Next Article in Journal
Endoscopic Ultrasound Guided Fine Needle Aspiration versus Endoscopic Ultrasound Guided Fine Needle Biopsy for Pancreatic Cancer Diagnosis: A Systematic Review and Meta-Analysis
Next Article in Special Issue
Nectins and Nectin-like Molecules in Colorectal Cancer: Role in Diagnostics, Prognostic Values, and Emerging Treatment Options: A Literature Review
Previous Article in Journal
Left Ventricle Wall Motion Analysis with Real-Time MRI Feature Tracking in Heart Failure Patients: A Pilot Study
Previous Article in Special Issue
Identification and Application of a Novel Immune-Related lncRNA Signature on the Prognosis and Immunotherapy for Lung Adenocarcinoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Bronchoalveolar Lavage Fluid-Isolated Biomarkers for the Diagnostic and Prognostic Assessment of Lung Cancer

by
Alexandros Kalkanis
1,*,
Dimitrios Papadopoulos
1,
Dries Testelmans
1,
Alexandra Kopitopoulou
2,
Eva Boeykens
2 and
Els Wauters
1
1
Department of Respiratory Diseases, University Hospitals Leuven, Campus Gasthuisberg, 3000 Leuven, Belgium
2
Department of Pulmonology, General Hospital Rivierenland, Campus Bornem, 2880 Bornem, Belgium
*
Author to whom correspondence should be addressed.
Diagnostics 2022, 12(12), 2949; https://doi.org/10.3390/diagnostics12122949
Submission received: 31 October 2022 / Revised: 18 November 2022 / Accepted: 22 November 2022 / Published: 25 November 2022
(This article belongs to the Special Issue Clinical Prognostic and Predictive Biomarkers)

Abstract

:
Lung cancer is considered one of the most fatal malignant neoplasms because of its late detection. Detecting molecular markers in samples from routine bronchoscopy, including many liquid-based cytology procedures, such as bronchoalveolar lavage fluid (BALF), could serve as a favorable technique to enhance the efficiency of a lung cancer diagnosis. BALF analysis is a promising approach to evaluating the tumor progression microenvironment. BALF’s cellular and non-cellular components dictate the inflammatory response in a cancer-proliferating microenvironment. Furthermore, it is an essential material for detecting clinically significant predictive and prognostic biomarkers that may aid in guiding treatment choices and evaluating therapy-induced toxicities in lung cancer. In the present article, we have reviewed recent literature about the utility of BALF analysis for detecting markers in different stages of tumor cell metabolism, employing either specific biomarker assays or broader omics approaches.

1. Introduction

Lung cancer is among the most common malignant neoplasms and is associated with a poor prognosis since it is usually diagnosed at an advanced stage [1]. However, recent advances in the field of systemic treatment have changed the landscape of the disease. Much of this progress has been achieved by the discovery and utility of predictive biomarkers, whose detection may guide targeted therapies in an attempt to a more personalized approach to treatment. In cancer research, a biomarker is a biological molecule whose levels can be measured in tissue or fluids and indicate the presence or progress of the disease or its response to specific therapies [2]. Thus, biomarkers may aid in determining the diagnosis, prognosis, and prediction of treatment response in cancer patients. Any component of the cancer cell metabolic pathway, starting from gene alterations and ending with the various metabolites, can be tested as a biomarker according to its biological role. In recent years, the contemporary measurement of multiple substances belonging to the same level in the metabolic cascade has been possible with the use of omics technologies, making feasible the identification of biomarker profiles or signatures [2].
A major problem in employing lung cancer biomarker testing in everyday clinical practice is obtaining adequate tissue samples. Most patients are diagnosed based on small bioptic or even more limited cytological materials obtained during bronchoscopy or transthoracic aspiration. These materials are often depleted during standard histological and histochemical procedures used for diagnosis and tumor subtyping, leaving no space for biomarker testing. Fluids, on the other hand, are promising materials for biomarker evaluation since most substances, from DNA to proteins and metabolites, are being excreted from cancer cells in soluble form. Bronchial or bronchoalveolar lavage fluid (BALF) is obtained by a lung irrigation technique routinely performed during bronchoscopy. It allows the harvesting of cellular and non-cellular contents of the bronchial and alveolar space, serving as an excellent marker of the tumor microenvironment [3]. Its collection requires a minimally invasive procedure that can be repeated with minimum risk, providing a valid and adequate medium for biomarker testing in lung cancer. In addition, due to its proximity to the neoplastic tissue, it could have higher sensitivity for biomarker detection, especially in locally advanced non-metastasized lung cancer, compared to other fluids commonly employed for the same reason, such as plasma or pleural fluid [4].
BALF analysis in lung cancer has multiple uses. Numerous research studies have demonstrated the benefit of employing BALF for the cytological identification of malignant lung neoplasms. Malignant cells may be shed from an adjacent lung tumor into the respective bronchoalveolar space and identified with conventional cytology. Although BALF cytology alone has a modest sensitivity for diagnosing lung cancer (between 29% and 69%), the specificity is exceptionally high (between 90% and 100%) [5]. Furthermore, its combination with other histological techniques seems to increase the diagnostic yield. The diagnostic accuracy of transbronchial lung biopsy in endoscopically non-visible lesions was enhanced when combined with BALF cytology [6]. Secondly, the parenchymal cells of the lungs secrete different types of soluble substances, which can be identified and measured in BALF after removing the cellular component with centrifugation (supernatant). With the advent of novel molecular techniques, it has become possible to amplify nucleic acids that are either extracted from cells or extracellular vesicles (EVs) or detected in cell-free form on BALF and perform a wide array of genomic, epigenomic, and transcriptomic testing. Finally, BALF analysis may aid in identifying various treatment-related adverse events in the lungs, such as pulmonary infections or pneumonitis.
This review aims to perform a brief and comprehensive overview of studies published in the previous decade that focus on the utility of BALF for detecting different forms of diagnostic, predictive, and prognostic biomarkers in lung cancer. Studies were identified by searching the MEDLINE/PubMed electronic database for relative reports dating from 2011 to September 2022. The search strategy included a combination of MeSH terms (biomarkers; bronchoalveolar lavage; lung neoplasm) and keywords (biomarker; [prognostic or predictive] marker; [bronchoalveolar or bronchial or lung] lavage; [bronchial or lung] washing; [lung or pulmonary] [cancer or neoplasm or tumor]). According to our judgement, some older seminal articles are also mentioned based on their influence on the afterward-growing literature on the subject.

2. BALF Biomarkers for the Diagnosis and Prognosis of Lung Cancer

2.1. Genetic Biomarkers

Recent studies have conducted various assessments on genetic markers at a large, genome-wide scale to identify more specific and sensitive molecules for targeted drug delivery methods to cure the disease. A recent study compared tumor-derived mutations in similar plasma and BALF samples from patients with non-small-cell lung cancer (NSCLC). Researchers discovered that BALF cell-free DNA (cfDNA) testing is more accurate than serum for detecting mutations related to lung cancer [7]. Clinical applications of BALF cfDNA testing include the detection of mutations in affected individuals and, perhaps, the detection of lung adenocarcinoma. It is convenient to identify mutations related to tumors by targeted sequencing of BALF cfDNA, and this method seems more robust compared to plasma screening. Bronchial washings are also a viable and practical choice for genome-wide molecular studies. Actionable mutations in cancer genes were detected with a higher than 90% concordance with respective gene mutations in tumor biopsies [8].

2.1.1. EGFR Mutations

With cfDNA collected through the BALF supernatant, a polymerase chain reaction (PCR) technique may identify functional EGFR mutations. This might be a quick and accurate way to diagnose NSCLC simultaneously using DNA analysis and morphology [9]. The tissue biopsy results are compared to the detectability of activating EGFR mutations in BALF and blood plasma samples in another study [10]. Under this investigation, BALF was found to be significantly more sensitive than liquid biopsy at detecting an EGFR mutation in the same patients (92.3% vs. 38.5%) [10]. A real-time peptide nucleic acid (PNA)-mediated polymerase chain clamping assay was used to detect the T790M EGFR mutation on both BALF and bronchial biopsy specimens of cancer-afflicted persons [11]. In a different, more thorough investigation, 20 patients with lung adenocarcinoma had their EGFR mutations and T790M mutations molecularly tested using cfDNA from BALF. The combination of PNA-mediated clamping PCR and the PANAMutyperTM R EGFR kit with PNA clamping-assisted fluorescence melting curve analysis produced 91.7% concordance with the results from a tumor biopsy [12]. In another study of patients with advanced NSCLC, cytology samples from BALF were used to identify the T790M EGFR mutation. The findings demonstrated that the clinical benefit of osimertinib treatment was predicted by cytology sample EGFR T790M positivity [13]. Isolated EVs from BALF include DNA that can be used for EGFR genotyping by liquid biopsy. Particularly when compared to liquid biopsy cfDNA, BALF EV DNA is tissue specific and incredibly sensitive. Additionally, tissue re-biopsy is less effective than BALF EV DNA at identifying the T790M mutation in individuals who developed resistance to EGFR tyrosine kinase inhibitors (TKIs) [14].

2.1.2. KRAS Mutations

Clinical uses for KRAS mutation analysis in lung disease include its use as a diagnostic indicator for cancer in sputum and BALF samples [15]. In adenocarcinomas of the lung and related sputum and BALF samples, matched KRAS mutations have been found [16]. Furthermore, it has been demonstrated that patients with negative cytological results may benefit from screening for KRAS mutations in BALF cells to help with a lung cancer diagnosis [17]. KRAS and p53 gene mutations in BALF may serve as helpful biomarkers for the diagnosis of peripheral NSCLC, according to the findings of a recent study [18].

2.1.3. ALK Translocations

Reverse transcription-PCR has been used to detect ALK translocations in cytology samples having a 97% concordance with tissue samples [19].

2.2. Epigenetic Biomarkers

DNA methylation markers are frequently employed as biomarkers for early diagnosis or recurrence of cancer. DNA methylation occurs when a methyl group is attached to a cytosine base located in a CpG dinucleotide, which may impact gene transcription. Aberrant DNA methylation can reduce the expression of tumor-suppressor genes and allow cancer cells proliferation. Kim et al. discovered that p16, RASSF1A, H-cadherin, and RAR beta gene methylation in tumors may be valuable biomarkers for the early diagnosis of NSCLC in BALF [20]. Moreover, several other studies also reported the potential application value of p16 promoter methylation in sputum for lung cancer diagnosis [21,22]. A large retrospective cohort showed improved diagnostic efficacy of DNA methylation biomarkers (DNA methylation of p16, TERT, WT1, and RASSF1 gene) in cytological bronchial washings evaluation [23]. When lung cancer patients have bronchial aspirates, the Epi pro-Lung BL Reflex Assay is a powerful and practical diagnostic technique for detecting elevated DNA methylation of the SHOX2 gene locus [24]. In comparison to conventional cytology analysis and serum CEA, the methylation analysis of the SHOX2 and RASSF1A panel in BALF using RT-PCR produced better diagnostic sensitivity (81.0%) and specificity (97.4%) [25]. NSCLC could be found by examining the DNA methylation of 7CpGs (TBX15, PHF11, TOX2, PRR15, TFAP2A, HOXA1, and PDGFRA genes) in bronchial washings [26]. A 5-marker (LHX9, GHSR, HOXA11, PTGER4-2, HOXB4-3) methylation model was recently found to have 70% sensitivity and 82% specificity for discriminating malignant from benign pulmonary nodules [27]. According to a meta-analysis, the detection of DNA methylation of the p16INK4a gene in BALF may be a potential biomarker for NSCLC diagnosis [28].

2.3. Post-Transcriptional Biomarkers

Transcriptome comparison allows for the discovery of genes that are differently expressed in various cell groups or in response to different treatments. Researchers have showed that clusters of deregulated miRNAs were expressed in the BALF of smoking-related diseases such as lung adenocarcinoma and COPD [29]. Similarly, the expression of two anti-apoptotic genes, survivin and livin, through their mRNA levels was observed in lung cancer patients versus patients with benign lung disease, using bronchial aspirates as a sample source [30]. Other studies have broadened the target transcriptomes to other specific panels of miRNAs that were significantly upregulated in patients with lung cancer [31,32]. Cluster analysis of the expression levels of a miRNA panel in BALF samples from NSCLC patients provided a diagnostic sensitivity of 85.7% and specificity of 100% [33]. Moreover, a prediction model of specific miRNA biomarkers from liquid cytological specimens has shown potential in discriminating squamous cell carcinoma from adenocarcinoma [34]. Transcriptome signatures can also be potentially helpful in predicting survival for patients with lung cancer. Patients with a low expression level of a 3-miRNA panel were associated with better overall survival [35]. Evaluation of EVs from plasma and BALF in patients with NSCLC and benign lung diseases revealed significant differences in exosome amount and miRNA content between fluids and patient groups, revealing their specific role as biomarkers [36]. A significant increase in the expression profile of immunoglobulin genes in BALF was found between lung cancer and healthy controls in a recent transcriptomics study, which generated a 53-gene signature that showed a significant correlation with inhibitory checkpoint PDCD1 [37].

2.4. Post-Translational Biomarkers

2.4.1. Proteins

Proteins in BALF can represent the physiological and pathological state of the lung. Proteins that lung cancer cells secrete into BALF may be utilized as biomarkers to detect and evaluate malignancy, aiding lung cancer diagnosis, prognosis, subtyping, and therapy response monitoring. Researchers have since long identified a link between bronchial secretions and serum tumor marker concentrations [38]. Among many proteins, the most specific is napsin A, a renowned immunohistochemical marker for lung cancer diagnosis in affected individuals [39]. In addition, levels of several BALF proteins located in antioxidant, inflammatory, or anti-angiogenic pathways, such as cytokines [40,41,42] and growth [43,44,45], pro-angiogenic [46,47,48], or complement [49,50,51] factors, have been evaluated as diagnostic or prognostic biomarkers employing antibody-specific immunoassays.
Combining proteomics, mass spectrometry (MS), and affinity chemistry-based methods have significantly improved our understanding of the protein oxidative changes that take place in many biological specimens under varied physiological and pathological circumstances in recent years (Figure 1). Glycoproteins are crucial in the standard processing of biological processes such as cell separation, growth, their close contact with their vicinity and invading of tumor cells into surrounding cells. A cohort study used solid-phase N-proteoglycans extraction, iTRAQ labeling, and liquid chromatography-tandem MS to analyze N-proteoglycans levels in BALF from patients with lung cancer and benign lung diseases. Levels of 8 glycoproteins (neutrophil elastase, integrin alpha-M, cullin-4B, napsin A, lysosome-associated membrane protein 2, cathepsin D, BPI fold-containing family B member 2, and neutrophil gelatinase-associated lipocalin) displayed more than two times rise in cancer BALF compared to benign BALF [52]. According to research by Uribarri et al. the expression levels of APOA1, CO4A, CRP, GSTP1, and SAMP led to a diagnostic panel for lung cancer that was 95% sensitive and 81% specific. The measurement of STMN1 and GSTP1 proteins enabled the two main subtypes of lung cancer (non-small-cell and small-cell) to be distinguished with 57% specificity and 90% sensitivity [53]. Ortea et al. identified different proteins in the BALF of patients with lung cancer, including glutathione S-transferase pi, haptoglobin, and complement C4-A. These different types of proteins could be proved as prominent biomarkers for disease diagnosis [54]. Almatroodi et al. performed a quantitative proteomic evaluative study on BALF of patients with lung adenocarcinoma, and their results indicated the occurrence of 1100 proteins in BALF. Among them, the ratio of over-expressed proteins in lung adenocarcinoma individuals were 33 compared to healthy individuals. S100-A8, thymidine phosphorylase, annexin A2, transglutaminase 2, and annexin A1 were lung cancer individuals’ most renowned over-expressed proteins [55]. In another prospective cohort of individuals with suspected lung cancer, proteomic analysis of BALF samples identified 133 proteins that could differentiate cancer and non-cancer patients [56]. Using label-free MS analysis of BALF, Hmmier et al. discovered that 4 proteins (cystatin-C, TIMP1, lipocalin 2, and HSP70/HSPA1A) were significantly overexpressed in lung cancer patients compared to healthy controls [57]. A recent investigation evaluated aberrant protein glycosylation using lectin microarrays in BALF. It revealed 15 lectins that could distinguish between the different lung cancer types and 14 lectins whose levels differed between early and advanced disease stages [58].

2.4.2. Cell Epitopes

The combination of immunoassays and flow cytometry in BALF supernatant enables the study of cell epitopes, mainly used in identifying immune cell subpopulations (Figure 2). T cells are essential for antitumor defense, but as cancer progresses, their population changes. Numerous suppressory and regulatory systems can prevent lymphocyte activation and prevent the identification of lung cancer antigens. Various research studies indicated that now lung cancer immunotherapeutic therapy aims to enhance the cytotoxological action of lymphocytes by restricting the activities of suppressor molecules such as cytotoxic T cell antigen 4 (CTLA-4) and programmed death-1 (PD-1) [59]. The examination of BALF revealed that one could easily analyze the different types of immune responses in the tumor microenvironment as well as an alteration in the status of various cells. A higher ratio of cytotoxic CD8+ lymphocytes, neutrophils, T cells, and CTLA-4+ T regulatory cells was found in BALF of the cancer-affected lung compared to the healthy contralateral lung [60]. Moreover, tissue samples of lung adenocarcinoma patients also indicated the presence of M2 polarization of macrophages, which are further associated with the onset of lung cancer [61]. Osińska et al. examined the BALF of 35 patients with lung cancer to figure out the existence of regulatory T cells (Tregs) and their specific role in boosting immunity. Flow cytometry analysis revealed that Treg cells were present in significant proportion in the local environment of lung cancer cells as compared to normal cells. These Tregs are renowned as anticancer defense cells because they have the potential to restrict the normal functioning of NK cells, T-lymphocytes, and dendritic cells, which ultimately enhances the immune tolerance level [62]. In 2018, Hu et al. examined the significant ratio of PD-1+ cells in BALF and peripheral blood in small-cell lung cancer (SCLC) individuals, and their levels dropped after chemotherapy. Therefore, these cells would be potential biomarkers for the diagnosis of diseases as these programmed death cells are recognized as checkpoints in the immune system, and their inhibition could potentially mediate the activation of T cells, ultimately exhibiting antitumor activity [63]. Moreover, another rational study was carried out on BALF as a prognostic agent for patients of lung cancer, and through a cytometric bead array, Hu et al. identified a higher level of interleukin (IL)-10 and IL-10+CD206+CD14+M2-like macrophages in the SCLC affected individuals as compared to NSCLC individuals. These markers were also correlated with advanced stage and reduced survival of SCLC patients [64]. Recently, a cohort study was conducted by Masuhiro et al. to examine the immune profile of tumor microenvironment through BALF of NSCLC individuals according to their response to immunotherapy. They observed a higher level of CXCL9 and a higher ratio of CD56+ cytotoxic T cells in BALF of patients responding to nivolumab compared to those not responding [65].
Some cells disassociate themselves from the primary tumor mass and enter the circulatory system. These cells are known as circulating tumor cells (CTCs) and emerge as a new target that offers clinical insights for the prediction and diagnosis of different types of cancers [66]. Recent studies have demonstrated that the precision and sensitivity of CTCs detected by the chromosomal enumeration probe 8 (CEP8) in lung cancer patients was, correspondingly, 83.3% and 98.6% [67]. Several studies have investigated these distinct CEP8+ CTCs, and the results have demonstrated a substantial correlation between these cells with the detection of lung cancer and also its prognosis with excellent precision and sensitivity [68,69]. Detection of CTCs in serum and BALF showed that CEP8+ CTCs could be used as a supplementary approach for individuals with solitary pulmonary nodules to diagnose lung cancer at an early stage [70].

2.5. Metabolite Biomarkers

Metabolites of tumor cells, such as amino acids, nucleic acids, lipids, and sugars, may reflect cellular metabolic processes that drive tumor formation and progression. Either by using metabolite-specific immunoassays or MS-based metabolomic approaches (Figure 3), recent studies have compared metabolites between lung cancer patients and controls. In an early study, levels of leukotriene B4 and cysteinyl leukotrienes, which are end products of arachidonic acid metabolism, were significantly elevated in BALF of NSCLC patients [71]. In another report, patients with NSCLC had significantly lower ATP and ADP concentrations in BALF than patients with COPD, an effect that was exerted by upregulated expression of P2Y1, P2X4, and P2X7 purinergic receptors [72]. Callejón-Leblic et al. identified 42 altered metabolites in BALF of lung cancer patients compared to non-cancer patients. Based on ROC curve analysis, levels of carnitine, adenine, choline, glycerol, and phosphoric acid show high sensitivity and specificity to distinguish between lung cancer and controls and should be considered as potential biomarkers [73].
Certain elements such as essential and non-essential metals (Fe, Cd, Zn, Co, Cu, Pb, V, Cr, etc.) are important for maintaining homeostasis and are involved in several cellular pathways. Excess or deficiency of such elements causes dyshomeostasis and disturbs cellular pathways, ultimately leading to the progression of serious illness [74]. So, identification of levels of such elements in body fluids such as serum and BALF can help to understand the mechanisms involved in the progression of serious illnesses such as lung cancer. As a result, variations in the particular elements’ concentrations and the profile of their chemical components can reveal the individual’s metabolic and nutritional status and therefore offers a prospective early prognosis of cancer progression. Belén et al. used triple quadrupole inductively coupled plasma MS analysis and identified the presence of metals in body fluids, including BALF, in lung cancer patients and concluded that such metals could be employed as a potent biological marker for more accurate and expeditious identification of lung adenocarcinoma in affected individuals [75].

2.6. Metagenomic Biomarkers

Microbiome generally contributes towards barrier formation, external communication, and immune homeostasis in healthy individuals. The microbiome can also trigger host immune responses against cancer cells. Several studies have shown the interconnectedness between lung cancer and the microbiome. For example, Wang et al. conducted a preliminary analysis of microbiome diversity in saliva and BALF of lung cancer patients [76]. They observed that the microbiome diversity is significantly less than in the healthy samples, indicating a link between cancer and microbiome. It can also be said that certain microbiome genera could act as potential lung cancer biomarkers. In this regard, Cheng et al. characterized microbiomes in BALF for potential associations with lung cancer [77]. The microbiome diversity was compared between diseased and healthy samples using principal coordinate analysis. They observed six different genera that were significantly enriched in BALF of cancer patients: Blautia, Capnocytophaga, Gemmiger, Oscillospira, Sediminibacterium, and TM7-3. Similarly, Lee et al. investigated the microbiome in BALF of the affected group and compared them with benign lung mass [78]. They reported that members of the genera Megasphaera and Veillonella are potential cancer markers as their concentrations are relatively higher in cancer patients than in the control group. In addition, Patnaik et al. observed that the lower airway microbiome could contribute to the recurrence of early-stage NSCLC [79]. They observed that the microbiome diversity differed significantly between patients with recurrence and non-recurrence after surgery. They also observed that the diversity among 16 out of 18 cancer recurrence patients was similar. Zheng et al. performed metagenomic sequencing on BALF samples from cancer patients and healthy controls [80]. Even though the diversity between both samples was comparable, some rare microbiota stood out in lung cancer samples. For example, species such as Bacteroides pyogenes, Chaetomium globosum, Lactobacillus rossiae, Magnetospirillum gryphiswaldense, Paenibacillus odorifer, and Pseudomonas entomophila were common in cancer samples. The results indicate that change in the microbiome’s diversity is directly associated with cancer. Moreover, Veillonella was found to increase tumor burden and decrease survival. Furthermore, the microbiome plays a crucial role in cancer cells’ response to immunotherapy. For example, Jang et al. analyzed the microbiome’s relationship with programmed death-ligand-1 (PD-L1) expression [81]. PD-L1 protein is typically involved in protecting non-cancerous/non-harmful cells from immune cells. The authors also found that the cells with higher PD-L1 expression levels respond better to immunotherapy than the cells with lower PD-L1 expression levels. The population of Veillonella was significantly higher in the group with the higher expression level of PD-L1, while Neisseria was abundant in the group with the lower expression of PD-L1. Moreover, the exact reason for a higher amount of Veillonella in cancer patients is still unknown, but several studies have developed links between different lung-related diseases and the microbiome genera under debate. For example, Veillonella has been found to be involved in pulmonary fibrosis and asthma development [82,83]. As both pulmonary disorders are related to the immune system and lungs, it is also expected that Veillonella will be higher in cancer patients as well.

3. BALF Biomarkers for the Identification of Adverse Events of Lung Cancer Treatment

3.1. Pulmonary Infections

It has been investigated that the risk of developing neutropenia is usually low in the case of lung adenocarcinoma immunotherapeutic procedures compared to chemotherapy [84]. Moreover, long-term immunosuppression therapies with high drug dosage (steroids, TNF-α) are required for immune-related adverse events (irAEs), e.g., colitis and pneumonitis, that ultimately enhance the risk of other infections [85]. Aspergillus fumigatus pneumonia diagnosed after BALF examination has been described in a metastatic melanoma patient treated with systemic corticosteroids and infliximab for an irAE due to ipilimumab [86]. Another report described the detection of Mycobacterium tuberculosis in BALF in a NSCLC patient during third-line nivolumab therapy [87]. Thus, the performance of bronchoscopy and subsequent serological and molecular testing of BALF is an established method for the diagnosis of such infectious complications in this unique patient population [88].

3.2. Therapy-Induced Pulmonary Toxicity

It has been observed that excess application of immunotherapeutic molecules has important side effects; among them, checkpoint inhibitor pneumonitis (CIP) is the most famous. Common clinical presentations of CIP are the onset of dyspnea, hypoxemia, and pulmonary infiltration [89,90]. For advanced NSCLC, the phase I trial of pembrolizumab and nivolumab reported 1 and 3 pneumonitis-related deaths, respectively [91,92]. The extensive examination of BALF showed infiltration and inflammation of lymphocytes in patients with CIP. In 2017, another case study reported T-lymphocytic alveolitis as extrinsic allergic alveolitis in 80% of affected individuals with CD8+ cell predominance [93]. Identifying the dysregulation of immune cell subpopulations in BALF could serve as a promising target for developing therapeutics to minimize adverse effects [94].
Drug-induced sarcoidosis reaction (DISR) can be another after-effect of cancer-related therapies. DISR is a systematic granulomatous reaction that arises after using chemotherapeutic agents and is indistinguishable from sarcoidosis [95]. DISR has been described after treatment with immune checkpoint inhibitors, including PD-L1 inhibitors, PD-1 inhibitors, and anti-CTLA-4 antibodies [96]. Thus, in the absence of biopsy specimens, the surrogate method for the diagnosis of DISR in affected patients is BALF sampling [97]. Montaudie et al. conducted research work, and they presented a DISR-based study that showed 32% lymphocytes along with the enhanced frequency of CD4/CD8 cells [98].
Radiation-induced pneumonitis is an inflammatory process occurring in reaction to chest radiotherapy used to treat thoracic malignancies. Early identification and treatment are crucial, considering the fact that its chronic phase constitutes lung fibrosis. Previous research has detected different types of serum biological markers, including TGF-β, IL-6, TNF-α, and IL-1, that predicted tissue injuries after radiotherapy [99,100]. Later, a series of research studies were performed by Kwang-Joo Park et al. on animal models for the evaluation of radiotherapy-associated injuries of lung tissues, in which the focus was on BALF. Their results revealed that the level of IL-1, NO, TGF-β, and neutrophils were high in BALF [101,102]. Additionally, lymphocytic alveolitis was also found in BALF of affected individuals who took radiotherapy for breast cancer. In most symptomatic cases, a significant rise in CD4+ T lymphocytes was observed, but no increase in CD4/CD8 ratio was found [103]. Crohns et al. reported that radiation therapy resulted in a significant increase in IL-6 in BALF, while elevated IL-8 levels in BALF are associated with worse survival in lung cancer patients receiving radiotherapy [104]. BALF was found to be more effective as compared to serum for the detection of surfactant protein D, which is an important biomarker for radiation-induced pneumonitis in tumor tissues of affected persons [105]. In a cohort study, the profile of cytokines or chemokines was analyzed in BALF samples from cancer-afflicted individuals before and after radiotherapy. This study showed the overexpression of cytokines, including PAI-1, CD154, IL-1ra, CXCL-1, IL-23, MIF, and IFN-γ, in the lungs of patients with high-grade radiation-induced pneumonitis even before radiotherapy [106].

4. The Value of Repeated BALF Examination in the Course of Lung Cancer

With the advent of modern molecular technologies and extensive research work in biological sciences, highly curable therapies for patients with lung cancer have been recently introduced. These treatment methods wrapped new therapeutic molecules as well as more specific biomarkers for the diagnosis of cancer tissues found in different sections of the lungs more accurately. Apart from their highly beneficial results, the continuous re-evaluation of cancer patients is necessary to gauge the tumor status before and after the application of each line of therapy. It has been proposed that the follow-up should not only be restricted to imaging but repeated histological assessment may be needed when relapses occur after therapeutic interventions. Zarogoulidis et al. performed a case study, and their results suggested that re-biopsy of different lung cancer tissues after the administration of TKIs would detect T790M mutations as well as an altered type of lung cancer [107]. Another cohort study reported that 63% of NSCLC patients showed onset of EGFR mutations and proliferation of cancer after treatment with EGFR-TKIs during re-biopsy, while a 33% frequency of T790M mutation was found [108]. Taking into account the low invasiveness and high sensitivity of BALF testing, a surrogate method of the classic re-biopsy technique could be repeated BALF assessment, not only for the detection of T790M mutations but also for a re-evaluation of biomarkers that may predict cancer recurrence or prognosis and guide further treatment choices.

5. Advantages and Disadvantages of BALF Biomarker Testing in Lung Cancer

Biomarker testing is a central component in nowadays personalized management of lung cancer patients. Many approved drugs for the treatment of stage IV NSCLC are being prescribed according to the detection and quantification of specific molecular markers, such as EGFR, ALK, ROS1, and BRAF mutations and PD-L1 expression. In the case of minimal or no tumor tissue material available for biomarker testing, BALF has the advantage of containing a wide variety of molecules, either cell derived or in soluble form, that resemble the tumor molecular profile. Biomarkers with the highest diagnostic, predictive, and prognostic accuracy from the studies reported above are presented in Table 1. High concordance to tissue markers and better accuracy than plasma in oncogene driver detection makes BALF the best alternative for predictive assessment in lung neoplasms. Panels of BALF-isolated biomarkers of pre- and post-transcriptional gene expression regulation, such as DNA methylation and miRNA expression, have higher diagnostic accuracy than BALF cytology for the detection of lung cancer. Moreover, prediction models derived from proteomic, metabolomic, and microbiomic studies have identified novel markers for the discrimination between cancer types and stages and the prediction of immunotherapy response or tumor progression/recurrence.
Main disadvantages of BALF biomarker testing are the lower yield of tumor cells or tumor nucleic acids compared to tissue samples, which could impede biomarker test performance, and the higher invasiveness compared to plasma sampling. Furthermore, continuous validation of the methylomic, transcriptomic, and proteomic signatures in BALF is needed so that they may assist in the diagnosis of small pulmonary nodules and guide treatment approaches in clinical practice.

6. Conclusions

It can be concluded that BALF is an appropriate medium that may aid in the diagnosis of lung cancer, in assessing prognosis and response to therapy, but also in the early identification of treatment-related adverse events. Furthermore, continuous re-evaluation of the genome and the protein and immune status of lung cancer cells is essential in the course of the disease. Repeated BALF examination is a cost-effective and easily accessible method for evaluating the microenvironment and immune status of cancer-afflicted lung tissues and provides a valid comparison of the disease status before and after treatment.

Author Contributions

Conceptualization, A.K. (Alexandros Kalkanis); methodology, A.K. (Alexandros Kalkanis) and D.P.; data curation, A.K. (Alexandros Kalkanis), D.P., D.T. and A.K. (Alexandra Kopitopoulou); writing—original draft preparation, A.K. (Alexandros Kalkanis) and D.P.; writing—review and editing, D.T., A.K. (Alexandra Kopitopoulou), E.B. and E.W.; supervision, E.W. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Howlader, N.; Noone, A.; Krapcho, M.; Miller, D.; Brest, A.; Yu, M.; Ruhl, J.; Tatalovich, Z.; Mariotto, A.; Lewis, D.; et al. SEER Cancer Statistics Review, 1975–2018; National Cancer Institute: Bethesda, MD, USA, 2021.
  2. Henry, N.L.; Hayes, D.F. Cancer biomarkers. Mol. Oncol. 2012, 6, 140–146. [Google Scholar] [CrossRef] [Green Version]
  3. Domagala-Kulawik, J. The relevance of bronchoalveolar lavage fluid analysis for lung cancer patients. Expert Rev. Respir. Med. 2020, 14, 329–337. [Google Scholar] [CrossRef]
  4. Lee, S.H.; Kim, E.Y.; Kim, T.; Chang, Y.S. Compared to plasma, bronchial washing fluid shows higher diagnostic yields for detecting EGFR-TKI sensitizing mutations by ddPCR in lung cancer. Respir. Res. 2020, 21, 142. [Google Scholar] [CrossRef] [PubMed]
  5. Wongsurakiat, P.; Wongbunnate, S.; Dejsomritrutai, W.; Charoenratanakul, S.; Tscheikuna, J.; Youngchaiyud, P.; Pushpakom, R.; Maranetra, N.; Nana, A.; Chierakul, N.; et al. Diagnostic value of bronchoalveolar lavage and postbronchoscopic sputum cytology in peripheral lung cancer. Respirology 1998, 3, 131–137. [Google Scholar] [CrossRef] [PubMed]
  6. Binesh, F.; Pirdehghan, A.; Mirjalili, M.R.; Samet, M.; Majomerd, Z.A.; Akhavan, A. Comparative assessment of the diagnostic value of transbronchial lung biopsy and bronchoalveolar lavage fluid cytology in lung cancer. Asian Pac. J. Cancer Prev. 2015, 16, 201–204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Nair, V.S.; Hui, A.B.-Y.; Chabon, J.J.; Esfahani, M.S.; Stehr, H.; Nabet, B.Y.; Zhou, L.; Chaudhuri, A.A.; Benson, J.; Ayers, K.; et al. Genomic profiling of bronchoalveolar lavage fluid in lung cancer. Cancer Res. 2022, 82, 2838. [Google Scholar] [CrossRef]
  8. Roncarati, R.; Lupini, L.; Miotto, E.; Saccenti, E.; Mascetti, S.; Morandi, L.; Bassi, C.; Rasio, D.; Callegari, E.; Conti, V.; et al. Molecular testing on bronchial washings for the diagnosis and predictive assessment of lung cancer. Mol. Oncol. 2020, 14, 2163–2175. [Google Scholar] [CrossRef] [PubMed]
  9. Kawahara, A.; Fukumitsu, C.; Taira, T.; Abe, H.; Takase, Y.; Murata, K.; Yamaguchi, T.; Azuma, K.; Ishii, H.; Takamori, S.; et al. Epidermal growth factor receptor mutation status in cell-free DNA supernatant of bronchial washings and brushings. Cancer Cytopathol. 2015, 123, 620–628. [Google Scholar] [CrossRef]
  10. Yanev, N.; Mekov, E.; Valev, D.; Yankov, G.; Milanov, V.; Bichev, S.; Gabrovska, N.; Kostadinov, D. EGFR mutation status yield from bronchoalveolar lavage in patients with primary pulmonary adenocarcinoma compared to a venous blood sample and tissue biopsy. PeerJ 2021, 9, e11448. [Google Scholar] [CrossRef]
  11. Prim, N.; Quoix, E.; Beau-Faller, M. Tumor cell content for selection of molecular techniques for T790M EGFR mutation detection in non-small cell lung cancer. J. Thorac. Oncol. 2011, 6, 1615–1616. [Google Scholar] [CrossRef] [PubMed]
  12. Park, S.; Hur, J.Y.; Lee, K.Y.; Lee, J.C.; Rho, J.K.; Shin, S.H.; Choi, C.-M. Assessment of EGFR mutation status using cell-free DNA from bronchoalveolar lavage fluid. Clin. Chem. Lab. Med. 2017, 55, 1489–1495. [Google Scholar] [CrossRef]
  13. Kiura, K.; Yoh, K.; Katakami, N.; Nogami, N.; Kasahara, K.; Takahashi, T.; Okamoto, I.; Cantarini, M.; Hodge, R.; Uchida, H. Osimertinib in patients with epidermal growth factor receptor T790M advanced non-small cell lung cancer selected using cytology samples. Cancer Sci. 2018, 109, 1177–1184. [Google Scholar] [CrossRef] [PubMed]
  14. Hur, J.Y.; Kim, H.J.; Lee, J.S.; Choi, C.-M.; Lee, J.C.; Jung, M.K.; Pack, C.G.; Lee, K.Y. Extracellular vesicle-derived DNA for performing EGFR genotyping of NSCLC patients. Mol. Cancer 2018, 17, 1–6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Gazdar, A.F.; Minna, J.D. Molecular detection of early lung cancer. J. Natl. Cancer Inst. 1999, 91, 299–301. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Ahrendt, S.A.; Chow, J.T.; Xu, L.-H.; Yang, S.C.; Eisenberger, C.F.; Esteller, M.; Herman, J.G.; Wu, L.; Decker, P.A.; Jen, J.; et al. Molecular detection of tumor cells in bronchoalveolar lavage fluid from patients with early stage lung cancer. J. Natl. Cancer Inst. 1999, 91, 332–339. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Oshita, F.; Nomura, I.; Yamada, K.; Kato, Y.; Tanaka, G.; Noda, K. Detection of K-ras mutations of bronchoalveolar lavage fluid cells aids the diagnosis of lung cancer in small pulmonary lesions. Clin. Cancer Res. 1999, 5, 617–620. [Google Scholar] [PubMed]
  18. Li, J.; Hu, Y.-M.; Wang, Y.; Tang, X.-P.; Shi, W.-L.; Du, Y.-J. Gene mutation analysis in non-small cell lung cancer patients using bronchoalveolar lavage fluid and tumor tissue as diagnostic markers. Int. J. Biol. Markers 2014, 29, 328–336. [Google Scholar] [CrossRef] [PubMed]
  19. Nakamichi, S.; Seike, M.; Miyanaga, A.; Chiba, M.; Matsuda, K.; Kobayashi, K.; Takahashi, A.; Takeuchi, S.; Minegishi, Y.; Kubota, K.; et al. RT-PCR for Detecting ALK Translocations in Cytology Samples from Lung Cancer Patients. Anticancer Res. 2017, 37, 3295. [Google Scholar] [PubMed]
  20. Kim, H.; Kwon, Y.M.; Kim, J.S.; Lee, H.; Park, J.-H.; Shim, Y.M.; Han, J.; Park, J.; Kim, D.-H. Tumor-specific methylation in bronchial lavage for the early detection of non-small-cell lung cancer. J. Clin. Oncol. 2004, 22, 2363–2370. [Google Scholar] [CrossRef]
  21. Destro, A.; Bianchi, P.; Alloisio, M.; Laghi, L.; di Gioia, S.; Malesci, A.; Cariboni, U.; Gribaudi, G.; Bulfamante, G.; Marchetti, A.; et al. K-ras and p16INK4A alterations in sputum of NSCLC patients and in heavy asymptomatic chronic smokers. Lung Cancer 2004, 44, 23–32. [Google Scholar] [CrossRef] [PubMed]
  22. Konno, S.; Morishita, Y.; Fukasawa, M.; Shu, Y.; Wang, D.; Tanaka, R.; Minami, Y.; Iijima, T.; Noguchi, M. Anthracotic index and DNA methylation status of sputum contents can be used for identifying the population at risk of lung carcinoma. Cancer Cytopathol. 2004, 102, 348–354. [Google Scholar] [CrossRef] [PubMed]
  23. Nikolaidis, G.; Raji, O.Y.; Markopoulou, S.; Gosney, J.R.; Bryan, J.; Warburton, C.; Walshaw, M.; Sheard, J.; Field, J.K.; Liloglou, T. DNA Methylation Biomarkers Offer Improved Diagnostic Efficiency in Lung Cancer. Cancer Res. 2012, 72, 5692–5701. [Google Scholar] [CrossRef] [Green Version]
  24. Dietrich, D.; Kneip, C.; Raji, O.; Liloglou, T.; Seegebarth, A.; Schlegel, T.; Flemming, N.; Rausch, S.; Distler, J.; Fleischhacker, M.; et al. Performance evaluation of the DNA methylation biomarker SHOX2 for the aid in diagnosis of lung cancer based on the analysis of bronchial aspirates. Int. J. Oncol. 2012, 40, 825–832. [Google Scholar] [CrossRef] [PubMed]
  25. Zhang, C.; Yu, W.; Wang, L.; Zhao, M.; Guo, Q.; Lv, S.; Hu, X.; Lou, J. DNA methylation analysis of the SHOX2 and RASSF1A panel in bronchoalveolar lavage fluid for lung cancer diagnosis. J. Cancer 2017, 8, 3585. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Um, S.-W.; Kim, Y.; Lee, B.B.; Kim, D.; Lee, K.-J.; Kim, H.K.; Han, J.; Kim, H.; Shim, Y.M.; Kim, D.-H. Genome-wide analysis of DNA methylation in bronchial washings. Clin. Epigenetics 2018, 10, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Li, L.; Ye, Z.; Yang, S.; Yang, H.; Jin, J.; Zhu, Y.; Tao, J.; Chen, S.; Xu, J.; Liu, Y.; et al. Diagnosis of pulmonary nodules by DNA methylation analysis in bronchoalveolar lavage fluids. Clin. Epigenetics 2021, 13, 185. [Google Scholar] [CrossRef]
  28. Tuo, L.; Sha, S.; Huayu, Z.; Du, K. P16INK4a gene promoter methylation as a biomarker for the diagnosis of non-small cell lung cancer: An updated meta-analysis. Thorac. Cancer 2018, 9, 1032–1040. [Google Scholar] [CrossRef]
  29. Molina-Pinelo, S.; Pastor, M.D.; Suarez, R.; Romero-Romero, B.; González De la Peña, M.; Salinas, A.; García-Carbonero, R.; De Miguel, M.J.; Rodríguez-Panadero, F.; Carnero, A.; et al. MicroRNA clusters: Dysregulation in lung adenocarcinoma and COPD. Eur. Respir. J. 2014, 43, 1740. [Google Scholar] [CrossRef] [PubMed]
  30. Li, J.; Chen, P.; Li, X.-Q.; Bao, Q.-L.; Dai, C.-H.; Ge, L.-P. Elevated levels of survivin and livin mRNA in bronchial aspirates as markers to support the diagnosis of lung cancer. Int. J. Cancer 2013, 132, 1098–1104. [Google Scholar] [CrossRef]
  31. Rehbein, G.; Schmidt, B.; Fleischhacker, M. Extracellular microRNAs in bronchoalveolar lavage samples from patients with lung diseases as predictors for lung cancer. Clin. Chim. Acta 2015, 450, 78–82. [Google Scholar] [CrossRef]
  32. Kim, J.E.; Eom, J.S.; Kim, W.-Y.; Jo, E.J.; Mok, J.; Lee, K.; Kim, K.U.; Park, H.-K.; Lee, M.K.; Kim, M.-H. Diagnostic value of microRNAs derived from exosomes in bronchoalveolar lavage fluid of early-stage lung adenocarcinoma: A pilot study. Thorac. Cancer 2018, 9, 911–915. [Google Scholar] [CrossRef]
  33. Kim, J.O.; Gazala, S.; Razzak, R.; Guo, L.; Ghosh, S.; Roa, W.H.; Bédard, E.L.R. Non-small Cell Lung Cancer Detection Using MicroRNA Expression Profiling of Bronchoalveolar Lavage Fluid and Sputum. Anticancer Res. 2015, 35, 1873. [Google Scholar] [PubMed]
  34. Li, H.; Jiang, Z.; Leng, Q.; Bai, F.; Wang, J.; Ding, X.; Li, Y.; Zhang, X.; Fang, H.; Yfantis, H.G.; et al. A prediction model for distinguishing lung squamous cell carcinoma from adenocarcinoma. Oncotarget 2017, 8, 50704–50714. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Mancuso, G.; Bovio, E.; Rena, O.; Rrapaj, E.; Mercalli, F.; Veggiani, C.; Paganotti, A.; Andorno, S.; Boldorini, R. Prognostic impact of a 3-MicroRNA signature in cytological samples of small cell lung cancer. Cancer Cytopathol. 2016, 124, 621–629. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Rodríguez, M.; Silva, J.; López-Alfonso, A.; López-Muñiz, M.B.; Peña, C.; Domínguez, G.; García, J.M.; López-Gónzalez, A.; Méndez, M.; Provencio, M.; et al. Different exosome cargo from plasma/bronchoalveolar lavage in non-small-cell lung cancer. Genes Chromosomes Cancer 2014, 53, 713–724. [Google Scholar] [CrossRef] [PubMed]
  37. Kuo, C.-H.S.; Liu, C.-Y.; Pavlidis, S.; Lo, Y.-L.; Wang, Y.-W.; Chen, C.-H.; Ko, H.-W.; Chung, F.-T.; Lin, T.-Y.; Wang, T.-Y.; et al. Unique Immune Gene Expression Patterns in Bronchoalveolar Lavage and Tumor Adjacent Non-Neoplastic Lung Tissue in Non-Small Cell Lung Cancer. Front. Immunol. 2018, 9, 232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Macchia, V.; Mariano, A.; Cavalcanti, M.; Coppa, A.; Cecere, C.; Fraioli, G.; Elia, S.; Ferrante, G. Tumor markers and lung cancer: Correlation between serum and bronchial secretion levels of CEA, TPA, CanAg CA-50, NSE and ferritin. Int. J. Biol. Markers 1987, 2, 151–156. [Google Scholar] [CrossRef]
  39. Edge, S.; Byrd, D.; Compton, C.; Fritz, A.; Greene, F.; Trotti, A. (Eds.) AJCC Cancer Staging Manual, 7th ed.; Springer: New York, NY, USA, 2010. [Google Scholar]
  40. Naumnik, W.; Naumnik, B.; Niewiarowska, K.; Ossolinska, M.; Chyczewska, E. Novel cytokines: IL-27, IL-29, IL-31 and IL-33. Can they be useful in clinical practice at the time diagnosis of lung cancer? Exp. Oncol. 2012, 34, 348–353. [Google Scholar]
  41. Naumnik, W.; Naumnik, B.; Niklińska, W.; Ossolińska, M.; Chyczewska, E. Clinical implications of hepatocyte growth factor, interleukin-20, and interleukin-22 in serum and bronchoalveolar fluid of patients with non-small cell lung cancer. In Advancements in Clinical Research; Pokorski, M., Ed.; Advances in Experimental Medicine and Biology; Springer: Cham, Switzerland, 2016; pp. 41–49. [Google Scholar]
  42. Kontakiotis, T.; Katsoulis, K.; Hagizisi, O.; Kougioulis, M.; Gerou, S.; Papakosta, D. Bronchoalveolar lavage fluid alteration in antioxidant and inflammatory status in lung cancer patients. Eur. J. Intern. Med. 2011, 22, 522–526. [Google Scholar] [CrossRef] [PubMed]
  43. Jakubowska, K.; Naumnik, W.; Niklińska, W.; Chyczewska, E. Clinical significance of HMGB-1 and TGF-β level in serum and BALF of advanced non-small cell lung cancer. In Respiratory Carcinogenesis; Pokorski, M., Ed.; Advances in Experimental Medicine and Biology; Springer: Cham, Switzerland, 2015; pp. 49–58. [Google Scholar]
  44. Chen, Z.; Xu, Z.; Sun, S.; Yu, Y.; Lv, D.; Cao, C.; Deng, Z. TGF-β1, IL-6 and TNF-α in Bronchoalveolar Lavage Fluid: Useful Markers for Lung Cancer? Sci. Rep. 2014, 4, 5595. [Google Scholar] [CrossRef] [Green Version]
  45. Xiong, W.; Ding, W.; Xu, M.; Pudasaini, B.; Sun, J.; Zhao, Y. The screening role of a biomarker panel in BALF among patients with cancer-suspected pulmonary nodules less than 8 mm. Clin. Respir. J. 2020, 14, 829–838. [Google Scholar] [CrossRef] [PubMed]
  46. Charpidou, A.; Gkiozos, I.; Konstantinou, M.; Eleftheraki, A.; Demertzis, P.; Harrington, K.; Polyzos, A.; Syrigos, K.N. Bronchial washing levels of vascular endothelial growth factor receptor-2 (VEGFR2) correlate with overall survival in NSCLC patients. Cancer Lett. 2011, 304, 144–153. [Google Scholar] [CrossRef] [PubMed]
  47. Cao, C.; Sun, S.F.; Lv, D.; Chen, Z.B.; Ding, Q.L.; Deng, Z.C. Utility of VEGF and sVEGFR-1 in bronchoalveolar lavage fluid for differential diagnosis of primary lung cancer. Asian Pac. J. Cancer Prev. 2013, 14, 2443–2446. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Chen, L.; Li, Q.; Zhou, X.-D.; Shi, Y.; Yang, L.; Xu, S.-L.; Chen, C.; Cui, Y.-H.; Zhang, X.; Bian, X.-W. Increased pro-angiogenic factors, infiltrating neutrophils and CD163+ macrophages in bronchoalveolar lavage fluid from lung cancer patients. Int. Immunopharmacol. 2014, 20, 74–80. [Google Scholar] [CrossRef]
  49. Pio, R.; Garcia, J.; Corrales, L.; Ajona, D.; Fleischhacker, M.; Pajares, M.J.; Cardenal, F.; Seijo, L.; Zulueta, J.J.; Nadal, E.; et al. Complement factor H is elevated in bronchoalveolar lavage fluid and sputum from patients with lung cancer. Cancer Epidemiol. Biomark. Prev. 2010, 19, 2665–2672. [Google Scholar] [CrossRef] [Green Version]
  50. Ajona, D.; Pajares, M.J.; Corrales, L.; Perez-Gracia, J.L.; Agorreta, J.; Lozano, M.D.; Torre, W.; Massion, P.P.; de-Torres, J.P.; Jantus-Lewintre, E.; et al. Investigation of Complement Activation Product C4d as a Diagnostic and Prognostic Biomarker for Lung Cancer. J. Natl. Cancer Inst. 2013, 105, 1385–1393. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Ajona, D.; Okrój, M.; Pajares, M.J.; Agorreta, J.; Lozano, M.D.; Zulueta, J.J.; Verri, C.; Roz, L.; Sozzi, G.; Pastorino, U.; et al. Complement C4d-specific antibodies for the diagnosis of lung cancer. Oncotarget 2018, 9, 6346–6355. [Google Scholar] [CrossRef] [Green Version]
  52. Li, Q.K.; Shah, P.; Li, Y.; Aiyetan, P.O.; Chen, J.; Yung, R.; Molena, D.; Gabrielson, E.; Askin, F.; Chan, D.W.; et al. Glycoproteomic analysis of bronchoalveolar lavage (BAL) fluid identifies tumor-associated glycoproteins from lung adenocarcinoma. J. Proteome Res. 2013, 12, 3689–3696. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Uribarri, M.; Hormaeche, I.; Zalacain, R.; Lopez-Vivanco, G.; Martinez, A.; Nagore, D.; Ruiz-Argüello, M.B. A new biomarker panel in bronchoalveolar lavage for an improved lung cancer diagnosis. J. Thorac. Oncol. 2014, 9, 1504–1512. [Google Scholar] [CrossRef] [Green Version]
  54. Ortea, I.; Rodríguez-Ariza, A.; Chicano-Gálvez, E.; Arenas Vacas, M.S.; Jurado Gámez, B. Discovery of potential protein biomarkers of lung adenocarcinoma in bronchoalveolar lavage fluid by SWATH MS data-independent acquisition and targeted data extraction. J. Proteom. 2016, 138, 106–114. [Google Scholar] [CrossRef] [PubMed]
  55. Almatroodi, S.A.; McDonald, C.F.; Collins, A.L.; Darby, I.A.; Pouniotis, D.S. Quantitative proteomics of bronchoalveolar lavage fluid in lung adenocarcinoma. Cancer Genom. Proteom. 2015, 12, 39–48. [Google Scholar]
  56. Carvalho, A.S.; Cuco, C.M.; Lavareda, C.; Miguel, F.; Ventura, M.; Almeida, S.; Pinto, P.; de Abreu, T.T.; Rodrigues, L.V.; Seixas, S.; et al. Bronchoalveolar Lavage Proteomics in Patients with Suspected Lung Cancer. Sci. Rep. 2017, 7, 42190. [Google Scholar] [CrossRef]
  57. Hmmier, A.; O’Brien, M.E.; Lynch, V.; Clynes, M.; Morgan, R.; Dowling, P. Proteomic analysis of bronchoalveolar lavage fluid (BALF) from lung cancer patients using label-free mass spectrometry. BBA Clin. 2017, 7, 97–104. [Google Scholar] [CrossRef] [PubMed]
  58. Liu, L.; Li, D.; Shu, J.; Wang, L.; Zhang, F.; Zhang, C.; Yu, H.; Chen, M.; Li, Z.; Guo, X. Protein Glycopatterns in Bronchoalveolar Lavage Fluid as Novel Potential Biomarkers for Diagnosis of Lung Cancer. Front. Oncol. 2021, 10, 568433. [Google Scholar] [CrossRef]
  59. Domagala-Kulawik, J.; Osinska, I.; Hoser, G.J. Mechanisms of immune response regulation in lung cancer. Transl. Lung Cancer 2014, 3, 15. [Google Scholar]
  60. Kwiecien, I.; Stelmaszczyk-Emmel, A.; Polubiec-Kownacka, M.; Dziedzic, D.; Domagala-Kulawik, J. Elevated regulatory T cells, surface and intracellular CTLA-4 expression and interleukin-17 in the lung cancer microenvironment in humans. Cancer Immunol. Immunother. 2017, 66, 161–170. [Google Scholar] [CrossRef] [Green Version]
  61. Zhang, B.; Yao, G.; Zhang, Y.; Gao, J.; Yang, B.; Rao, Z.; Gao, J. M2-polarized tumor-associated macrophages are associated with poor prognoses resulting from accelerated lymphangiogenesis in lung adenocarcinoma. Clinics 2011, 66, 1879–1886. [Google Scholar] [CrossRef] [Green Version]
  62. Osińska, I.; Stelmaszczyk-Emmel, A.; Polubiec-Kownacka, M.; Dziedzic, D.; Domagała-Kulawik, J. CD4+/CD25high/FoxP3+/CD127−regulatory T cells in bronchoalveolar lavage fluid of lung cancer patients. Hum. Immunol. 2016, 77, 912–915. [Google Scholar] [CrossRef]
  63. Hu, X.; Gu, Y.; Li, D.; Zhao, S.; Hua, S.; Jiang, Y. Analyzing the percentage of different PD-1+ T cell subsets in peripheral blood and bronchoalveolar lavage fluid of small cell lung cancer patients: A prospective study. Clin. Exp. Pharmacol. Physiol. 2019, 46, 1074–1083. [Google Scholar] [CrossRef] [PubMed]
  64. Hu, X.; Gu, Y.; Zhao, S.; Hua, S.; Jiang, Y. Immunotherapy. Increased IL-10+ CD206+ CD14+ M2-like macrophages in alveolar lavage fluid of patients with small cell lung cancer. Cancer Immunol. 2020, 69, 2547–2560. [Google Scholar] [CrossRef] [PubMed]
  65. Masuhiro, K.; Tamiya, M.; Fujimoto, K.; Koyama, S.; Naito, Y.; Osa, A.; Hirai, T.; Suzuki, H.; Okamoto, N.; Shiroyama, T.; et al. Bronchoalveolar lavage fluid reveals factors contributing to the efficacy of PD-1 blockade in lung cancer. JCI Insight 2022, 7, e157915. [Google Scholar] [CrossRef] [PubMed]
  66. Krebs, M.G.; Hou, J.-M.; Ward, T.H.; Blackhall, F.H.; Dive, C. Circulating tumour cells: Their utility in cancer management and predicting outcomes. Ther. Adv. Med. Oncol. 2010, 2, 351–365. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Chen, Q.; Ge, F.; Cui, W.; Wang, F.; Yang, Z.; Guo, Y.; Li, L.; Bremner, R.M.; Lin, P.P. Lung cancer circulating tumor cells isolated by the EpCAM-independent enrichment strategy correlate with Cytokeratin 19-derived CYFRA21-1 and pathological staging. Clin. Chim. Acta 2013, 419, 57–61. [Google Scholar] [CrossRef] [PubMed]
  68. Wang, P.-P.; Liu, S.-H.; Chen, C.-T.; Lv, L.; Li, D.; Liu, Q.-Y.; Liu, G.-L.; Wu, Y. Circulating tumor cells as a new predictive and prognostic factor in patients with small cell lung cancer. J. Cancer 2020, 11, 2113. [Google Scholar] [CrossRef]
  69. Yang, D.; Yang, X.; Li, Y.; Zhao, P.; Fu, R.; Ren, T.; Hu, P.; Wu, Y.; Yang, H.; Guo, N. Clinical significance of circulating tumor cells and metabolic signatures in lung cancer after surgical removal. J. Transl. Med. 2020, 18, 1–13. [Google Scholar] [CrossRef]
  70. Zhong, M.; Zhang, Y.; Pan, Z.; Wang, W.; Zhang, Y.; Weng, Y.; Huang, H.; He, Y.; Liu, O. Clinical Utility of Circulating Tumor Cells in the Early Detection of Lung Cancer in Patients with a Solitary Pulmonary Nodule. Technol. Cancer Res. Treat. 2021, 20, 15330338211041465. [Google Scholar] [CrossRef]
  71. Ciebiada, M.; Górski, P.; Antczak, A. Eicosanoids in Exhaled Breath Condensate and Bronchoalveolar Lavage Fluid of Patients with Primary Lung Cancer. Dis. Markers 2012, 32, 562862. [Google Scholar] [CrossRef]
  72. Schmid, S.; Kübler, M.; Korcan Ayata, C.; Lazar, Z.; Haager, B.; Hoßfeld, M.; Meyer, A.; Cicko, S.; Elze, M.; Wiesemann, S.; et al. Altered purinergic signaling in the tumor associated immunologic microenvironment in metastasized non-small-cell lung cancer. Lung Cancer 2015, 90, 516–521. [Google Scholar] [CrossRef]
  73. Callejón-Leblic, B.; García-Barrera, T.; Grávalos-Guzmán, J.; Pereira-Vega, A.; Gómez-Ariza, J.L. Metabolic profiling of potential lung cancer biomarkers using bronchoalveolar lavage fluid and the integrated direct infusion/ gas chromatography mass spectrometry platform. J. Proteom. 2016, 145, 197–206. [Google Scholar] [CrossRef] [Green Version]
  74. Lui, G.Y.; Kovacevic, Z.; Richardson, V.; Merlot, A.M.; Kalinowski, D.S.; Richardson, D.R. Targeting cancer by binding iron: Dissecting cellular signaling pathways. Oncotarget 2015, 6, 18748. [Google Scholar] [CrossRef] [Green Version]
  75. Callejon-Leblic, B.; Gómez-Ariza, J.L.; Pereira-Vega, A.; Garcia-Barrera, T. Metal dyshomeostasis based biomarkers of lung cancer using human biofluids. Metallomics 2018, 10, 1444–1451. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Wang, K.; Huang, Y.; Zhang, Z.; Liao, J.; Ding, Y.; Fang, X.; Liu, L.; Luo, J.; Kong, J. A preliminary study of microbiota diversity in saliva and bronchoalveolar lavage fluid from patients with primary bronchogenic carcinoma. Med. Sci. Monit. 2019, 25, 2819. [Google Scholar] [CrossRef] [PubMed]
  77. Cheng, C.; Wang, Z.; Wang, J.; Ding, C.; Sun, C.; Liu, P.; Xu, X.; Liu, Y.; Chen, B.; Gu, B. Characterization of the lung microbiome and exploration of potential bacterial biomarkers for lung cancer. Transl. Lung Cancer Res. 2020, 9, 693–704. [Google Scholar] [CrossRef] [PubMed]
  78. Lee, S.H.; Sung, J.Y.; Yong, D.; Chun, J.; Kim, S.Y.; Song, J.H.; Chung, K.S.; Kim, E.Y.; Jung, J.Y.; Kang, Y.A.; et al. Characterization of microbiome in bronchoalveolar lavage fluid of patients with lung cancer comparing with benign mass like lesions. Lung Cancer 2016, 102, 89–95. [Google Scholar] [CrossRef]
  79. Patnaik, S.K.; Cortes, E.G.; Kannisto, E.D.; Punnanitinont, A.; Dhillon, S.S.; Liu, S.; Yendamuri, S. Lower airway bacterial microbiome may influence recurrence after resection of early-stage non–small cell lung cancer. J. Thorac. Cardiovasc. Surg. 2021, 161, 419–429.e16. [Google Scholar] [CrossRef]
  80. Zheng, L.; Sun, R.; Zhu, Y.; Li, Z.; She, X.; Jian, X.; Yu, F.; Deng, X.; Sai, B.; Wang, L.; et al. Lung microbiome alterations in NSCLC patients. Sci. Rep. 2021, 11, 11736. [Google Scholar] [CrossRef]
  81. Jang, H.J.; Choi, J.Y.; Kim, K.; Yong, S.H.; Kim, Y.W.; Kim, S.Y.; Kim, E.Y.; Jung, J.Y.; Kang, Y.A.; Park, M.S.; et al. Relationship of the lung microbiome with PD-L1 expression and immunotherapy response in lung cancer. Respir. Res. 2021, 22, 322. [Google Scholar] [CrossRef]
  82. Espuela-Ortiz, A.; Lorenzo-Diaz, F.; Baez-Ortega, A.; Eng, C.; Hernandez-Pacheco, N.; Oh, S.S.; Lenoir, M.; Burchard, E.G.; Flores, C.; Pino-Yanes, M. Bacterial salivary microbiome associates with asthma among African American children and young adults. Pediatric Pulmonol. 2019, 54, 1948–1956. [Google Scholar] [CrossRef]
  83. Molyneaux, P.L.; Cox, M.J.; Wells, A.U.; Kim, H.C.; Ji, W.; Cookson, W.O.; Moffatt, M.F.; Kim, D.S.; Maher, T.M. Changes in the respiratory microbiome during acute exacerbations of idiopathic pulmonary fibrosis. Respir. Res. 2017, 18, 1–6. [Google Scholar] [CrossRef] [Green Version]
  84. Yu, D.P.; Cheng, X.; Liu, Z.D.; Xu, S.F. Comparative beneficiary effects of immunotherapy against chemotherapy in patients with advanced NSCLC: Meta-analysis and systematic review. Oncol. Lett. 2017, 14, 1568–1580. [Google Scholar] [CrossRef] [Green Version]
  85. Weber, J.S.; Kähler, K.C.; Hauschild, A. Management of immune-related adverse events and kinetics of response with ipilimumab. J. Clin. Oncol. 2012, 30, 2691–2697. [Google Scholar] [CrossRef] [PubMed]
  86. Kyi, C.; Hellmann, M.D.; Wolchok, J.D.; Chapman, P.B.; Postow, M.A. Opportunistic infections in patients treated with immunotherapy for cancer. J. Immunother. Cancer 2014, 2, 19. [Google Scholar] [CrossRef] [Green Version]
  87. Fujita, K.; Terashima, T.; Mio, T. Anti-PD1 antibody treatment and the development of acute pulmonary tuberculosis. J. Thorac. Oncol. 2016, 11, 2238–2240. [Google Scholar] [CrossRef] [PubMed]
  88. Jain, P.; Sandur, S.; Meli, Y.; Arroliga, A.C.; Stoller, J.K.; Mehta, A.C. Role of flexible bronchoscopy in immunocompromised patients with lung infiltrates. Chest 2004, 125, 712–722. [Google Scholar] [CrossRef] [PubMed]
  89. Hellmann, M.D.; Rizvi, N.A.; Goldman, J.W.; Gettinger, S.N.; Borghaei, H.; Brahmer, J.R.; Ready, N.E.; Gerber, D.E.; Chow, L.Q.; Juergens, R.A.; et al. Nivolumab plus ipilimumab as first-line treatment for advanced non-small-cell lung cancer (CheckMate 012): Results of an open-label, phase 1, multicohort study. Lancet Oncol. 2017, 18, 31–41. [Google Scholar] [CrossRef] [Green Version]
  90. Suresh, K.; Naidoo, J.; Lin, C.T.; Danoff, S. Immune checkpoint immunotherapy for non-small cell lung cancer: Benefits and pulmonary toxicities. Chest 2018, 154, 1416–1423. [Google Scholar] [CrossRef]
  91. Garon, E.B.; Rizvi, N.A.; Hui, R.; Leighl, N.; Balmanoukian, A.S.; Eder, J.P.; Patnaik, A.; Aggarwal, C.; Gubens, M.; Horn, L.; et al. Pembrolizumab for the treatment of non–small-cell lung cancer. N. Engl. J. Med. 2015, 372, 2018–2028. [Google Scholar] [CrossRef]
  92. Gettinger, S.N.; Horn, L.; Gandhi, L.; Spigel, D.R.; Antonia, S.J.; Rizvi, N.A.; Powderly, J.D.; Heist, R.S.; Carvajal, R.D.; Jackman, D.M.; et al. Overall survival and long-term safety of nivolumab (anti–programmed death 1 antibody, BMS-936558, ONO-4538) in patients with previously treated advanced non–small-cell lung cancer. J. Clin. Oncol. 2015, 33, 2004. [Google Scholar] [CrossRef]
  93. Delaunay, M.; Cadranel, J.; Lusque, A.; Meyer, N.; Gounant, V.; Moro-Sibilot, D.; Michot, J.-M.; Raimbourg, J.; Girard, N.; Guisier, F.; et al. Immune-checkpoint inhibitors associated with interstitial lung disease in cancer patients. Eur. Respir. J. 2017, 50, 1700050. [Google Scholar] [CrossRef]
  94. Suresh, K.; Naidoo, J.; Zhong, Q.; Xiong, Y.; Mammen, J.; de Flores, M.V.; Cappelli, L.; Balaji, A.; Palmer, T.; Forde, P.M.; et al. The alveolar immune cell landscape is dysregulated in checkpoint inhibitor pneumonitis. J. Clin. Investig. 2019, 129, 4305–4315. [Google Scholar] [CrossRef] [Green Version]
  95. Chopra, A.; Nautiyal, A.; Kalkanis, A.; Judson, M.A. Drug-induced sarcoidosis-like reactions. Chest 2018, 154, 664–677. [Google Scholar] [CrossRef] [PubMed]
  96. Gkiozos, I.; Kopitopoulou, A.; Kalkanis, A.; Vamvakaris, I.N.; Judson, M.A.; Syrigos, K.N. Sarcoidosis-like reactions induced by checkpoint inhibitors. J. Thorac. Oncol. 2018, 13, 1076–1082. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Costabel, U.; Hunninghake, G. ATS/ERS/WASOG statement on sarcoidosis. Sarcoidosis statement committee. American thoracic society. European respiratory society. World association for sarcoidosis and other granulomatous disorders. Eur. Respir. J. 1999, 14, 735–737. [Google Scholar] [CrossRef] [PubMed]
  98. Montaudié, H.; Pradelli, J.; Passeron, T.; Lacour, J.P.; Leroy, S. Pulmonary sarcoid-like granulomatosis induced by nivolumab. Br. J. Dermatol. 2017, 176, 1060–1063. [Google Scholar] [CrossRef]
  99. Zhao, L.; Wang, L.; Ji, W.; Wang, X.; Zhu, X.; Hayman, J.A.; Kalemkerian, G.P.; Yang, W.; Brenner, D.; Lawrence, T.S.; et al. Elevation of plasma TGF-β1 during radiation therapy predicts radiation-induced lung toxicity in patients with non-small-cell lung cancer: A combined analysis from Beijing and Michigan. Int. J. Radiat. Oncol. Biol. Phys. 2009, 74, 1385–1390. [Google Scholar] [CrossRef]
  100. Chen, Y.; Rubin, P.; Williams, J.; Hernady, E.; Smudzin, T.; Okunieff, P. Circulating IL-6 as a predictor of radiation pneumonitis. Int. J. Radiat. Oncol. Biol. Phys. 2001, 49, 641–648. [Google Scholar] [CrossRef]
  101. Park, K.-J.; Oh, Y.-T.; Kil, W.-J.; Park, W.; Kang, S.-H.; Chun, M. Bronchoalveolar lavage findings of radiation induced lung damage in rats. J. Radiat. Res. 2009, 50, 177–182. [Google Scholar] [CrossRef] [Green Version]
  102. Hong, J.-H.; Jung, S.-M.; Tsao, T.C.Y.; Wu, C.-J.; Lee, C.-Y.; Chen, F.-H.; Hsu, C.-H.; Mcbride, W.H.; Chiang, C.-S. Bronchoalveolar lavage and interstitial cells have different roles in radiation-induced lung injury. Int. J. Radiat. Biol. 2003, 79, 159–167. [Google Scholar] [CrossRef]
  103. Toma, C.L.; Serbescu, A.; Alexe, M.; Cervis, L.; Ionita, D.; Bogdan, M.A. The bronchoalveolar lavage pattern in radiation pneumonitis secondary to radiotherapy for breast cancer. Maedica 2010, 5, 250. [Google Scholar]
  104. Crohns, M.; Saarelainen, S.; Laine, S.; Poussa, T.; Alho, H.; Kellokumpu-Lehtinen, P. Cytokines in bronchoalveolar lavage fluid and serum of lung cancer patients during radiotherapy—Association of interleukin-8 and VEGF with survival. Cytokine 2010, 50, 30–36. [Google Scholar] [CrossRef]
  105. Yamagishi, T.; Kodaka, N.; Kurose, Y.; Watanabe, K.; Nakano, C.; Kishimoto, K.; Oshio, T.; Niitsuma, K.; Matsuse, H. Analysis of predictive parameters for the development of radiation-induced pneumonitis. Ann. Thorac. Med. 2017, 12, 252. [Google Scholar] [PubMed]
  106. Aso, S.; Navarro-Martin, A.; Castillo, R.; Padrones, S.; Castillo, E.; Montes, A.; Martínez, J.I.; Cubero, N.; López, R.; Rodríguez, L.; et al. Severity of radiation pneumonitis, from clinical, dosimetric and biological features: A pilot study. Radiat. Oncol. 2020, 15, 1–10. [Google Scholar] [CrossRef] [PubMed]
  107. Zarogoulidis, P.; Rapti, A.; Sardeli, C.; Chinelis, P.; Athanasiadou, A.; Paraskevaidou, K.; Kallianos, A.; Veletza, L.; Trakada, G.; Hohenforst-Schmidt, W.; et al. Re-biopsy after relapse of targeted therapy. T790M after epidermal growth factor mutation, where and why based on a case series. Respir. Med. Case Rep. 2017, 21, 171–175. [Google Scholar] [CrossRef] [PubMed]
  108. Kawamura, T.; Kenmotsu, H.; Taira, T.; Omori, S.; Nakashima, K.; Wakuda, K.; Ono, A.; Naito, T.; Murakami, H.; Mori, K.; et al. Rebiopsy for patients with non-small-cell lung cancer after epidermal growth factor receptor-tyrosine kinase inhibitor failure. Cancer Sci. 2016, 107, 1001–1005. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Example of a proteome from a bronchoalveolar lavage fluid sample in a lung cancer patient performed by two-dimensional gel electrophoresis and silver staining. Circled spots represent differentially expressed proteins between lung cancer and controls. Reprinted from [53] with permission from Elsevier.
Figure 1. Example of a proteome from a bronchoalveolar lavage fluid sample in a lung cancer patient performed by two-dimensional gel electrophoresis and silver staining. Circled spots represent differentially expressed proteins between lung cancer and controls. Reprinted from [53] with permission from Elsevier.
Diagnostics 12 02949 g001
Figure 2. Immunostaining for cell epitopes in macrophages from bronchoalveolar lavage fluid samples in a lung cancer patient and healthy control. Reprinted from [48] with permission from Elsevier.
Figure 2. Immunostaining for cell epitopes in macrophages from bronchoalveolar lavage fluid samples in a lung cancer patient and healthy control. Reprinted from [48] with permission from Elsevier.
Diagnostics 12 02949 g002
Figure 3. Typical mass spectra from lung cancer patients (a) and controls (b) by direct infusion triple quadrupole time-of-flight mass spectrometry in bronchoalveolar lavage fluid for the detection of metabolite expressions. Reprinted from [73] with permission from Elsevier.
Figure 3. Typical mass spectra from lung cancer patients (a) and controls (b) by direct infusion triple quadrupole time-of-flight mass spectrometry in bronchoalveolar lavage fluid for the detection of metabolite expressions. Reprinted from [73] with permission from Elsevier.
Diagnostics 12 02949 g003
Table 1. Overview of lung cancer biomarkers, their measurement methods in bronchoalveolar lavage fluid, their respective diagnostic/predictive roles, and accuracy results from studies included in the review.
Table 1. Overview of lung cancer biomarkers, their measurement methods in bronchoalveolar lavage fluid, their respective diagnostic/predictive roles, and accuracy results from studies included in the review.
StudyPatientsControlsProcessingDetectionBiomarkerRoleAccuracy
Genetic/epigenetic biomarkers
Lee 2020 [4]n = 73 (M 38)
Mean age: 65.3 ± 9.8 y
Type: AC 65, SQCC 7, other 1
Stage: I 20, II 10, III 10, IV 27
-Cell-free DNA from BALF supernatantDroplet digital allele-specific PCREGFR L858R mutationPrediction of tumor molecular profileAUC 0.96
Acc 95%
EGFR E19del mutationAUC 0.86
Acc 85%
Nair 2022 [7]n = 35 (M 18)
Age: 40–83 y
Type: AC 30, SQCC 2, NSCLC NOS 1, SCC 1, other 1
Stage: I 18, II 6, III 6, IV 5
n = 21 (M 12)
Age: 46–76 y
Cell-free DNA from BALF supernatantDeep sequencing11 gene feature classifierLC diagnosisAUC 0.84
Sens 69%
Spec 100%
n = 31-Any tumor variant detectedPrediction of tumor molecular profileAcc 81%
Roncarati 2020 [8]n = 91 (M 60)
Age: 47–85 y
Type: AC 41, SQCC 31, SCC 11, undefined 7
Stage: I 13, II 7, III 25, IV 43
n = 31 (M 21)
Age: 42–86 y
Cellular DNA/RNA from BALF cell pelletDroplet digital methylation-specific PCRCDH1 methylationLC diagnosisSens 64%
Spec 74%
DLC1 methylationSens 37%
Spec 94%
PRPH methylationSens 40%
Spec 100%
RASSF1A methylationSens 46%
Spec 100%
4-gene methylation panelAUC 0.93
Sens 97%
Spec 74%
-Next-generation sequencingALK fusionsPrediction of tumor molecular profileAcc 96%
BRAF V600E mutationAcc 100%
EGFR mutationsAcc 97%
ERBB2/HER2 mutationsAcc 100%
KRAS mutationsAcc 90%
MET mutationsAcc 100%
ROS1 fusionsAcc 100%
Kawahara 2015 [9]n = 42 (M 23)
Age: 42–84 y
Type: AC
-Cell-free DNA from BALF supernatantAllele-specific PCR/
FRET-PHFA
EGFR mutationsPrediction of tumor molecular profileAcc 47%
Yanev 2021 [10]n = 26 (M 13)
Mean age: 63.3 y
Type: AC
-Cell-free DNA from BALF supernatantAllele-specific PCREGFR mutationsPrediction of tumor molecular profileAcc 92%
Park 2017 [12]n = 20 (M 5)
Age: 43–77 y
Type: AC
Stage: I 1, II 1, III 1, IV 17
-Cell-free DNA from BALF supernatantPNA-mediated clamping PCR/ PNA clamping-assisted fluorescence melting curve analysisEGFR mutationsPrediction of tumor molecular profileAcc 92%
Prediction of treatment response (EGFR TKI)-
Hur 2018 [14]n = 23-EV DNA from BALF EV pelletPNA-mediated clamping PCREGFR mutationsPrediction of tumor molecular profileAcc 100%
Cell-free DNA from BALF supernatantAcc 71%
Ahrendt 1999 [16]n = 50
Type: AC 25, SQCC 23, other 2
Stage: I 28, II 15, III 7
-Cellular DNA from BALF cell pelletOligonucleotide plaque hybridizationp53 mutationsPrediction of tumor molecular profileAcc 39%
Allele-specific PCRKRAS mutationsAcc 50%
Methylation-specific PCRp16 methylationAcc 63%
Microsatellite fragment analysis15 microsatellite markersAcc 14%
Combined panelAcc 53%
Oshita 1999 [17]n = 20
Type: AC
n = 13Cellular DNA from BALF cell pelletAllele-specific PCRKRAS codon 12 mutationLC diagnosisSens 79%
Spec 64%
Li 2014 [18]n = 48 (M 27)
Age: 42–75 y
Type: AC 32, SQCC 14, LCC 2
Stage: I 19, II 25, III 4
n = 26 (M 17)
Age: 28–70 y
Cellular DNA from BALF cell pelletPCR single-strand conformation polymorphismKRAS mutationsLC diagnosisSens 38%
Spec 92%
Prediction of tumor molecular profileAcc 72%
p53 mutationsLC diagnosisSens 44%
Spec 96%
Prediction of tumor molecular profileAcc 75%
Combined panelLC diagnosisSens 67%
Spec 89%
Prediction of tumor molecular profileAcc 70%
Nakamichi 2017 [19]n = 36-Cellular RNA from BALF cell pelletmRNA-specific reverse transcription-PCRALK translocationsPrediction of tumor molecular profileAcc 97%
Kim 2004 [20]n = 85 (M 57)
Mean age: 65 ± 17 y
Type: AC 31, SQCC 43, other 11
Stage: I 52, II 33
n = 127 (M 84)
Mean age: 62 ± 14 y
Cellular DNA from BALF cell pelletMethylation-specific PCRp16 methylationLC diagnosisSens 16%
Spec 94%
RARβ methylationSens 15%
Spec 87%
H-cadherin methylationSens 13%
Spec 97%
RASSF1A methylationSens 18%
Spec 96%
Nikolaidis 2012 [23]n = 139 (M 80)
Mean age: 68.4 ± 8.1 y
Type: AC 22, SQCC 31, SCC 39, LCC 16, other 20, unknown 11
n = 109 (M 63)
Mean age: 67.6 ± 8.8 y
Cellular DNA from BALF cell pelletMethylation-specific PCRMethylation panel (p16, RASSF1, WT1, TERT)LC diagnosisSens 82%
Spec 91%
Dietrich 2012 [24]n = 125 (M 72)
Age: 46–85 y
Type: AC 26, SQCC 28, NSCLC NOS 9, SCC 40, other 22
n = 125 (M 61)
Age: 45–86 y
Cellular DNA from BALF cell pelletMethylation-specific PCRSHOX2 methylationLC diagnosisAUC 0.94
Sens 78%
Spec 96%
Zhang 2017 [25]n = 284 (M 212)
Age: 31–85
Type: AC 92, SQCC 107, SCC 42, LCC 5, unknown 38
Stage: I 28, II 30, III 133, IV 93
n = 38 (M 28)
Age: 29–75 y
Cellular DNA from BALF cell pelletMethylation-specific PCRMethylation panel (SHOX2, RASSF1A)LC diagnosisAUC 0.89
Sens 81%
Spec 97%
Um 2018 [26]n = 31n = 10Cellular DNA from BALF cell pelletDNA methylation microarrayMethylation panel (TFAP2A, TBX15,
PHF11, TOX2, PRR15, PDGFRA, HOXA11)
LC diagnosisAUC 0.87
Sens 87%
Spec 83%
Li 2021 [27]n = 52 (M 33)
Type: AC 37, SQCC 10, other 5
Stage: I 34, II 1, III 1, IV 4, unknown 12
n = 59 (M 33)Cellular DNA from BALF cell pelletMethylation-specific PCRMethylation panel (LHX9, GHSR, HOXA11,
PTGER4-2, HOXB4-3)
LC diagnosisAUC 0.82
Sens 70%
Spec 82%
Post-transcriptional biomarkers
Molina-Pinelo 2014 [29]n = 48 (M 40)
Type: AC
Stage: I-II 2, III 15, IV 31
n = 16 (M 15)Cellular RNA from BALF cell pelletMicroRNA microfluidic card arrayFour upregulated miRNA clusters (chromosome loci 13q31.3, 7q22.1, Xq26.2, 11q13.1)LC diagnosis-
Li 2013 [30]n = 70 (M 52)
Mean age: 64 ± 24 y
Type: AC 37, SQCC 25, LCC 4, SCC 4
Stage: I 10, II 24, III 25, IV 11
n = 26 (M 19)
Mean age: 55 ± 19 y
Cellular RNA from BALF cell pelletmRNA-specific reverse transcription-PCRSurvivin expression ratio > 0.35LC diagnosisAUC 0.83
Sens 83%
Spec 96%
Livin expression ratio > 0.3AUC 0.68
Sens 63%
Spec 92%
Rehbein 2015 [31]n = 30 (M 21)
Median age 64.5 y
n = 30 (M 17)
Median age 63.5 y
Cell-free RNA from BALF supernatantMicroRNA microfluidic card arrayFive upregulated miRNAs (U6 snRNA, hsa-miR 1285, hsa-miR 1303, hsa-miR 29a-5p, hsa-miR 650)LC diagnosis-
Kim 2018 [32]n = 13 (M 7)
Age: 47–72 y
Type: AC
Stage: I 10, II 3
n = 15EV RNA from BALF EV pelletmiRNA-specific reverse transcription-PCRmiR-126 and Let-7a were significantly upregulatedLC diagnosis-
Kim 2015 [33]n = 21 (M 17)
Age: 46–84 y
Type: AC 13, SQCC 5, LCC 3
Stage: I 12, II 9
n = 10 (M 8)
Age: 30–77 y
Cellular RNA from BALF cell pelletmiRNA-specific reverse transcription-PCRHigh expression cluster of a 5-miRNA panel (miR-21, miR-143, miR-155, miR-210, miR-372) LC diagnosisSens 86%
Spec 100%
Li 2017 [34]n = 127 (M 82)
Age: 66 ± 8 y
Type: AC 45, SQCC 82
Stage: I 52, II 38, III-IV 37
-Cellular RNA from BALF cell pelletDroplet digital miRNA-specific PCR2-miRNA (miR-205-5p, miR-944) prediction modelDiscrimination of SQCC from ACAUC 0.997
Sens 95%
Spec 97%
Mancuso 2016 [35]n = 50 (M 32)
Age: 34–82 y
Type: SCC
Stage: III 18, IV 32
-Cellular RNA from BALF cell pelletmiRNA-specific reverse transcription-PCRAbove median expression levels of a 3-miRNA panel (miR-192, miR-200c, miR-205)Overall survival (worse)-
Rodríguez 2014 [36]n = 30 (M 23)
Age: 45–83 y
Type: AC 14, SQCC 16
n = 75 (M 46)
Age: 18–87 y
EV RNA from BALF EV pelletMicroRNA real-time PCR array10 miRNAs were upregulated and 10 downregulatedLC diagnosis-
Kuo 2018 [37]n = 34 (M 19)
Mean age: 58.5 ± 12.8 y
Type: AC 26, SQCC 8
Stage: III 11, IV 23
n = 14 (M 7)
Mean age: 53.3 ± 11.4 y
Cellular RNA from BALF cell pelletmRNA-specific reverse transcription-PCR9-gene (SPP1, CEACAM6, MMP7, SLC40A1, IGJ, IGKC, CPA3, YES1, CXCL13) prediction modelLC diagnosisAUC 0.92
Post-translational biomarkers (proteins, cell epitopes, metabolites)
Macchia 1987 [38]n = 37
Type: AC 4, SQCC 23, LCC 3, SCC 7
n = 20Cell-free BALF supernatantRIACEALC diagnosisSens 57%
Spec 65%
TPASens 65%
Spec 20%
NSESCC diagnosisSens 71%
Spec 90%
FerritinSens 71%
Spec 100%
CanAg CA-50Sens 100%
Spec 55%
Naumnik 2012 [40]n = 45 (M 38)
Mean age: 61.9 ± 4 y
Type: AC 9, SQCC 22, NSCLC NOS 14
Stage: III 18, IV 27
n = 15 (M 13)
Mean age: 60.1 ± 5 y
Cell-free BALF supernatantELISAIL-27 (↑)LC diagnosis-
IL-27, IL-29 (↓)Discrimination of advanced stage-
Naumnik 2016 [41]n = 46 (M 46)
Mean age: 63 ± 3 y
Type: AC 10, SQCC 25, LCC 11
Stage: III 20, IV 26
n = 15 (M 12)
Mean age 60 ± 4 y
Cell-free BALF supernatantELISAHGF, IL-22 (↓)LC diagnosis-
IL-22 (↑)Overall survival (worse)-
Kontakiotis 2011 [42]n = 42 (M 42)
Age: 43–80 y
Type: AC 7, SQCC 22, SCC 10, other 3
n = 16 (M 16)
Age: 45–77 y
Cell-free BALF supernatantELISATNF-α (↑)LC diagnosis-
Colorimetric assayTotal antioxidants, glutathione (↑)-
Jakubowska 2015 [43]n = 45 (M 38)
Mean age: 61.7 ± 8.3 y
Type: AC 20, SQCC 22, LCC 3
Stage: III 18, IV 27
n = 15 (M 13)
Mean age: 60.1 ± 5.0 y
Cell-free BALF supernatantELISATGF-β (↑)LC diagnosis-
Chen 2014 [44]n = 45 (M 28)
Mean age: 60.8 ± 1.2 y
Type: AC 11, SQCC 18, SCC 10, other 6
n = 33 (M 19)
Mean age: 58.2 ± 1.7 y
Cell-free BALF supernatantELISATGF-β1 >10.85 pg/mlLC diagnosisAUC 0.7
Sens 62%
Spec 61%
Xiong 2020 [45]n = 219 (M 150)
Mean age: 68.4 ± 18.8 y
Type: AC 136, SQCC 43, SCC 35, other 5
Stage: 0 38, I 93, II 50, III 28, IV 10
n = 186 (M 125)
Mean age: 40.6 ± 15.5 y
Cell-free BALF supernatantELISAVEGF >234.1 pg/mL, TGF-β >81.8 pg/mL, HGF 44.6 pg/mL (at least 2 positive)
LC diagnosisAUC 0.81
Sens 82%
Spec 61%
Charpidou 2011 [46]n = 40 (M 37)
Age: 45–82 y
Type: AC 12, SQCC 19, other 9
Stage: I 3, III 14, IV 23
-Cell-free BALF supernatantELISAVEGF (↓)Prediction of treatment response (chemotherapy) -
VEGFR1 >53.2 pg/mlProgression free survival (worse)-
VEGFR2 >705.3 pg/mlOverall survival (worse)-
Cao 2013 [47]n = 37 (M 28)
Mean age: 55.4 ± 8.4 y
Type: AC 15, SQCC 19, SCC 3
Stage: I 23, II 9, III 5
n = 19 (M 12)
Mean age: 48.1 ± 9.2 y
Cell-free BALF supernatantELISAVEGF >214 pg/mlLC diagnosisAUC 0.86
Sens 82%
Spec 84%
Chen 2014 [48]n = 54 (M 60)
Median age: 60 y
Type: AC 9, SQCC 36, SCC 9
n = 12 (M 6)
Median age: 37 y
Cell-free BALF supernatantELISAIL-8, VEGF (↑)LC diagnosis-
Pio 2010 [49]n = 56 (M 45)
Age: 38–83 y
Type: AC 12, SQCC 24, LCC 4, SCC 9, other 7
n = 22 (M 14)
Age: 30–82 y
Cell-free BALF supernatantELISA Complement factor H >1 μg/mLLC diagnosisSens 62%
Spec 77%
Albumin >17 μg/mLSens 68%
Spec 71%
Ajona 2013 [50]n = 50 (M 41)
Type: AC 12, SQCC 22, SCC 9, other 7
n = 22 (M 14)Cell-free BALF supernatantELISAComplement C4-derived fragmentsLC diagnosisAUC 0.73
Ajona 2018 [51]n = 49 (M 40)
Type: AC 12, SQCC 22, SCC 8, other 7
n = 22 (M 14)Cell-free BALF supernatantELISAComplement C4dLC diagnosisAUC 0.80
Li 2013 [52]n = 18 (M 7)
Age: 51–83 y
Type: AC
n = 6 (M 3)
Age: 18–85 y
Cell-free proteins from BALF supernatantELISANapsin A >55 ng/mg total proteinLC diagnosisAUC 0.85
Sens 84%
Spec 67%
Uribarri 2014 [53]n = 204 (M 177)
Mean age: 63.0 ± 10.7 y
Type: AC 59, SQCC 80, SCC 63, other 2
Stage: I 14, II 4, III 60, IV 109, undefined 17
n = 48 (M 38)
Mean age: 54.9 ± 14.0 y
Cell-free proteins from BALF supernatantFluorescent bead-based immunoassay5-protein (APOA1, CO4A, CRP, GSTP1, SAMP) prediction modelLC diagnosisAUC 0.94
Sens 95%
Spec 81%
2-protein (STMN1, GSTP1) prediction modelDiscrimination of SCC from NSCLCAUC 0.80
Sens 90%
Spec 57%
Ortea 2016 [54]n = 12 (M 8)
Median age: 64 y
Type: AC
Stage: I-II 2, III-IV 10
n = 10 (M 10)
Median age: 61
Cell-free proteins from BALF supernatantLiquid chromatography–mass spectrometryDiscriminant analysis of a 44-protein panelLC diagnosisSens 92%
Spec 70%
Almatroodi 2015 [55]n = 8 (M 5)
Mean age: 68.1 ± 7.6 y
Type: AC
Stage: I 2, II 2, III 1, IV 3
n = 8 (M 3)
Mean age: 60 ± 8.7 y
Cellular proteins from BALF cell pelletsLiquid chromatography–mass spectrometry33 upregulated proteinsLC diagnosis-
Carvalho 2017 [56]n = 49
Type: AC 28, SQCC 10, SCC 4, LCC 1, other 6
n = 41Cell-free proteins from BALF supernatantLiquid chromatography–mass spectrometryDifferent spectral count values from all abundant proteinsLC diagnosis-
133 differentially expressed proteins-
Hmmier 2017 [57]n = 26 (M 13)
Mean age: 65 y
Type: AC 13, SQCC 13
Stage: I-II 15, III-IV 11
n = 16 (M 8)
Mean age: 56 y
Cell-free proteins from BALF supernatantLiquid chromatography–mass spectrometry267 differentially expressed proteinsAC diagnosis-
261 differentially expressed proteinsSQCC diagnosis-
292 differentially expressed proteinsDiscrimination of SQCC from AC-
Liu 2021 [58]n = 85 (M 60)
Type: AC 32, SQCC 32, SCC 21
Stage: I-II 30, III-IV 42, unknown 13
n = 33 (M 20)Cell-free proteins from BALF supernatantLectin microarray3-lectin (ECA, GSL-I, RCA120) prediction modelLC diagnosisAUC 0.96
Sens 92%
Spec 94%
4-lectin (DBA, STL, UEA-I, BPL) prediction modelDiscrimination of AC from other subtypesAUC 0.62
Sens 71%
Spec 59%
1-lectin (PNA) prediction modelDiscrimination of AC from other subtypesAUC 0.69
Sens 80%
Spec 67%
6-lectin (STL, BS-I, PTL-II, SBA, PSA, GNA) prediction modelDiscrimination of AC from other subtypesAUC 0.72
Sens 72%
Spec 68%
6-lectin (MAL-II, LTL, GSL-I, RCA120, PTL-II, PWM) prediction modelDiscrimination of early from advanced stageAUC 0.86
Sens 83%
Spec 81%
Kwiecien 2017 [60]n = 18 (M 12)
Age: 50–81 y
Type: AC 4, SQCC 9, NSCLC NOS 4
Stage: I 4, II 11, III 3
-Immune cells from BALF cell pelletsAntibody-specific flow cytometry% Tregs, CTLA-4+ Tregs (↑)LC diagnosis (affected vs. healthy lung)-
Hu 2019 [63]n = 52 (M 29)
Age: 39–73 y
Type: NSCLC 26, SCC 26
n = 20 (M 12)
Age: 35–75 y
Immune cells from BALF cell pelletsAntibody-specific flow cytometry% PD-1+ Tph (↓), PD-1+ Tfh/Tph (↑)SCC diagnosis-
Hu 2020 [64]n = 67 (M 46)
Age: 39–75 y
Type: AC 18, SQCC 17, SCC 32
Stage: 0–IIIA 39, IIIB-IV 28
n = 14 (M 10)
Age: 33–71 y
Immune cells from BALF cell pelletsAntibody-specific flow cytometryTregs (↑)LC diagnosis
Discrimination of SCC from NSCLC
Discrimination of advanced SCC
-
IL-10+ CD206+ CD14+ M2-like macrophages (↑)LC diagnosis
Discrimination of SCC from NSCLC
Discrimination of advanced SCC
Overall survival (worse)
-
Cell-free BALF supernatantCytometric bead
array
IL-10 (↑)LC diagnosis
Discrimination of SCC from NSCLC
Discrimination of advanced SCC
Overall survival (worse)
-
Masuhiro 2022 [65]n = 12 (M 9)
Age: 55–70 y
Type: AC 7
-Cell-free BALF supernatantCytometric
bead array
CXCL9 (↑)Prediction of treatment response (immunotherapy)-
Bacterial DNA from BALF supernatant16S rRNA sequencingBacterial alpha diversity (↑), Proteobacteria (↓), Bacteroidetes (↑)-
n = 7Cellular RNA from BALF cell pelletsRNA sequencing87 genes were upregulated and 28 were downregulated-
Zhong 2021 [70]n = 12n = 6Tumor cells from BALF cell pelletsAntibody-specific immunostaining + fluorescence in situ hybridizationCirculating tumor cell count ≥2LC diagnosisSens 75%
Spec 100%
Schmid 2015 [72]n = 26 (M 16)
Mean age: 60.2 ± 8.3 y
Type: AC 20, SQCC 6
n = 21 (M 13)
Mean age: 64.7 ± 8.4 y
Cell-free BALF supernatantBLEIAATP, ADP (↑)LC diagnosis-
Cellular RNA from BALF cell pelletsmRNA-specific reverse transcription-PCRCD39 (↑)LC diagnosis
Discrimination of metastatic disease
-
P2X4, P2X7, P2Y1 (↑)Discrimination of metastatic disease-
Callejón-Leblic 2016 [73]n = 24 (M 16)
Mean age: 66 ± 11 y
n = 31 (M 23)
Mean age: 56 ± 13 y
Cell-free metabolites from BALF supernatantDirect infusion mass spectrometryCarnitine LC diagnosisAUC 0.88
AdenineAUC 0.83
CholineAUC 0.78
Gas chromatography–mass spectrometryGlycerolAUC 0.89
Phosphoric acidAUC 0.79
Callejón-Leblic 2018 [75]n = 24 (M 20)
Mean age: 65 ± 13 y
Type: NSCLC 22, SCC 2
n = 31 (M 27)
Mean age: 54 ± 14 y
Cell-free elements from BALF supernatantInductive coupled plasma mass spectrometryMnLC diagnosisAUC 0.75
V/Cu ratioAUC 0.76
Suresh 2019 [94]n = 18 (M 13)
Type: NSCLC 15, other 3
-Immune cells from BALF cell pelletsAntibody-specific flow cytometry% PD-1hi/CTLA-4hi Tregs (↓), % IL-1RA-expressing B cells (↓), % central memory T cells (↑), % CD8+ TNF-αhi T cells (↑), % IL-1βhi monocytes (↑)Prediction of CIP development-
Cell-free BALF supernatantV-plex immunoassaysIL-1β (↓), IL-8 (↓), MIP-3α (↓), IL-12p40 (↑), IP-10 (↑)-
Crohns 2010 [104]n = 36 (M 29)
Age: 47–82 y
Type: AC 1, SQCC 33, SCC 2
Stage: I 1, III 18, IV 17
n = 36 (M 16)
Age: 18–75 y
Cell-free BALF supernatantELISAIl-6 (↑)LC diagnosis-
IL-8 (↑)Overall survival (worse)-
Yamagishi 2017 [105]n = 22 (M 16)
Type: AC 8, SQCC 8, SCC 4, unknown 2
-Cell-free BALF supernatantELISAMMP-9 (↑)Prediction of radiation pneumonitis-
VEGF (↓)-
Metagenomic biomarkers
Wang 2019 [76]n = 51 (M 31)
Type: AC 18, SQCC 19, SCC 14
n = 15 (M 8)Bacterial DNA from BALF cell pellet16S rRNA sequencingMicrobial diversity (↓)LC diagnosis-
TreponemaAUC 0.86
Cheng 2020 [77]n = 32 (M 23)
Mean age: 64.3 ± 8.4 y
Type: AC 16, SQCC 9, SCC 7
Stage: I 7, III 6, IV 19
n = 22 (M 12)
Mean age: 56.5 ± 14.3 y
Bacterial DNA from BALF supernatant16S rRNA sequencing10-genera (f:Pseudomonadaceae, Capnocytophaga, Stenotrophomonas, Microbacterium, Gemmiger, c:TM7-3, Oscillospira, Blautia, Lautropia, Sediminibacterium) prediction modelLC diagnosisAUC 0.79
Lee 2016 [78]n = 20 (M 13)
Median age: 64 y
Type: AC 13, SQCC 5, SCC 2
Stage: II 6, III 8, IV 6
n = 8 (M 7)
Median age: 58.5 y
Bacterial DNA from BALF supernatant16S rRNA pyrosequencingVeillonellaLC diagnosisAUC 0.86
MegasphaeraAUC 0.78
Combined panelAUC 0.89
Patnaik 2021 [79]n = 36 (M 16)
Type: AC 24, SQCC 11, other 1
Stage: I
-Bacterial DNA from BALF supernatant16S rRNA sequencing19-genera microbiome signaturePrediction of recurrence after surgeryAUC 0.77
Zheng 2021 [80]n = 32
Type: NSCLC
Stage: I-II 23, III-IV 9
n = 15Bacterial DNA from BALF supernatant16S rRNA sequencingDifferentiated abundance of 19 speciesLC diagnosis-
Jang 2021 [81]n = 11 (M 9)
Median age: 63 y
Type: AC 8, SQCC 3
Stage: III 5, IV 6
-Bacterial DNA from BALF supernatant16S rRNA sequencingHaemophilus influenzae (↓), Neisseria perflava (↓), Veillonella dispar (↑)Prediction of treatment response (immunotherapy)-
BALF, bronchoalveolar lavage fluid; M, male; AC, adenocarcinoma; SQCC, squamous cell carcinoma; NSCLC; non-small-cell lung cancer; NOS, not otherwise specified; SCC, small-cell carcinoma; LCC, large cell carcinoma; EV, extracellular vesicle; PCR, polymerase chain reaction; FRET-PHFA, fluorescence resonance energy transfer-based preferential homoduplex formation assay; PNA, peptide nucleic acid; RIA, radioimmunoassay; ELISA, enzyme-linked immunosorbent assay; BLEIA, bioluminescent enzyme immunoassay; LC, lung cancer; TKI, tyrosine kinase inhibitor; CIP, checkpoint inhibitor pneumonitis; AUC, area under the ROC curve; Acc, overall accuracy; Sens, sensitivity; Spec, specificity. Note: Upward arrows correspond to increased and downward arrows to decreased detection of the biomarker respectively to predict each outcome. Sensitivity is the probability that a test result will be positive when the disease is present. Specificity is the probability that a test result will be negative when the disease is not present. Overall accuracy is the overall probability that a patient is correctly classified. Area under the ROC curve is the probability that a classifier will distinguish between two classes (disease vs. normal).
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kalkanis, A.; Papadopoulos, D.; Testelmans, D.; Kopitopoulou, A.; Boeykens, E.; Wauters, E. Bronchoalveolar Lavage Fluid-Isolated Biomarkers for the Diagnostic and Prognostic Assessment of Lung Cancer. Diagnostics 2022, 12, 2949. https://doi.org/10.3390/diagnostics12122949

AMA Style

Kalkanis A, Papadopoulos D, Testelmans D, Kopitopoulou A, Boeykens E, Wauters E. Bronchoalveolar Lavage Fluid-Isolated Biomarkers for the Diagnostic and Prognostic Assessment of Lung Cancer. Diagnostics. 2022; 12(12):2949. https://doi.org/10.3390/diagnostics12122949

Chicago/Turabian Style

Kalkanis, Alexandros, Dimitrios Papadopoulos, Dries Testelmans, Alexandra Kopitopoulou, Eva Boeykens, and Els Wauters. 2022. "Bronchoalveolar Lavage Fluid-Isolated Biomarkers for the Diagnostic and Prognostic Assessment of Lung Cancer" Diagnostics 12, no. 12: 2949. https://doi.org/10.3390/diagnostics12122949

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop