inhibitory Effect on Lung Carcinogenesis through Targeting the Tumor Immune Microenvironment. Adv. Sci. 2021, 8, e2100629. [Google Scholar] [CrossRef] [PubMed]
  106. Zhu, S.; Chen, J.; Yu, L.; Li, J.; You, S.; Zheng, Y.; Zhuang, W.; Qiu, B.; Huang, Y. Detection of fucosylated extracellular vesicles miR-4732-5p related to diagnosis of early lung adenocarcinoma by the electrochemical biosensor. Sci. Rep. 2024, 14, 11217. [Google Scholar] [CrossRef]
  107. Tang, X.; Liu, S.; Cui, Y.; Zhao, Y. MicroRNA-4732 is downregulated in non-small cell lung cancer and inhibits tumor cell proliferation, migration, and invasion. Respir. Med. Res. 2021, 80, 100865. [Google Scholar] [CrossRef]
  108. Hu, Y.; Bai, J.; Zhou, D.; Zhang, L.; Chen, X.; Chen, L.; Liu, Y.; Zhang, B.; Li, H.; Yin, C. The miR-4732-5p/XPR1 axis suppresses the invasion, metastasis, and epithelial-mesenchymal transition of lung adenocarcinoma via the PI3K/Akt/GSK3beta/Snail pathway. Mol. Omics 2022, 18, 417–429. [Google Scholar] [CrossRef]
  109. Vosa, U.; Vooder, T.; Kolde, R.; Fischer, K.; Valk, K.; Tonisson, N.; Roosipuu, R.; Vilo, J.; Metspalu, A.; Annilo, T. Identification of miR-374a as a prognostic marker for survival in patients with early-stage nonsmall cell lung cancer. Genes Chromosomes Cancer 2011, 50, 812–822. [Google Scholar] [CrossRef]
  110. Jin, J.; Cui, Y.; Niu, H.; Lin, Y.; Wu, X.; Qi, X.; Bai, K.; Zhang, Y.; Wang, Y.; Bu, H. NSCLC Extracellular Vesicles Containing miR-374a-5p Promote Leptomeningeal Metastasis by Influencing Blood–Brain Barrier Permeability. Mol. Cancer Res. 2024, 22, 699–710. [Google Scholar] [CrossRef]
  111. Guo, Q.; Wang, H.; Xu, Y.; Wang, M.; Tian, Z. miR-374a-5p inhibits non-small cell lung cancer cell proliferation and migration via targeting NCK1. Exp. Ther. Med. 2021, 22, 943. [Google Scholar] [CrossRef]
  112. Ling, D.J.; Chen, Z.S.; Zhang, Y.D.; Liao, Q.D.; Feng, J.X.; Zhang, X.Y.; Shi, T.S. MicroRNA-145 inhibits lung cancer cell metastasis. Mol. Med. Rep. 2015, 11, 3108–3114. [Google Scholar] [CrossRef]
  113. Li, B.; Ding, C.M.; Li, Y.X.; Peng, J.C.; Geng, N.; Qin, W.W. MicroRNA-145 inhibits migration and induces apoptosis in human non-small cell lung cancer cells through regulation of the EGFR/PI3K/AKT signaling pathway. Oncol. Rep. 2018, 40, 2944–2954. [Google Scholar] [CrossRef]
  114. Jiang, L.P.; He, C.Y.; Zhu, Z.T. Role of microRNA-21 in radiosensitivity in non-small cell lung cancer cells by targeting PDCD4 gene. Oncotarget 2017, 8, 23675–23689. [Google Scholar] [CrossRef]
  115. Jiang, Y.; Chen, X.; Tian, W.; Yin, X.; Wang, J.; Yang, H. The role of TGF-beta1-miR-21-ROS pathway in bystander responses induced by irradiated non-small-cell lung cancer cells. Br. J. Cancer 2014, 111, 772–780. [Google Scholar] [CrossRef] [PubMed]
  116. Han, M.; Wang, Y.; Liu, M.; Bi, X.; Bao, J.; Zeng, N.; Zhu, Z.; Mo, Z.; Wu, C.; Chen, X. MiR-21 regulates epithelial-mesenchymal transition phenotype and hypoxia-inducible factor-1alpha expression in third-sphere forming breast cancer stem cell-like cells. Cancer Sci. 2012, 103, 1058–1064. [Google Scholar] [CrossRef] [PubMed]
  117. Xie, Z.; Cao, L.; Zhang, J. miR-21 modulates paclitaxel sensitivity and hypoxia-inducible factor-1alpha expression in human ovarian cancer cells. Oncol. Lett. 2013, 6, 795–800. [Google Scholar] [CrossRef]
  118. Jiang, S.; Wang, R.; Yan, H.; Jin, L.; Dou, X.; Chen, D. MicroRNA-21 modulates radiation resistance through upregulation of hypoxia-inducible factor-1alpha-promoted glycolysis in non-small cell lung cancer cells. Mol. Med. Rep. 2016, 13, 4101–4107. [Google Scholar] [CrossRef]
  119. Sasahira, T.; Kurihara, M.; Bhawal, U.K.; Ueda, N.; Shimomoto, T.; Yamamoto, K.; Kirita, T.; Kuniyasu, H. Downregulation of miR-126 induces angiogenesis and lymphangiogenesis by activation of VEGF-A in oral cancer. Br. J. Cancer 2012, 107, 700–706. [Google Scholar] [CrossRef]
  120. Ma, Y.; Shen, N.; Wicha, M.S.; Luo, M. The Roles of the Let-7 Family of MicroRNAs in the Regulation of Cancer Stemness. Cells 2021, 10, 2415. [Google Scholar] [CrossRef] [PubMed]
  121. Kumar, M.S.; Erkeland, S.J.; Pester, R.E.; Chen, C.Y.; Ebert, M.S.; Sharp, P.A.; Jacks, T. Suppression of non-small cell lung tumor development by the let-7 microRNA family. Proc. Natl. Acad. Sci. USA 2008, 105, 3903–3908. [Google Scholar] [CrossRef] [PubMed]
  122. Ruan, L.; Chen, J.; Ruan, L.; Tan, A.; Wang, P. miR-34a inhibits tumorigenesis of NSCLC via targeting SIRT6. Int. J. Clin. Exp. Pathol. 2018, 11, 1135–1145. [Google Scholar]
  123. Zhang, T.; Hu, Y.; Yang, N.; Yu, S.; Pu, X. The microRNA-34 Family and Its Functional Role in Lung Cancer. Am. J. Clin. Oncol. 2024, 47, 448–457. [Google Scholar] [CrossRef]
  124. Ballou, L.M.; Lin, R.Z. Rapamycin and mTOR kinase inhibitors. J. Chem. Biol. 2008, 1, 27–36. [Google Scholar] [CrossRef]
  125. Costa, D.B.; Nguyen, K.S.; Cho, B.C.; Sequist, L.V.; Jackman, D.M.; Riely, G.J.; Yeap, B.Y.; Halmos, B.; Kim, J.H.; Janne, P.A.; et al. Effects of erlotinib in EGFR mutated non-small cell lung cancers with resistance to gefitinib. Clin. Cancer Res. 2008, 14, 7060–7067. [Google Scholar] [CrossRef]
  126. Hu, Y.; Hong, Y.; Xu, Y.; Liu, P.; Guo, D.H.; Chen, Y. Inhibition of the JAK/STAT pathway with ruxolitinib overcomes cisplatin resistance in non-small-cell lung cancer NSCLC. Apoptosis 2014, 19, 1627–1636. [Google Scholar] [CrossRef] [PubMed]
  127. Ferrara, N.; Hillan, K.J.; Novotny, W. Bevacizumab (Avastin), a humanized anti-VEGF monoclonal antibody for cancer therapy. Biochem. Biophys. Res. Commun. 2005, 333, 328–335. [Google Scholar] [CrossRef] [PubMed]
  128. Herbst, R.S.; Giaccone, G.; de Marinis, F.; Reinmuth, N.; Vergnenegre, A.; Barrios, C.H.; Morise, M.; Felip, E.; Andric, Z.; Geater, S.; et al. Atezolizumab for First-Line Treatment of PD-L1-Selected Patients with NSCLC. N. Engl. J. Med. 2020, 383, 1328–1339. [Google Scholar] [CrossRef] [PubMed]
  129. Zou, R.; Loke, S.Y.; Tan, V.K.; Quek, S.T.; Jagmohan, P.; Tang, Y.C.; Madhukumar, P.; Tan, B.K.; Yong, W.S.; Sim, Y.; et al. Development of a microRNA Panel for Classification of Abnormal Mammograms for Breast Cancer. Cancers 2021, 13, 2130. [Google Scholar] [CrossRef]
  130. Dama, E.; Colangelo, T.; Cuttano, R.; Dziadziuszko, R.; Dandekar, T.; Widlak, P.; Rzyman, W.; Veronesi, G.; Bianchi, F. A plasma 9-microRNA signature for lung cancer early detection: A multicenter analysis. Biomark. Res. 2025, 13, 74. [Google Scholar] [CrossRef]
  131. Sur, D.; Advani, S.; Braithwaite, D. MicroRNA panels as diagnostic biomarkers for colorectal cancer: A systematic review and meta-analysis. Front. Med. 2022, 9, 915226. [Google Scholar] [CrossRef]
  132. Zhong, Y.; Ding, X.; Bian, Y.; Wang, J.; Zhou, W.; Wang, X.; Li, P.; Shen, Y.; Wang, J.J.; Li, J.; et al. Discovery and validation of extracellular vesicle-associated miRNAs as noninvasive detection biomarkers for early-stage non-small-cell lung cancer. Mol. Oncol. 2021, 15, 2439–2452. [Google Scholar] [CrossRef] [PubMed]
  133. Ying, L.; Du, L.; Zou, R.; Shi, L.; Zhang, N.; Jin, J.; Xu, C.; Zhang, F.; Zhu, C.; Wu, J.; et al. Development of a serum miRNA panel for detection of early stage non-small cell lung cancer. Proc. Natl. Acad. Sci. USA 2020, 117, 25036–25042. [Google Scholar] [CrossRef]
  134. Kamkar, L.; Saberi, S.; Totonchi, M.; Kavousi, K. Circulating microRNA panels for multi-cancer detection and gastric cancer screening: Leveraging a network biology approach. BMC Med. Genom. 2025, 18, 27. [Google Scholar] [CrossRef]
  135. Wu, P.; Li, D.; Zhang, C.; Dai, B.; Tang, X.; Liu, J.; Wu, Y.; Wang, X.; Shen, A.; Zhao, J.; et al. A unique circulating microRNA pairs signature serves as a superior tool for early diagnosis of pan-cancer. Cancer Lett. 2024, 588, 216655. [Google Scholar] [CrossRef]
  136. Wang, Y.; Zhao, H.; Gao, X.; Wei, F.; Zhang, X.; Su, Y.; Wang, C.; Li, H.; Ren, X. Identification of a three-miRNA signature as a blood-borne diagnostic marker for early diagnosis of lung adenocarcinoma. Oncotarget 2016, 7, 26070–26086. [Google Scholar] [CrossRef]
  137. Wang, P.; Yang, D.; Zhang, H.; Wei, X.; Ma, T.; Cheng, Z.; Hong, Q.; Hu, J.; Zhuo, H.; Song, Y.; et al. Early Detection of Lung Cancer in Serum by a Panel of MicroRNA Biomarkers. Clin. Lung Cancer 2015, 16, 313–319.e1. [Google Scholar] [CrossRef] [PubMed]
  138. Zheng, D.; Haddadin, S.; Wang, Y.; Gu, L.Q.; Perry, M.C.; Freter, C.E.; Wang, M.X. Plasma microRNAs as novel biomarkers for early detection of lung cancer. Int. J. Clin. Exp. Pathol. 2011, 4, 575–586. [Google Scholar]
  139. Lv, S.; Xue, J.; Wu, C.; Wang, L.; Wu, J.; Xu, S.; Liang, X.; Lou, J. Identification of A Panel of Serum microRNAs as Biomarkers for Early Detection of Lung Adenocarcinoma. J. Cancer 2017, 8, 48–56. [Google Scholar] [CrossRef]
  140. Peng, H.; Wang, J.; Li, J.; Zhao, M.; Huang, S.K.; Gu, Y.Y.; Li, Y.; Sun, X.J.; Yang, L.; Luo, Q.; et al. A circulating non-coding RNA panel as an early detection predictor of non-small cell lung cancer. Life Sci. 2016, 151, 235–242. [Google Scholar] [CrossRef] [PubMed]
  141. Yang, X.; Zhang, Q.; Zhang, M.; Su, W.; Wang, Z.; Li, Y.; Zhang, J.; Beer, D.G.; Yang, S.; Chen, G. Serum microRNA Signature Is Capable of Early Diagnosis for Non-Small Cell Lung Cancer. Int. J. Biol. Sci. 2019, 15, 1712–1722. [Google Scholar] [CrossRef] [PubMed]
  142. Leng, Q.; Lin, Y.; Jiang, F.; Lee, C.J.; Zhan, M.; Fang, H.; Wang, Y.; Jiang, F. A plasma miRNA signature for lung cancer early detection. Oncotarget 2017, 8, 111902–111911. [Google Scholar] [CrossRef]
  143. Abdipourbozorgbaghi, M.; Vancura, A.; Radpour, R.; Haefliger, S. Circulating miRNA panels as a novel non-invasive diagnostic, prognostic, and potential predictive biomarkers in non-small cell lung cancer (NSCLC). Br. J. Cancer 2024, 131, 1350–1362. [Google Scholar] [CrossRef] [PubMed]
  144. Wang, K.; Yuan, Y.; Cho, J.H.; McClarty, S.; Baxter, D.; Galas, D.J. Comparing the MicroRNA spectrum between serum and plasma. PLoS ONE 2012, 7, e41561. [Google Scholar] [CrossRef] [PubMed]
  145. Kim, J.O.; Gazala, S.; Razzak, R.; Guo, L.; Ghosh, S.; Roa, W.H.; Bedard, E.L. Non-small cell lung cancer detection using microRNA expression profiling of bronchoalveolar lavage fluid and sputum. Anticancer Res. 2015, 35, 1873–1880. [Google Scholar]
  146. Gahlawat, A.W.; Fahed, L.; Witte, T.; Schott, S. Total circulating microRNA level as an independent prognostic marker for risk stratification in breast cancer. Br. J. Cancer 2022, 127, 156–162. [Google Scholar] [CrossRef]
  147. Fehlmann, T.; Lehallier, B.; Schaum, N.; Hahn, O.; Kahraman, M.; Li, Y.; Grammes, N.; Geffers, L.; Backes, C.; Balling, R.; et al. Common diseases alter the physiological age-related blood microRNA profile. Nat. Commun. 2020, 11, 5958. [Google Scholar] [CrossRef]
  148. Rao, Y.S.; Mott, N.N.; Wang, Y.; Chung, W.C.; Pak, T.R. MicroRNAs in the aging female brain: A putative mechanism for age-specific estrogen effects. Endocrinology 2013, 154, 2795–2806. [Google Scholar] [CrossRef]
  149. Peltier, H.J.; Latham, G.J. Normalization of microRNA expression levels in quantitative RT-PCR assays: Identification of suitable reference RNA targets in normal and cancerous human solid tissues. RNA 2008, 14, 844–852. [Google Scholar] [CrossRef]
  150. Maiocchi, S.; Collins, E.N.; Peterson, A.R.; Alexander, K.C.; McGlamery, D.J.; Cassidy, N.A.; Ikonomidis, J.S.; Akerman, A.W. Plasma microrna quantification protocol. Vessel Plus 2023, 7, 27. [Google Scholar] [CrossRef]
  151. Faraldi, M.; Gomarasca, M.; Sansoni, V.; Perego, S.; Banfi, G.; Lombardi, G. Normalization strategies differently affect circulating miRNA profile associated with the training status. Sci. Rep. 2019, 9, 1584. [Google Scholar] [CrossRef]
  152. Kauko, O.; Laajala, T.D.; Jumppanen, M.; Hintsanen, P.; Suni, V.; Haapaniemi, P.; Corthals, G.; Aittokallio, T.; Westermarck, J.; Imanishi, S.Y. Label-free quantitative phosphoproteomics with novel pairwise abundance normalization reveals synergistic RAS and CIP2A signaling. Sci. Rep. 2015, 5, 13099. [Google Scholar] [CrossRef] [PubMed]
  153. Marabita, F.; de Candia, P.; Torri, A.; Tegner, J.; Abrignani, S.; Rossi, R.L. Normalization of circulating microRNA expression data obtained by quantitative real-time RT-PCR. Brief. Bioinform. 2016, 17, 204–212. [Google Scholar] [CrossRef]
  154. Dhilipkannah, P.; Sachdeva, A.; Holden, V.K.; Jiang, F. Integrative Biomarker Panel for Improved Lung Cancer Diagnosis Using Plasma microRNAs and Sputum Bacterial DNA. Curr. Oncol. 2024, 31, 5949–5959. [Google Scholar] [CrossRef] [PubMed]
  155. Hu, Y.; Wang, L.; Gu, J.; Qu, K.; Wang, Y. Identification of microRNA differentially expressed in three subtypes of non-small cell lung cancer and in silico functional analysis. Oncotarget 2017, 8, 74554–74566. [Google Scholar] [CrossRef] [PubMed]
  156. Sens, A.; Rischke, S.; Hahnefeld, L.; Dorochow, E.; Schafer, S.M.G.; Thomas, D.; Kohm, M.; Geisslinger, G.; Behrens, F.; Gurke, R. Pre-analytical sample handling standardization for reliable measurement of metabolites and lipids in LC-MS-based clinical research. J. Mass. Spectrom. Adv. Clin. Lab. 2023, 28, 35–46. [Google Scholar] [CrossRef]
  157. Zafar, S.; Hafeez, A.; Shah, H.; Mutiullah, I.; Ali, A.; Khan, K.; Figueroa-Gonzalez, G.; Reyes-Hernandez, O.D.; Quintas-Granados, L.I.; Pena-Corona, S.I.; et al. Emerging biomarkers for early cancer detection and diagnosis: Challenges, innovations, and clinical perspectives. Eur. J. Med. Res. 2025, 30, 760. [Google Scholar] [CrossRef]
  158. Metcalf, G.A.D. MicroRNAs: Circulating biomarkers for the early detection of imperceptible cancers via biosensor and machine-learning advances. Oncogene 2024, 43, 2135–2142. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Article Metrics

Citations

Article Access Statistics

Multiple requests from the same IP address are counted as one view.