Prognostic and predictive significance of soluble programmed death ligand 1 in bronchoalveolar lavage fluid in stage IV non-small cell lung cancer
Original Article

Prognostic and predictive significance of soluble programmed death ligand 1 in bronchoalveolar lavage fluid in stage IV non-small cell lung cancer

So-yun Kim1#, Dongil Park1#, Pureum Sun2, Nayoung Kim3, Dahye Lee2, Duk Ki Kim1, Song-I Lee1, Jeong Eun Lee1, Chaeuk Chung1, Da Hyun Kang1 ORCID logo

1Department of Internal Medicine, College of Medicine, Chungnam National University, Daejeon, Korea; 2Institute for Medical Sciences, College of Medicine, Chungnam National University, Daejeon, Korea; 3Cancer Research Institute, Chungnam National University, Daejeon, Korea

Contributions: (I) Conception and design: DH Kang, C Chung; (II) Administrative support: SY Kim, DH Kang, C Chung; (III) Provision of study materials or patients: SY Kim, D Park, DK Kim, SI Lee, JE Lee; (IV) Collection and assembly of data: P Sun, N Kim, D Lee; (V) Data analysis and interpretation: SY Kim, D Park, DH Kang, C Chung; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

#These authors contributed equally to this work.

Correspondence to: Da Hyun Kang, MD, PhD; Chaeuk Chung, MD, PhD. Department of Internal Medicine, College of Medicine, Chungnam National University, 282 Munhwa-ro, Jung-gu, Daejeon, Korea. Email: ibelieveu113@naver.com or ibelieveu113@cnuh.co.kr; universe7903@gmail.com or cuchung@cnu.ac.kr.

Background: Patients with non-small cell lung cancer (NSCLC) have been shown to exhibit elevated levels of soluble programmed death-ligand 1 (sPD-L1) in the blood, associated with poor survival in NSCLC. The bronchoalveolar lavage fluid (BALF) composition reflects the tumor microenvironment of lung cancer. In this study, we investigated sPD-L1 levels in BALF and its role as a prognostic and predictive marker in patients with stage IV NSCLC.

Methods: We prospectively obtained BALF from lung cancer patients who underwent bronchoscopy between January 2020 and September 2022 at Chungnam National University Hospital (CNUH). Finally, 94 NSCLC stage IV patients were included in this study. Soluble PD-L1 levels in BALF were measured using a human PD-L1 Quantikine ELISA kit.

Results: The correlation between PD-L1 expression in tumor cells and sPD-L1 in BALF was weakly positive (rho =0.314, P=0.002). The median overall survival (OS) of the low sPD-L1 in BALF group was 16.47 months [95% confidence interval (CI): 11.15–21.79 months], which is significantly longer than 8.87 months (95% CI: 0.0–19.88 months, P=0.001) in the high sPD-L1 in BALF group. In 64 patients treated with or without immune checkpoint inhibitors (ICIs), sPD-L1 in BALF was significantly associated with progression-free survival (PFS) and OS. In the subgroup analysis of 31 patients treated with ICI, the objective response rate (ORR) in the low sPD-L1 BALF group was significantly higher than in high sPD-L1 in BALF group (ORR: 60.9% vs. 12.5%, P=0.02).

Conclusions: Soluble PD-L1 in BALF is a potential prognostic indicator for patients with stage IV NSCLC and a predictive marker for ICI treatment response.

Keywords: Non-small cell lung cancer (NSCLC); soluble programmed death-ligand 1 (sPD-L1); biomarker; bronchoalveolar lavage fluid (BALF)


Submitted May 02, 2024. Accepted for publication Jul 10, 2024. Published online Aug 23, 2024.

doi: 10.21037/tlcr-24-392


Highlight box

Key findings

• Soluble programmed death-ligand 1 (sPD-L1) in bronchoalveolar lavage fluid (BALF) was a significant prognostic factor affecting both progression-free survival (PFS) and overall survival (OS) in patients with non-small cell lung cancer (NSCLC) who received treatment. sPD-L1 in BALF can serve as a predictive and prognostic marker for the efficacy and outcomes of immune checkpoint inhibitor (ICI) treatment.

What is known and what is new?

• High levels of sPD-L1 in the blood are associated with poor survival and response to ICI treatment in lung cancer.

• We demonstrated a positive correlation between programmed death-ligand 1 (PD-L1) expression in tumor cells and sPD-L1 levels in BALF. Patients with high sPD-L1 in BALF showed a significantly lower response to ICI treatment.

What is the implication, and what should change now?

• We demonstrated the potential of sPD-L1 in BALF as a promising biomarker in NSCLC prognosis and ICI efficacy. Our results suggest that ICI combination therapy using cytotoxic agents may be better than ICI monotherapy when the levels of both PD-L1 in tumor cells and sPD-L1 in BALF are high.


Introduction

Although the development of various drugs, such as targeted agents and immune checkpoint inhibitors (ICIs), have contributed to the survival rate improvement of patients with advanced-stage cancer, lung cancer remains the leading cause of cancer-related mortality worldwide (1). The evolution of various anticancer agents has resulted in the development of more diverse treatment options for patients with lung cancer; however, treatment responses vary widely. Numerous studies on the prognosis of lung cancer and predicting the efficacy of existing treatments have been conducted. In patients receiving ICIs, which have recently become the standard treatment for advanced lung cancer, programmed death ligand 1 (PD-L1) expression in tumor cells is currently the only predictive marker used in clinical practice (2-4).

PD-L1 is a transmembrane protein that binds to its receptor programmed death-1 (PD-1) expressed by T cells and other immune cells (5,6). The interaction between PD-L1 and PD-1 suppresses T cell function and blocks antitumor immune responses, leading to tumor progression (7). ICIs block the interaction between PD-L1 and PD-1, enhancing anti-tumor immunity (8-11). However, PD-L1 expression in tumor cells has not been standardized as a prognostic marker for lung cancer. Some studies have shown that high PD-L1 expression in tumor cells is associated with poor prognosis in non-small cell lung cancer (NSCLC) (12,13). Other studies have suggested that the relationship between PD-L1 expression in tumor cells and prognosis differs depending on the cancer stage (14). PD-L1 expression in tumor cells is determined using tissue biopsy, which is an invasive procedure. Moreover, it is sometimes difficult to obtain sufficient tissue samples to detect PD-L1 expression in tumor cells, depending on the patient’s condition, tumor location, and tumor size (15). Additionally, PD-L1 expression in tumor cells differs depending on tumor size and heterogeneity (16,17).

Due to the high variability and difficulty in obtaining sufficient tissue samples, circulating blood-based biomarkers are being investigated to predict the response to ICIs and prognosis in lung cancer patients. Unlike PD-L1 expression in tumor cells, blood-based biomarkers offer advantages in terms of ease of sampling, repeatability, and reduced invasiveness (18). Circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), and blood tumor mutational burden (bTMB) are some common blood-based biomarkers (19). Some studies have shown that soluble PD-L1 (sPD-L1) levels in the blood are elevated in patients with cancer compared with those in healthy controls (20-22). Other studies have reported that high levels of sPD-L1 in the blood are associated with poor survival and response to ICI treatment in lung cancer (23,24). However, blood-based biomarkers have some limitations in patients with early-stage cancer that exhibit a low tumor burden (25-27).

Compared to tissue or blood, bronchoalveolar lavage fluid (BALF) demonstrates an enhanced ability for the preservation of tumor-related DNA, improved sensitivity for detecting tumor-derived mutations, and a high potential to reflect intra-tumoral heterogeneity in lung cancer (27,28). BALF samples can reflect tumor microenvironments and are more sensitive than blood or other fluid samples as bronchoscopy operators proximally target pulmonary lesions to obtain BALF samples (29). Using saline, large amounts of tumor-related BALF samples can be obtained (27). In addition, BALF samples could enable the analysis of the cellular and non-cellular contents of the bronchial and alveolar spaces, serving as excellent markers of the tumor microenvironment (30). Therefore, BALF may be more optimal for assessing immune profiles in the lung tumor microenvironment and predicting the efficacy of ICIs (31).

Accordingly, in the present study, we investigated sPD-L1 levels in BALF and their role as a prognostic and predictive marker of ICI treatment response in patients with advanced lung cancer. We present this article in accordance with the STROBE reporting checklist (available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-24-392/rc).


Methods

Study design and participants

This study was conducted in accordance with the Declaration of Helsinki (as revised in 2013) and approved by the Institutional Review Board of Chungnam National University Hospital (IRB No. 2018-01-059). All study samples were obtained after acquiring the study participants’ written informed consent. We prospectively obtained BALF from lung cancer patients who underwent bronchoscopy between clinical need from January 2020 and September 2022 at Chungnam National University Hospital (CNUH). A flowchart depicting the study population is shown in Figure 1. A total of 266 BALF samples were collected. Of these, 43 samples were excluded, including 11 BALF samples obtained from recurrent lesions, 14 samples collected for other reasons (e.g., pneumonia), and 18 BALF samples with incomplete lung cancer staging. A total of 223 samples from patients with lung cancer with complete cancer staging were included. These samples were obtained during the initial bronchoscopy conducted for the diagnosis of lung cancer without prior treatment for lung cancer. The stage and histology of lung cancer are powerful factors affecting overall survival (OS); therefore, in the present study, we included 94 patients initially diagnosed with stage IV NSCLC. We performed a subgroup analysis on 64 patients treated with or without ICIs at our hospital, and among them, we also performed a subgroup analysis on 31 patients who received ICI treatment.

Figure 1 Flow diagram of the patients included in the study. *, we analyzed the overall survival of these patients (N=94); , subgroup analysis was performed for patients with or without ICI treatment (N=64); , subgroup analysis was performed for patients who received ICI treatment (N=31). BALF, bronchoalveolar lavage fluid; CNUH, Chungnam National University Hospital; SCLC, small cell lung cancer; NSCLC, non-small cell lung cancer; ICI, immune checkpoint inhibitor.

Soluble PD-L1 in BALF

Bronchoscopy was performed using a flexible bronchoscope (Olympus, Tokyo, Japan). BALF was obtained in accordance with official recommendations (32) by the instillation of isotonic saline solution in a total of 100 mL into the wedged pulmonary segment that showed the most prominent finding on chest computed tomography (CT). Through gentle suction of the injected solution, the BALF was collected and pooled in a collection tube. The BALF was deemed to be of sufficient quality when the volume, and the number of cells per mL, of BALF were >30 mL and >60,000, respectively, and there was no increase in the number of epithelial cells. For processing the samples, BALF was filter through a 100 µm cell strainer (SPL) to remove clumps and debris and centrifuged at 2,000 rpm for 10 min at room temperature. Separated cell-pellets were treated with ammonium-chloride-potassium lysis buffer (BioLegend) to lyse the red blood cells (RBCs). After separating cell-pellets, the supernatant was aliquoted and stored at −80 ℃ until analysis. Thawed samples were used for measuring sPD-L1 levels in BALF using a human PD-L1 Quantikine ELISA kit (DB7H10; R&D Systems, Minneapolis, MN, USA).

PD-L1 expression in tumor tissue

PD-L1 expression in tumor cells was evaluated using the PD-L1 IHC 22C3 pharmDx test (Agilent Technologies, Santa Clara, CA, USA) on the Dako Autostainer and the PD-L1 IHC SP263 test on the Ventana BenchMark platform. The percentage of tumor cells showing immunoreactivity was quantified according to the respective manufacturer’s guidelines. Positive expression was defined by membrane staining of cancer cells, with cytoplasmic reactions disregarded. PD-L1 protein expression was assessed based on the proportion of viable tumor cells displaying partial or complete membrane staining [tumor proportion score (TPS)] (33). The study categorized PD-L1 expression into three groups based on TPS cut-offs: no expression (<1%), low expression (1–49%), and high expression (≥50%). Classification of subgroups according to PD-L1 expression relied on the 22C3 pharmDx assay results, with patients lacking 22C3 pharmDx results categorized based on SP263 assay results, as both assays are interchangeable (33).

Treatment response and survival analysis

Treatment response was assessed using CT. Treatment response was assessed based on the Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1. Progression-free survival (PFS) was defined as the time from the date of the first drug administration to the date of documented progression or death from any cause. OS was defined as the period from the date of first drug administration to the date of death or the last day of follow-up.

Statistical analysis

The demographic and clinical parameters were analyzed using descriptive statistics. The correlation between PD-L1 expression in tumor cells and sPD-L1 in BALF was analyzed using the Student’s t-test, analysis of variance (ANOVA), and Spearman’s rho method because the data did not follow a normal distribution. Conventional receiver operating characteristic curves were generated to calculate the sensitivity and specificity of biomarkers, conventional receiver operating characteristic (ROC) curves were generated. The optimal cut-off value was determined as the point at which the Youden index was maximized by the ROC curve. The relationships between sPD-L1 in BALF and clinical variables were analyzed using Pearson’s chi-square test. Kaplan-Meier survival analyses and Cox proportional hazard models were used to analyze PFS and OS. Statistical significance was set at P<0.05. SPSS version 26 (IBM Corp., Armonk, NY, USA), Excel 2016 (Microsoft, Redmond, WA, USA), and PRISM version 9 (GraphPad Software Inc., Boston, MA, USA) were used for all statistical analyses and graphics generation.


Results

Soluble PD-L1 levels in BALF in patients with stage IV NSCLC

The trends of the levels of sPD-L1 in BALF are shown in Figure 2A. The median value of sPD-L1 in BALF was 1.45 pg/mL and the mean value was 10.12 pg/mL. We investigated the correlation between PD-L1 expression in tumor cells and sPD-L1 levels in BALF. A Spearman’s rho value of 0.314 suggested that the correlation between these two factors was weakly positive (P=0.002; Figure 2B). ANOVA revealed that the mean value of sPD-L1 in BALF was significantly different (P=0.02) in the no PD-L1 expression group (2.5 pg/mL), low PD-L1 expression group (8.78 pg/mL), and high PD-L1 expression group (24.31 pg/mL) (Figure 2C). In addition, the mean value of PD-L1 expression in tumor cells was significantly different (P=0.01) in patients in the low sPD-L1 in BALF (18.2%) and high sPD-L1 in BALF (44.86%) groups, based on the cutoff value determined by ROC curve analysis (7.35 pg/mL) (Figure 2D).

Figure 2 Distribution and correlation analysis of PD-L1 and sPD-L1 expression in BALF. (A) Scatterplot showing sPD-L1 levels in BALF in all patients (N=94). (B) Graph showing weakly positive correlation between PD-L1 expression in tumor cells and sPD-L1 levels in BALF (P=0.002, Spearman’s rho value =0.314). (C) Mean sPD-L1 in BALF based on tumor PD-L1 expression. (D) Mean tumor PD-L1 expression based on sPD-L1 in BALF. BALF, bronchoalveolar lavage fluid; PD-L1, programmed death-ligand 1; sPD-L1, soluble programmed death-ligand 1.

Patient baseline characteristics according to sPD-L1 in BALF

Based on the cutoff value for sPD-L1 in BALF determined by ROC curve analysis (7.35 pg/mL), all patients were classified into the low sPD-L1 in BALF group (71 patients) and high sPD-L1 in BALF group (23 patients). The clinical parameters of the two groups are summarized in Table 1. There were no significant differences between the two groups in terms of sex, smoking status, Eastern Cooperative Oncology Group Performance Score (ECOG PS), histology, comorbidities, ICI use, targeted mutations, brain metastasis, or NLR. Mean age and PD-L1 expression in tumor cells were significantly different between the two groups.

Table 1

Baseline characteristics of total patients (N=94)

Variables Whole group Low BALF sPDL1 (N=71) High BALF sPDL1 (N=23) P value
Age (years) 72.11±9.4 70.99±9.7 75.77±7.22 0.04*
   <65 18 (19.1) 17 (23.9) 1 (4.3)
   ≥65 76 (80.9) 54 (76.1) 22 (95.7)
Sex 0.95
   Male 69 (73.4) 52 (73.2) 17 (73.9)
   Female 25 (26.6) 19 (26.8) 6 (26.1)
Smoking status 0.54
   Never 25 (26.6) 20 (28.2) 5 (21.7)
   Former 37 (39.4) 29 (40.8) 8 (34.8)
   Current 32 (34.0) 22 (31.0) 10 (43.5)
Smoking history (packs × years) 31.54±25.97 31.12±26.54 24.55±7.22 0.86
ECOG PS 0.66
   0 18 (19.1) 14 (19.7) 4 (17.4)
   1 56 (59.6) 44 (62.0) 12 (52.2)
   2 15 (16.0) 10 (14.1) 5 (21.7)
   3 5 (5.3) 3 (4.2) 2 (8.7)
Histology 0.32
   AD 61 (64.9) 49 (69.0) 12 (52.2)
   SCC 28 (29.8) 19 (26.8) 9 (39.1)
   Other 5 (5.3) 3 (4.2) 2 (8.7)
Comorbidity (COPD) 41 (43.6) 31 (43.7) 10 (43.5) 0.75
ICI use 0.83
   Yes 31 (33.0) 23 (32.4) 8 (34.8)
   No 63 (67.0) 48 (67.6) 15 (65.2)
PD-L1 expression in tumor cell 0.001*
   No 48 (51.1) 44 (62.0) 4 (17.4)
   Low 16 (17.0) 9 (12.7) 7 (30.4)
   High 28 (29.8) 17 (23.9) 11 (47.8)
   N/A 2 (2.1) 1 (1.4) 1 (4.3)
EGFR 0.14
   Wild-type 67 (71.3) 48 (67.6) 19 (82.6)
   Mutant 23 (24.5) 20 (28.2) 3 (13.0)
   N/A 4 (4.3) 3 (4.2) 1 (4.3)
ALK rearrangement 0.91
   Negative 77 (81.9) 60 (84.5) 17 (73.9)
   Positive 5 (5.3) 4 (5.6) 1 (4.3)
   N/A 12 (12.8) 7 (9.9) 5 (21.7)
BRAF 0.34
   Wild-type 41 (43.6) 28 (39.4) 13 (56.5)
   Mutant 2 (2.1) 2 (2.8) 0 (0.0)
   N/A 51 (54.3) 41 (57.7) 10 (43.5)
Brain metastasis 0.07
   Yes 26 (27.7) 23 (32.4) 3 (13.0)
   No 68 (72.3) 48 (67.6) 20 (87.0)
NLR 0.51
   <2.3 26 (27.7) 21 (29.6) 5 (21.7)
   ≥2.3 66 (70.2) 49 (69.0) 17 (73.9)
   N/A 2 (2.1) 1 (1.4) 1 (4.3)
BAL cells (%)
   Neutrophil 54.0±28.3 51.5±30.0 67.3±11.3 0.32
   Lymphocyte 9.6±9.3 9.5±9.8 10.5±6.8 0.85
   Eosinophil 0.84±1.1 0.7±0.9 1.5±1.9 0.48
   Macrophage 34.7±26.3 37.5±27.6 20.0±10.6 0.23

Data are presented as mean ± standard deviation or n (%). *, P<0.05. ECOG PS, Eastern Cooperative Oncology Group performance status; AD, adenocarcinoma; SCC, squamous cell carcinoma; COPD, chronic obstructive pulmonary disease; ICI, immune checkpoint inhibitor; PD-L1, programmed death ligand 1; EGFR, epidermal growth factor receptor; ALK, anaplastic lymphoma kinase; BRAF, v-Raf, murine sarcoma viral oncogene homolog B; NLR, neutrophil-to-lymphocyte ratio; BAL, bronchoalveolar lavage; BALF, bronchoalveolar lavage fluid; sPDL1, soluble programmed death ligand 1; N/A, not available.

OS according to sPD-L1 in BALF

To investigate the prognostic role of sPD-L1 in BALF in patients with NSCLC, we used an ROC curve to determine the optimal cut-off level of sPD-L1 in BALF for the prediction of survival (Figure 3A). We found that a cutoff value of 7.35 pg/mL distinguished best survival and yielded an AUC of 0.626 (P=0.045). Univariate and multivariate analyses were performed for prognostic factors, including sPD-L1 in BALF, age, sex, smoking status, ECOG PS, comorbidity, histology, PD-L1 expression in tumor cells, and brain metastasis (Table S1). Univariate analysis revealed that a longer OS was associated with never smoking, better performance status, no PD-L1 expression in tumor cells, and low sPD-L1 in BALF. Multivariate logistic regression analyses showed that only sPD-L1 in BALF was significantly associated with OS. The median OS of the low sPD-L1 in BALF group was 16.47 months [95% confidence interval (CI): 11.15–21.79 months], which is significantly longer than 8.87 months (95% CI: 0.0–19.88 months) in the high sPD-L1 in BALF group. The hazard ratio (HR) for OS was 2.349 in the high sPD-L1 in BALF group than in the low sPD-L1 in BALF group. In the survival graph, patients with high sPD-L1 in BALF exhibited poorer OS outcomes than patients with low sPD-L1 in BALF (P=0.001, Figure 3B).

Figure 3 OS in all patients based on soluble PD-L1 levels in BALF (N=94). (A) ROC curve of sPD-L1 levels for survival rate in total patients (N=94) showing an AUC value of 0.626 (P=0.045) based on the 7.35 pg/mL cutoff. (B) Graph showing significantly longer median OS of the low sPD-L1 in BALF group (16.47 months; 95% CI: 11.15–21.79) than in the high sPD-L1 in BALF group (8.87 months; 95% CI: 0.0–19.88). AUC, area under the curve; OS, overall survival; sPD-L1, soluble programmed death-ligand 1; PD-L1, programmed death-ligand 1; BALF, bronchoalveolar lavage fluid; ROC, receiver operating characteristic; CI, confidence interval.

Survival analysis in patients who received first-line treatment

Of the 94 patients finally enrolled in the study, we separately analyzed 64 patients who received first-line treatment in our hospital. Of the 64 patients, 49 were classified into the low sPD-L1 in BALF group and 15 in the high sPD-L1. Except for PD-L1 expression in tumor cells (P=0.02), none of the other variables differed significantly between the two groups (Table 2).

Table 2

Baseline characteristics of patients treated with or without ICIs (N=64)

Variables Low BALF sPDL1 (N=49) High BALF sPDL1 (N=15) P value
Age (years) 71.38±9.311 75.27±6.923 0.17
   <65 11 (22.4) 1 (6.7)
   ≥65 38 (77.6) 14 (93.3)
Sex 0.36
   Male 37 (75.5) 13 (86.7)
   Female 12 (24.5) 2 (13.3)
Smoking status 0.12
   Never 14 (28.6) 2 (13.3)
   Former 20 (40.8) 4 (26.7)
   Current 15 (30.6) 9 (60.0)
Smoking history (pack × year) 33.827±28.596 37.980±21.186 0.61
ECOG PS 0.79
   0 7 (14.3) 2 (13.3)
   1 33 (67.3) 9 (60.0)
   2 8 (16.3) 4 (26.7)
   3 1 (2.0) 0 (0.0)
Comorbidity (COPD) 23 (46.9) 7 (46.7) 0.71
Histology 0.15
   AD 36 (73.5) 7 (46.7)
   SCC 11 (22.4) 7 (46.7)
   Other 2 (4.1) 1 (6.7)
ICI use 0.60
   Yes 20 (40.8) 5 (33.3)
   No 29 (59.2) 10 (66.7)
Chemotherapy combined 0.31
   ICI only 7 (14.3) 3 (20.0)
   Combined 13 (26.5) 2 (13.3)
Best response to treatment 0.50
   CR 2 (4.1) 0 (0.0)
   PR 28 (57.1) 7 (46.7)
   SD 13 (26.5) 4 (26.7)
   PD 6 (12.2) 4 (26.7)
First response to treatment 0.47
   CR 2 (4.1) 0 (0.0)
   PR 19 (38.8) 6 (40.0)
   SD 22 (44.9) 5 (33.3)
   PD 6 (12.2) 4 (26.7)
ORR 61.2% 46.7% 0.32
PD-L1 expression in tumor cell 0.02*
   No 29 (59.2) 3 (20.0)
   Low 8 (16.3) 6 (40.0)
   High 12 (24.5) 6 (40.0)
EGFR 0.13
   Wild-type 32 (65.3) 13 (86.7)
   Mutant 16 (32.7) 2 (13.3)
   N/A 1 (2.0) 0 (0.0)
ALK rearrangement 0.38
   Negative 42 (85.7) 11 (73.3)
   Positive 3 (6.1) 0 (0.0)
   N/A 4 (8.2) 4 (26.7)
BRAF 0.38
   Wild-type 23 (46.9) 9 (60.0)
   Mutant 2 (4.1) 0 (0.0)
   N/A 24 (49.0) 6 (40.0)
Brain metastasis 0.11
   Yes 17 (34.7) 2 (13.3)
   No 32 (65.3) 13 (86.7)
NLR 0.96
   <2.3 16 (32.7) 5 (33.3)
   ≥2.3 33 (67.3) 10 (66.7)

Data are presented as mean ± standard deviation or n (%). *, P<0.05. ICIs, immune checkpoint inhibitors; ECOG PS, Eastern Cooperative Oncology Group performance status; COPD, chronic obstructive pulmonary disease; AD, adenocarcinoma; SCC, squamous cell carcinoma; CR, complete response; PR, partial response; SD, stable disease; PD, progressive disease; ORR, objective response rate; PD-L1, programmed death ligand 1; EGFR, epidermal growth factor receptor; ALK, anaplastic lymphoma kinase; BRAF, v-Raf murine sarcoma viral oncogene homolog B; NLR, neutrophil-to-lymphocyte ratio; BALF, bronchoalveolar lavage fluid; sPDL1, soluble programmed death ligand 1; N/A, not available.

We performed univariate and multivariate analyses for prognostic factors, including sPD-L1 levels in BALF, age, sex, smoking status, ECOG PS, comorbidity, histology, ICI use for chemotherapy, ICI and chemotherapy combination therapy, PD-L1 expression in tumor cells, molecular expression (EGFR, ALK, and BRAF), and brain metastasis (Table 3). Univariate and multivariate analyses revealed that both histology and sPD-L1 in BALF were significantly associated with PFS. Univariate analysis for OS showed that sPD-L1 in BALF was the only significant factor associated with OS. In the survival graph of 64 patients who received first-line treatment at our hospital, the high sPD-L1 in BALF group exhibited poorer PFS and OS than the low sPD-L1 in BALF group. The median PFS of the low sPD-L1 in BALF group was 10.2 months (95% CI: 6.45–13.95 months), which was significantly longer than 3.9 months (95% CI: 1.38–6.43 months) in the high sPD-L1 in BALF group (P=0.001, Figure 4A). The median OS of the low sPD-L1 in BALF group was 20.57 months (95% CI: 13.86–27.27 months), which was significantly longer than 10.60 months (95% CI: 5.87–15.33 months) in the high sPD-L1 in BALF group (P=0.003, Figure 4B).

Table 3

Univariate and multivariate analyses of PFS and OS in patients treated with or without ICIs (N=64)

Variables N=64 PFS OS
Univariate analysis Multivariate analysis Univariate analysis
Median (95% CI) (months) Exp(B)
(95% CI)
P value Exp(B)
(95% CI)
P value Median (95% CI) (months) Exp(B)
(95% CI)
P value
Age (years) 0.96 0.58
   <65 12 10.17
(2.97–12.70)
1 14.90
(10.25–17.72)
1
   ≥65 52 7.75
(5.30–10.58)
1.021
(0.493–2.113)
9.00
(5.98–11.31)
1.279
(0.530–3.087)
Sex 0.17 0.39
   Male 50 6.28
(4.37–10.08)
1 12.62
(8.97–14.20)
1
   Female 14 12.30
(7.47–14.00)
0.614
(0.305–1.235)
15.10
(12.90–17.93)
0.696
(0.304–1.593)
Smoking status 0.11 0.33
   Never 16 12.70
(7.93–14.00)
1 15.60
(13.77–17.93)
1
   Former 24 5.12
(3.10–10.20)
2.167
(1.031–4.557)
11.82
(6.69–14.20)
1.896
(0.797–4.510)
   Current 24 8.32
(3.83–10.67)
1.728
(0.810–3.684)
12.75
(6.52–16.37)
1.387
(0.573–3.360)
ECOG PS 0.12 0.10
   0 9 12.97
(9.93–16.60)
1 16.30
(9.97–18.10)
1
   1 42 6.78
(3.92–10.50)
2.321
(0.901–5.980)
12.93
(9.73–14.40)
3.226
(0.757–13.757)
   2 12 6.25
(2.37–10.93)
3.037
(1.047–8.807)
10.73
(4.08–15.93)
5.366
(1.172–24.574)
   3 1 12.97
(12.97–12.97)
0 (0) 12.90
(12.90–12.90)
0 (0)
Comorbidity 0.33 0.08
   COPD 30 5.75
(3.83–10.50)
1.352
(0.740–2.470)
12.38
(7.47–12.20)
1.931
(0.914–4.080)
Histology 0.01* 0.02* 0.60
   AD 43 10.67
(4.72–12.40)
1 1 14.00
(12.13–16.18)
1
   SCC 18 7.02
(4.57–10.20)
1.502
(0.796–2.834)
0.994
(0.481–2.051)
11.18
(8.72–13.60)
1.439
(0.688–3.007)
   Other 3 2.97
(0.23–5.30)
5.177
(1.500–17.874)
5.718
(1.643–19.905)
13.43
(7.60–15.80)
1.402
(0.326–6.023)
ICI use for 1st treatment 0.82 0.72
   Yes 25 7.93
(3.88–10.67)
0.932
(0.517–1.681)
12.63
(9.97–14.97)
0.882
(0.439–1.773)
   No 39 9.07
(4.93–12.20)
1 13.76
(9.42–15.60)
1
ICI + chemotherapy combined for 1st treatment 0.34 0.34
   ICI only 10 4.53
(0.62–12.27)
1 11.70
(2.77–16.28)
1
   Combined 15 9.53
(4.38–12.98)
0.627
(0.238–1.657)
12.63
(11.05–14.97)
0.580
(0.186–1.814)
PD-L1 expression in tumor cell 0.77 0.26
   No 32 9.33
(4.93–12.40)
1 13.90
(10.87–16.37)
1
   Low 14 5.67
(3.90–10.50)
1.286
(0.629–2.628)
11.18
(7.97–14.80)
1.955
(0.867–4.409)
   High 18 8.50
(2.42–12.97)
1.025
(0.519–1.986)
13.37
(5.43–15.52)
1.362
(0.618–3.006)
EGFR 0.11 0.49
   Wild-type 45 7.47
(4.38–9.93)
1 12.63
(10.30–13.77)
1
   Mutant 18 12.60
(9.03–14.57)
0.593
(0.309–1.137)
15.50
(10.58–17.92)
0.767
(0.363–1.620)
ALK rearrangement 0.18 0.19
   Negative 53 7.93
(4.93–10.50)
1 13.60
(10.60–15.50)
1
   Positive 3 12.97
(12.40–12.97)
0.281
(0.038–2.053)
12.90
(12.90–14.97)
0.045
(0.000–57.157)
BRAF 0.38 0.19
   Wild-type 32 4.90
(3.77–9.97)
1 11.50
(5.43–14.00)
1
   Mutant 2 10.62
(7.47–13.77)
0.419
(0.057–3.097)
16.30
(13.77–18.83)
0.043
(0.000–57.999)
Brain metastasis 0.55 0.17
   Yes 19 6.03
(3.10–13.00)
1.201
(0.659–2.189)
14.40
(4.07–16.07)
1
   No 45 9.53
(5.40–11.07)
1 12.97
(11.18–14.28)
1.601
(0.814–3.151)
sPD-L1 in BALF (pg/mL) 0.003* 0.006* 0.003*
   <7.35 49 10.20
(6.45–13.95)
1 1 20.57
(13.86–27.27)
1
   ≥7.35 15 3.90
(1.38–6.43)
2.637
(1.381–5.038)
2.872
(1.362–6.055)
10.60
(5.87–15.33)
2.771
(1.389–5.530)

*, P<0.05. PFS, progression-free survival; CI, confidence interval; OS, overall survival; ICIs, immune checkpoint inhibitors; ECOG PS, Eastern Cooperative Oncology Group performance status; COPD, chronic obstructive pulmonary disease; AD, adenocarcinoma; SCC, squamous cell carcinoma; PD-L1, programmed death ligand 1; EGFR, epidermal growth factor receptor; ALK, anaplastic lymphoma kinase; BRAF, v-Raf murine sarcoma viral oncogene homolog B; BALF, bronchoalveolar lavage fluid; sPD-L1, soluble programmed death ligand 1.

Figure 4 PFS and OS in patients with or without ICI treatment (N=64). (A) The median PFS of the low sPD-L1 in BALF group was 10.20 months (95% CI: 6.45–13.95 months), which was significantly longer than 3.90 months (95% CI: 1.38–6.43 months) in the high sPD-L1 in BALF group (P=0.001). (B) The median OS of the low sPD-L1 in BALF group was 20.57 months (95% CI: 13.86–27.27 months), which was significantly longer than 10.60 months (95% CI: 5.87–15.33 months) in the high sPD-L1 in BALF group (P=0.003). PFS, progression-free survival; sPD-L1, soluble programmed death-ligand 1; BALF, bronchoalveolar lavage fluid; OS, overall survival; ICI, immune checkpoint inhibitor; CI, confidence interval.

Survival analysis in patients treated with immunotherapy

We analyzed 31 patients treated with ICI separately. The 31 patients were classified into the low sPD-L1 in BALF (23 patients) and high sPD-L1 in BALF group (8 patients). There were no significant differences in baseline characteristics between the two groups (Table 4). The objective response rate (ORR) was significantly higher in the low sPD-L1 in BALF group than in the high sPD-L1 in BALF group (ORR: 60.9% vs. 12.5%, P=0.02, Figure 5A).

Table 4

Baseline characteristics of patients who received ICI treatment (N=31)

Variables Low BALF sPD-L1 (N=23) High BALF sPD-L1 (N=8) P value
Age (years) 71.30±7.82 75.38±2.07 0.21
  <65 4 (17.4) 0 (0.0)
  ≥65 19 (82.6) 8 (100.0)
Sex 0.55
   Male 22 (95.7) 8 (100.0)
   Female 1 (4.3) 0 (0.0)
Smoking status 0.50
   Never 1 (4.3) 0 (0.0)
   Former 10 (43.5) 2 (25.0)
   Current 12 (52.2) 6 (75.0)
Smoking history (pack × year) 51.44±23.76 43.09±13.86 0.36
ECOG PS 0.18
   0 1 (4.3) 2 (25.0)
   1 18 (78.3) 4 (50.0)
   2 4 (17.4) 2 (25.0)
   3 0 (0.0) 0 (0.0)
Comorbidity (COPD) 15 (65.2) 4 (50.0) 0.28
Histology 0.06
   AD 14 (60.9) 1 (12.5)
   SCC 7 (30.4) 6 (75)
   Other 2 (8.7) 1 (12.5)
Chemotherapy combined 0.12
   ICI only 10 (43.5) 6 (75.0)
   Combined 13 (56.5) 2 (25.0)
Agent (ICI) 0.30
   Pembrolizumab 20 (87.0) 5 (62.5)
   Atezolizumab 3 (13.0) 3 (37.5)
ICI treatment line 0.30
   1st 20 (87.0) 5 (62.5)
   ≥2nd 3 (13.0) 3 (37.5)
First response to treatment 0.19
   PR 11 (47.8) 1 (12.5)
   SD 8 (34.8) 4 (50.0)
   PD 4 (17.4) 3 (37.5)
BR to treatment 0.06
   PR 14 (60.9) 1 (12.5)
   SD 5 (21.7) 4 (50.0)
   PD 4 (17.4) 3 (37.5)
ORR 60.9% 12.5% 0.02*
PD-L1 expression in tumor cell 0.09
   No 10 (43.5) 1 (12.5)
   Low 5 (21.7) 5 (62.5)
   High 8 (34.8) 2 (25.0)
IRAE 0.47
   Yes 9 (39.1) 2 (25.0)
   No 14 (60.9) 6 (75.0)
Type of IRAE 0.23
   Pneumonitis 4 (17.4) 0 (0.0)
   Thyroid disease 0 (0.0) 1 (12.5)
   Dermatitis 4 (17.4) 1 (12.5)
   Pleural effusion 1 (4.3) 0 (0.0)
Brain metastasis 0.11
   Yes 6 (26.1) 0 (0.0)
   No 17 (73.9) 8 (100.0)
NLR 0.11
   <2.3 7 (30.4) 5 (62.5)
   ≥2.3 16 (69.6) 3 (37.5)

Data are presented as mean ± standard deviation or n (%). *, P<0.05. ICI, immune checkpoint inhibitor; ECOG PS, Eastern Cooperative Oncology Group performance status; COPD, chronic obstructive pulmonary disease; AD, adenocarcinoma; SCC, squamous cell carcinoma; PR, partial response; SD, stable disease; PD, progressive disease; BR, best response; ORR, objective response rate; PD-L1, programmed death ligand 1; IRAE, immune-related adverse events; NLR, neutrophil-to-lymphocyte ratio; BALF, bronchoalveolar lavage fluid; sPD-L1, soluble programmed death ligand 1.

Figure 5 ORR, PFS, and OS in patients who received immunotherapy (N=31). (A) Bar graph showing significantly higher ORR in patients with low sPD-L1 in BALF (60.9%) than in those with high sPD-L1 in BALF (12.5%) (P=0.02). (B) Graph showing significantly longer median PFS in the low sPD-L1 in BALF (7.57 months; 95% CI: 3.65–11.48) than in the high sPD-L1 in BALF group (2.0 months; 95% CI: 0.0–5.74). (C) Graph showing significantly longer median OS in the low sPD-L1 in BALF (not reached) than in the high sPD-L1 in BALF group (4.17 months; 95% CI: 0.0–14.38). ORR, objective response rate; sPD-L1, soluble programmed death-ligand 1; BALF, bronchoalveolar lavage fluid; PFS, progression-free survival; OS, overall survival; CI, confidence interval.

We performed univariate analysis for prognostic factors, including sPD-L1 levels in BALF, age, sex, smoking status, ECOG PS, comorbidity, histology, ICI and chemotherapy combined therapy, PD-L1 expression in tumor cells, and brain metastasis (Table 5). Univariate analysis revealed no significant factors associated with PFS. Although there was not significantly, the high sPD-L1 in BALF group showed poor PFS than the low sPD-L1 in BALF group. The median PFS of the low sPD-L1 in BALF group was 7.57 months (95% CI: 3.65–11.48 months), which was longer than 2.0 months (95% CI: 0.0–5.74 months) in the high sPD-L1 in BALF group (Figure 5B). Univariate analysis for OS revealed sPD-L1 in BALF as the only significant factor associated with OS (P=0.01). In the Kaplan-Meier survival curve for OS, the high sPD-L1 in BALF group exhibited poorer outcomes than the low sPD-L1 in BALF group. The median OS of the low sPD-L1 in BALF group was not reached, which was significantly longer than 4.17 months (95% CI: 0.0–14.38 months) in the high sPD-L1 in BALF group (P=0.01, Figure 5C).

Table 5

Univariate analysis of PFS and OS in patients who received ICI treatment (N=31)

Variables N=31 PFS OS
Univariate analysis Univariate analysis
Median (95% CI) (months) Exp(B) (95% CI) P value Median (95% CI) (months) Exp(B) (95% CI) P value
Age (years) 0.90 0.20
   <65 4 7.63 (2.97–9.13) 1 15.75 (7.60–19.50) 1
   ≥65 27 4.77 (2.73–7.93) 1.084 (0.317–3.709) 12.13 (4.53–13.43) 3.443 (0.454–26.093)
Sex 0.97 0.91
   Male 30 4.82 (3.37–8.17) 1 12.37 (5.90–13.76) 1
   Female 1 7.93 (7.93–7.93) 1.039 (0.138–7.832) 16.07 (16.07–16.07) 1.120 (0.146–8.582)
Smoking status 0.65 0.48
   Never 1 7.93 (7.93–7.93) 1 16.07 (16.07–16.07) 1
   Former 12 3.05 (1.10–8.53) 1.239 (0.155–9.932) 11.82 (1.30–13.82) 1.277 (0.156–10.480)
   Current 18 5.67 (3.83–9.13) 0.831 (0.107–6.462) 12.78 (6.07–15.67) 0.700 (0.086–5.674)
ECOG PS 0.56
  0 3 4.20 (1.13–11.33) 1 9.97 (1.27–16.77) 1 0.92
  1 22 4.81 (2.73–8.80) 2.797 (0.367–21.285) 12.62 (4.55–13.60) 1.490 (0.193–11.513)
  2 6 6.61 (2.47–11.63) 2.249 (0.261–19.349) 13.20 (5.03–17.32) 1.580 (0.176–14.170)
  3 0
Comorbidity (COPD) 19 7.53 (2.00–9.43) 0.460 (0.182–1.166) 0.09 12.63 (2.73–14.20) 0.850 (0.307–2.356) 0.76
Histology 0.11 0.54
   AD 15 7.93 (1.92–9.90) 1 13.77 (4.32–17.25) 1
   SCC 13 4.20 (3.77–7.57) 1.726 (0.683–4.365) 10.60 (4.17–13.13) 1.831 (0.626–5.359)
   Other 3 2.97 (0.23–5.30) 3.950 (0.998–15.634) 13.43 (7.60–15.80) 1.387 (0.280–6.879)
ICI + chemotherapy combined 0.08 0.11
   ICI only 16 3.80 (1.10–5.30) 1 7.93 (1.33–15.80) 1
   Combined 15 8.80 (4.00–9.43) 0.480 (0.208–1.108) 12.63 (11.05–14.58) 0.451 (0.166–1.226)
PD-L1 expression in tumor cell 0.96 0.82
   No 11 6.47 (2.97–9.13) 1 12.60 (4.17–16.37) 1
   Low 10 4.48 (2.00–9.43) 0.948 (0.352–2.555) 11.05 (2.38–14.97) 1.438 (0.437–4.736)
   High 10 6.22 (1.10–9.45) 0.869 (0.321–2.351) 13.13 (2.77–16.77) 1.352 (0.412–4.435)
Brain metastasis 0.27 0.16
   Yes 6 8.53 (3.20–13.98) 0.547 (0.184–1.630) 16.42 (8.48–26.27) 1
   No 25 4.20 (2.73–7.57) 1 11.50 (4.55–13.13) 0.358 (0.079–1.610)
sPD-L1 in BALF (pg/mL) 0.06 0.01*
   <7.35 23 7.57 (3.65–11.48) 1 Not reached 1
   ≥7.35 8 2.0 (0.0–5.74) 2.319 (0.928–5.796) 4.17 (0.0–14.38) 3.414 (1.250–9.322)

*, P<0.05. PFS, progression-free survival; OS, overall survival; ICI, immune checkpoint inhibitor; ECOG PS, Eastern Cooperative Oncology Group performance status; COPD, chronic obstructive pulmonary disease; AD, adenocarcinoma; SCC, squamous cell carcinoma; PD-L1, programmed death ligand 1; BALF, bronchoalveolar lavage fluid; sPDL1, soluble programmed death ligand 1.


Discussion

To our knowledge, this is the first study to analyze the predictive and prognostic significance of sPD-L1 in the BALF of patients with stage IV NSCLC (Figure 6), and to discover the correlation between sPD-L1 in BALF and PD-L1 expression in tumor cells. We found that sPD-L1 in BALF was a significant prognostic factor affecting both PFS and OS in patients with NSCLC who received treatment, regardless of the type of treatment agent. Among patients receiving ICI treatment, those with high sPD-L1 in BALF showed a significantly lower response to ICI treatment, confirming that sPD-L1 in BALF can serve as a predictive and prognostic marker for the efficacy and outcomes of ICI therapy.

Figure 6 Graphical summary of the study. NSCLC, non-small cell lung cancer; BAL, bronchoalveolar lavage; PD-L1, programmed death-ligand 1; sPD-L1, soluble programmed death-ligand 1.

We enrolled 94 NSCLC patients who underwent bronchoscopy for the diagnosis of lung cancer. These patients had not received any anticancer treatment for lung cancer, minimizing the potential confounding effects associated with changes in PD-L1 expression in tumor cells after chemotherapy, as observed in previous studies (34-36). In addition, we examined the influence of factors affecting sPD-L1 in BALF. sPD-L1 in BALF is elevated in patients with COPD or a history of smoking (37-39). We categorized patients into several groups based on COPD and smoking status, conducted chi-square tests, and demonstrated that these factors were not associated with sPD-L1 levels in the BALF of NSCLC patients.

As immunotherapy has become the standard treatment for lung cancer, it is crucial to identify potential biomarkers suitable for screening patients who may benefit from immunotherapy (40). PD-L1 expression in tumor cells, a suboptimal marker for predicting the therapeutic efficacy of NSCLC immunotherapy, is currently the most established and widely used biomarker for immunotherapy for NSCLC (4,41,42). High PD-L1 expression in tumor cell is associated with a favorable response to ICIs (2,43). Furthermore, PD-L1 expression is associated with increased tumor proliferation and aggressiveness in lung cancer (44). However, small biopsies can misclassify PD-L1 expression in tumor cells by up to 35% and PD-L1 expression of tumor cells can vary depending on sample size of biopsy (16,17). The site of biopsy and primary and metastatic lesions may also affect changes in PD-L1 expression (40). Therefore, the prognostic and predictive performance of PD-L1 expression in tumor cells remains suboptimal.

sPD-L1 is usually detected in the blood samples of patients with cancer. Soluble PD-1 (sPD-1) and sPD-L1, both soluble forms of checkpoints, are elevated in patients with advanced cancer, and high levels of sPD-L1 in the blood indicate a poorer prognosis in patients with lung cancer (20,22,45,46). However, sPD-L1 levels in the blood are also elevated in other diseases such as sepsis, acute respiratory distress syndrome (ARDS), acute pancreatitis, and auto-inflammatory diseases (5). Moreover, as a circulating protein, it reflects conditions not specifically implicated in tumors. However, sPD-L1 in BALF is typically collected in close proximity to tumor lesions, making it more specific to the tumor microenvironment (27,28,30). In addition, obtaining sPD-L1 in BALF is less invasive than biopsy and repeated examinations, and obtaining a substantial sample quantity is easier than biopsy. In a previous study, it was reported that increased sPD-L1 levels in the BALF of ARDS patients was associated with increased lung inflammation and ARDS severity (47). Another study reported that sPD-L1 level in the BALF was increased in patients with neutrophilic asthma and that sPD-L1 was associated with Th17/IL-17 immune responses (48). However, thus far, no study has investigated the sPD-L1 expression in the BALF of lung cancer patients.

In this study, we demonstrated a positive correlation between PD-L1 expression in tumor cells and sPD-L1 levels in BALF. However, in contrast to the results of previous studies that showed that patients with high PD-L1 expression in tumor cells showed a better response to ICI treatment, we found that patients with higher soluble PD-L1 in BALF showed a poorer response to ICI treatment. We classified patients into four groups based on PD-L1 expression in tumor cells and sPD-L1 levels in BALF (Table S2). Even in patients with no or low tumor PD-L1 expression, the ORR was approximately 60% when BALF sPD-L1 levels were low. The ORR in patients with high tumor PD-L1 expression and high sPD-L1 in BALF was 0%, whereas that in patients with high tumor PD-L1 expression and low sPD-L1 in BALF was 62.5%. The efficacy of ICI treatment may decrease when both PD-L1 expression in tumors and sPD-L1 levels in BALF are high, although it has been reported that there is no significant difference in efficacy between ICI monotherapy and ICI combination therapy when PD-L1 expression in tumor cells is high (49,50). Although it was difficult to determine this because the number of patients in each group was very small, our results suggest that ICI combination therapy using cytotoxic agents may be better than ICI monotherapy when the levels of both PD-L1 in tumor cells and sPD-L1 in BALF are high. Moreover, among patients with similar PD-L1 expression in tumor cells, sPD-L1 in BALF could be a valuable indicator for predicting the response to ICI.

This study had several limitations. First, it was conducted at a single center, resulting in a relatively small number of participants. In particular, the number of patients who received ICI treatment was small; therefore, validation of our results in a larger cohort is required. Second, the mechanism by which sPD-L1 in BALF contributes to poor prognosis in patients with lung cancer and is associated with reduced efficacy of ICI treatment remains to be elucidated.


Conclusions

We demonstrated the potential of sPD-L1 in BALF as a promising biomarker in NSCLC prognosis and ICI efficacy, offering advantages such as ease of sampling, less invasive procedure compared to traditional biopsy, and increased sensitivity for reflecting the tumor microenvironment compared to other established blood biomarkers.


Acknowledgments

The authors would like to thank Editage (www.editage.co.kr) for English language editing.

Funding: This research was supported by the Basic Science Research Program of the National Research Foundation of Korea (NRF), funded by the Ministry of Science and Technology (Nos. NRF-2022R1C1C1007301 and NRF-2022R1A2C2010148). This research was supported by grants from the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare, Republic of Korea (Nos. HR20C0025 and HR22C1734). This work was supported by research fund of Chungnam National University Hospital.


Footnote

Reporting Checklist: The authors have completed the STROBE reporting checklist. Available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-24-392/rc

Data Sharing Statement: Available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-24-392/dss

Peer Review File: Available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-24-392/prf

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-24-392/coif). The authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved. This study was conducted per the Declaration of Helsinki (as revised in 2013) and approved by the Institutional Review Board of Chungnam National University Hospital (IRB No. 2018-01-059). Informed consent was obtained from all participants involved in the study.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Siegel RL, Miller KD, Wagle NS, et al. Cancer statistics, 2023. CA Cancer J Clin 2023;73:17-48. [Crossref] [PubMed]
  2. Patel SP, Kurzrock R. PD-L1 Expression as a Predictive Biomarker in Cancer Immunotherapy. Mol Cancer Ther 2015;14:847-56. [Crossref] [PubMed]
  3. Grigg C, Rizvi NA. PD-L1 biomarker testing for non-small cell lung cancer: truth or fiction? J Immunother Cancer 2016;4:48. [Crossref] [PubMed]
  4. Gridelli C, Ardizzoni A, Barberis M, et al. Predictive biomarkers of immunotherapy for non-small cell lung cancer: results from an Experts Panel Meeting of the Italian Association of Thoracic Oncology. Transl Lung Cancer Res 2017;6:373-86. [Crossref] [PubMed]
  5. Bailly C, Thuru X, Quesnel B. Soluble Programmed Death Ligand-1 (sPD-L1): A Pool of Circulating Proteins Implicated in Health and Diseases. Cancers (Basel) 2021;13:3034. [Crossref] [PubMed]
  6. Zhu X, Lang J. Soluble PD-1 and PD-L1: predictive and prognostic significance in cancer. Oncotarget 2017;8:97671-82. [Crossref] [PubMed]
  7. Alsaab HO, Sau S, Alzhrani R, et al. PD-1 and PD-L1 Checkpoint Signaling Inhibition for Cancer Immunotherapy: Mechanism, Combinations, and Clinical Outcome. Front Pharmacol 2017;8:561. [Crossref] [PubMed]
  8. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 2012;12:252-64. [Crossref] [PubMed]
  9. Francisco LM, Salinas VH, Brown KE, et al. PD-L1 regulates the development, maintenance, and function of induced regulatory T cells. J Exp Med 2009;206:3015-29. [Crossref] [PubMed]
  10. Boussiotis VA. Molecular and Biochemical Aspects of the PD-1 Checkpoint Pathway. N Engl J Med 2016;375:1767-78. [Crossref] [PubMed]
  11. Chen DS, Irving BA, Hodi FS. Molecular pathways: next-generation immunotherapy--inhibiting programmed death-ligand 1 and programmed death-1. Clin Cancer Res 2012;18:6580-7. [Crossref] [PubMed]
  12. Li H, Xu Y, Wan B, et al. The clinicopathological and prognostic significance of PD-L1 expression assessed by immunohistochemistry in lung cancer: a meta-analysis of 50 studies with 11,383 patients. Transl Lung Cancer Res 2019;8:429-49. [Crossref] [PubMed]
  13. Pan ZK, Ye F, Wu X, et al. Clinicopathological and prognostic significance of programmed cell death ligand1 (PD-L1) expression in patients with non-small cell lung cancer: a meta-analysis. J Thorac Dis 2015;7:462-70. [PubMed]
  14. Chang CH, Shih AC, Chang YH, et al. The Prognostic Significance of PD1 and PDL1 Gene Expression in Lung Cancer: A Meta-Analysis. Front Oncol 2021;11:759497. [Crossref] [PubMed]
  15. Lee SH, Kim EY, Kim T, et al. 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. [Crossref] [PubMed]
  16. Bigras G, Mairs S, Swanson PE, et al. Small Biopsies Misclassify up to 35% of PD-L1 Assessments in Advanced Lung Non-Small Cell Lung Carcinomas. Appl Immunohistochem Mol Morphol 2018;26:701-8. [Crossref] [PubMed]
  17. Thunnissen E, Kerr KM, Dafni U, et al. Programmed death-ligand 1 expression influenced by tissue sample size. Scoring based on tissue microarrays' and cross-validation with resections, in patients with, stage I-III, non-small cell lung carcinoma of the European Thoracic Oncology Platform Lungscape cohort. Mod Pathol 2020;33:792-801. [Crossref] [PubMed]
  18. An HJ, Chon HJ, Kim C. Peripheral Blood-Based Biomarkers for Immune Checkpoint Inhibitors. Int J Mol Sci 2021;22:9414. [Crossref] [PubMed]
  19. Ancel J, Dormoy V, Raby BN, et al. Soluble biomarkers to predict clinical outcomes in non-small cell lung cancer treated by immune checkpoints inhibitors. Front Immunol 2023;14:1171649. [Crossref] [PubMed]
  20. Zhang J, Gao J, Li Y, et al. Circulating PD-L1 in NSCLC patients and the correlation between the level of PD-L1 expression and the clinical characteristics. Thorac Cancer 2015;6:534-8. [Crossref] [PubMed]
  21. Cheng S, Zheng J, Zhu J, et al. PD-L1 gene polymorphism and high level of plasma soluble PD-L1 protein may be associated with non-small cell lung cancer. Int J Biol Markers 2015;30:e364-8. [Crossref] [PubMed]
  22. Okuma Y, Hosomi Y, Nakahara Y, et al. High plasma levels of soluble programmed cell death ligand 1 are prognostic for reduced survival in advanced lung cancer. Lung Cancer 2017;104:1-6. [Crossref] [PubMed]
  23. Oh SY, Kim S, Keam B, et al. Soluble PD-L1 is a predictive and prognostic biomarker in advanced cancer patients who receive immune checkpoint blockade treatment. Sci Rep 2021;11:19712. [Crossref] [PubMed]
  24. Zhu X, Lang J. Soluble PD-1 and PD-L1: predictive and prognostic significance in cancer. Oncotarget 2017;8:97671-82. [Crossref] [PubMed]
  25. Zhao X, Han RB, Zhao J, et al. Comparison of epidermal growth factor receptor mutation statuses in tissue and plasma in stage I-IV non-small cell lung cancer patients. Respiration 2013;85:119-25. [Crossref] [PubMed]
  26. Underhill HR, Kitzman JO, Hellwig S, et al. Fragment Length of Circulating Tumor DNA. PLoS Genet 2016;12:e1006162. [Crossref] [PubMed]
  27. Zhang X, Li C, Ye M, et al. Bronchial Washing Fluid Versus Plasma and Bronchoscopy Biopsy Samples for Detecting Epidermal Growth Factor Receptor Mutation Status in Lung Cancer. Front Oncol 2021;11:602402. [Crossref] [PubMed]
  28. Nair VS, Hui AB, Chabon JJ, et al. Genomic Profiling of Bronchoalveolar Lavage Fluid in Lung Cancer. Cancer Res 2022;82:2838-47. [Crossref] [PubMed]
  29. Meyer KC. Bronchoalveolar lavage as a diagnostic tool. Semin Respir Crit Care Med 2007;28:546-60. [Crossref] [PubMed]
  30. Domagala-Kulawik J. The relevance of bronchoalveolar lavage fluid analysis for lung cancer patients. Expert Rev Respir Med 2020;14:329-37. [Crossref] [PubMed]
  31. Masuhiro K, Tamiya M, Fujimoto K, et al. Bronchoalveolar lavage fluid reveals factors contributing to the efficacy of PD-1 blockade in lung cancer. JCI Insight 2022;7:e157915. [Crossref] [PubMed]
  32. Baughman RP. Technical aspects of bronchoalveolar lavage: recommendations for a standard procedure. Semin Respir Crit Care Med 2007;28:475-85. [Crossref] [PubMed]
  33. Park HY, Oh IJ, Kho BG, et al. Clinical Characteristics of Korean Patients with Lung Cancer Who Have Programmed Death-Ligand 1 Expression. Tuberc Respir Dis (Seoul) 2019;82:227-33. [Crossref] [PubMed]
  34. Funaki S, Shintani Y, Kawamura T, et al. Chemotherapy enhances programmed cell death 1/ligand 1 expression via TGF-β induced epithelial mesenchymal transition in non-small cell lung cancer. Oncol Rep 2017;38:2277-84. [Crossref] [PubMed]
  35. Shin J, Chung JH, Kim SH, et al. Effect of Platinum-Based Chemotherapy on PD-L1 Expression on Tumor Cells in Non-small Cell Lung Cancer. Cancer Res Treat 2019;51:1086-97. [Crossref] [PubMed]
  36. Rojkó L, Reiniger L, Téglási V, et al. Chemotherapy treatment is associated with altered PD-L1 expression in lung cancer patients. J Cancer Res Clin Oncol 2018;144:1219-26. [Crossref] [PubMed]
  37. Polverino F, Mirra D, Yang CX, et al. Similar programmed death ligand 1 (PD-L1) expression profile in patients with mild COPD and lung cancer. Sci Rep 2022;12:22402. [Crossref] [PubMed]
  38. Ritzmann F, Borchardt K, Vella G, et al. Blockade of PD-1 decreases neutrophilic inflammation and lung damage in experimental COPD. Am J Physiol Lung Cell Mol Physiol 2021;320:L958-68. [Crossref] [PubMed]
  39. Li L, Yan J, Ma LQ, et al. Effects of Maxingloushi decoction on immune inflammation and programmed death markers in mice with chronic obstructive pulmonary disease. World J Emerg Med 2022;13:32-7. [Crossref] [PubMed]
  40. Wang L, Hu Y, Wang S, et al. Biomarkers of immunotherapy in non-small cell lung cancer. Oncol Lett 2020;20:139. [Crossref] [PubMed]
  41. Herbst RS, Baas P, Kim DW, et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial. Lancet 2016;387:1540-50. [Crossref] [PubMed]
  42. Mok TSK, Wu YL, Kudaba I, et al. Final analysis of the phase III KEYNOTE-042 study: Pembrolizumab (Pembro) versus platinum-based chemotherapy (Chemo) as first-line therapy for patients (Pts) with PD-L1-positive locally advanced/metastatic NSCLC. Ann Oncol 2019;30:ii38-ii68. [Crossref]
  43. Yu H, Boyle TA, Zhou C, et al. PD-L1 Expression in Lung Cancer. J Thorac Oncol 2016;11:964-75. [Crossref] [PubMed]
  44. Pawelczyk K, Piotrowska A, Ciesielska U, et al. Role of PD-L1 Expression in Non-Small Cell Lung Cancer and Their Prognostic Significance according to Clinicopathological Factors and Diagnostic Markers. Int J Mol Sci 2019;20:824. [Crossref] [PubMed]
  45. Khan M, Zhao Z, Arooj S, et al. Soluble PD-1: Predictive, Prognostic, and Therapeutic Value for Cancer Immunotherapy. Front Immunol 2020;11:587460. [Crossref] [PubMed]
  46. Costantini A, Julie C, Dumenil C, et al. Predictive role of plasmatic biomarkers in advanced non-small cell lung cancer treated by nivolumab. Oncoimmunology 2018;7:e1452581. [Crossref] [PubMed]
  47. Morrell ED, Dmyterko V, Stapleton RD, et al. Soluble PD-L1 and PD-1 Concentrations Are Associated with Alveolar Inflammation and Severity of Lung Injury in Subjects with ARDS. American Journal of Respiratory and Critical Care Medicine 2019;199:A6133.
  48. Ren YY, Dong HT, Liao JY, et al. The expression and function of programmed death-ligand 1 and related cytokines in neutrophilic asthma. Ann Transl Med 2021;9:1727. [Crossref] [PubMed]
  49. Wang Y, Han H, Zhang F, et al. Immune checkpoint inhibitors alone vs immune checkpoint inhibitors-combined chemotherapy for NSCLC patients with high PD-L1 expression: a network meta-analysis. Br J Cancer 2022;127:948-56. [Crossref] [PubMed]
  50. Pérol M, Felip E, Dafni U, et al. Effectiveness of PD-(L)1 inhibitors alone or in combination with platinum-doublet chemotherapy in first-line (1L) non-squamous non-small-cell lung cancer (Nsq-NSCLC) with PD-L1-high expression using real-world data. Ann Oncol 2022;33:511-21. [Crossref] [PubMed]
Cite this article as: Kim SY, Park D, Sun P, Kim N, Lee D, Kim DK, Lee SI, Lee JE, Chung C, Kang DH. Prognostic and predictive significance of soluble programmed death ligand 1 in bronchoalveolar lavage fluid in stage IV non-small cell lung cancer. Transl Lung Cancer Res 2024;13(8):1888-1906. doi: 10.21037/tlcr-24-392

Download Citation