Open Access

Impact of the immune molecular profile of the tumor microenvironment on the prognosis of NSCLC

  • Authors:
    • Hangjie Ying
    • Qingqing Hang
    • Guoping Cheng
    • Shifeng Yang
    • Xiaojing Lai
    • Min Fang
  • View Affiliations

  • Published online on: February 14, 2023     https://doi.org/10.3892/ol.2023.13717
  • Article Number: 131
  • Copyright: © Ying et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The present study aimed to clarify the association between macrophages, tumor neo‑vessels and programmed cell death‑ligand 1 (PD‑L1) in the tumor microenvironment and the clinicopathological features of patients with non‑small cell lung cancer (NSCLC), and to explore the prognostic factors of stromal features in NSCLC. To determine this, tissue microarrays containing samples of 92 patients with NSCLC were studied using immunohistochemistry and immunofluorescence. The quantitative data demonstrated that in tumor islets, the number of CD68+ and CD206+ tumor‑associated macrophages (TAMs) was 8‑348 (median, 131) and 2‑220 (median, 52), respectively (P<0.001). In tumor stroma, the number of CD68+ and CD206+ TAMs was 23‑412 (median, 169) and 7‑358 (median, 81), respectively (P<0.001). The number of CD68+ TAMs in each location of the tumor islets and tumor stroma was significantly higher than that of CD206+ TAMs, and they were significantly correlated (P<0.0001). The quantitative density of CD105 and PD‑L1 in tumor tissues was 19‑368 (median, 156) and 9‑493 (median, 103), respectively. Survival analysis revealed that a high density of CD68+ TAMs in tumor stroma and islets and a high density of CD206+ TAMs and PD‑L1 in tumor stroma were associated with worse prognosis (both P<0.05). Collectively, the survival analysis demonstrated that the high‑density group was related to a worse prognosis regardless of combined neo‑vessels and PD‑L1 expression with the CD68+ TAMs in tumor islets and stroma, or CD206+ TAMs in tumor islets and stroma. To the best of our knowledge, the present study was the first to provide a multi‑component combined prognostic survival analysis of different types of macrophages in different regions with tumor neo‑vessels and PD‑L1, which demonstrated the importance of macrophages in tumor stroma.

Introduction

Lung cancer is the most common cause of cancer-related death, of which non-small cell lung cancer (NSCLC) comprises 85–90% (1). Despite the development of treatments, the 5-year survival rate is <15% (2). Paget's ‘seed-soil’ theory demonstrates that the tumor microenvironment (TME) has a vital role in tumor growth and progression (3). The TME is a complex and dynamic community, among which immune infiltrating cells and vascular endothelial cells are the most representative factors (4). Macrophages, important representatives of immune infiltrating cells, act as vital components of the host's defense, antigen-presenting cells and effector cells, and may be classified into the classic M1 type or the alternative M2 type (5,6). M1 type macrophages are a tumor suppressor type that participate in inflammatory response, pathogen removal and antitumor immunity, while M2 type macrophages promote the occurrence and development of tumors by inducing angiogenesis and anti-inflammation (7). Macrophages have different biological properties due to different distributions (8), so the selection of macrophage markers and tissue sites of interest affect the prognostic role of tumor-associated macrophages (TAMs) on NSCLC, to a certain extent. CD68+ is the TAM marker that is commonly considered to be a pan-macrophage marker, but it cannot distinguish between M1 and M2 subtypes (9). M2 macrophages have a variety of surface markers, including CD163, CD204 and CD206. Among them, CD206, which is expressed on the surface of most classes of macrophages and dendritic cell subgroups, is routinely used to identify the M2 subtype (10). Certain studies suggest that a higher stromal TAM density is an independent prognostic factor and leads to poor prognosis (11,12). However, M2 macrophage density is more closely related to poor prognosis than CD68+ TAMs (13,14).

Studies have confirmed that a tumor is only a minor and asymptomatic lesion prior to the formation of angiogenesis (15,16). With the formation of angiogenesis, the tumor size rapidly increases and has an enhanced potential for distant metastasis (17,18). CD105 is an endoglin used to evaluate blood vessels that is considered to only recognize abnormal blood vessels induced by tumors, and it has higher specificity than CD34 (19,20). In addition, studies have indicated that TAMs and cancer cells may promote tumor angiogenesis and metastasis (2126). Immune checkpoint inhibitors targeting the programmed cell death protein-1 (PD-1)/PD-ligand 1 (PD-L1) pathway have demonstrated an impressive clinical benefit in NSCLC and the expression of PD-L1 may partially predict the treatment effectiveness (27). TAMs are able to secrete vascular endothelial growth factors, such as IL-10, secrete IL-1 β, induce regulatory T cells, increase PD-L1 expression in tumor cells and inhibit the function of effector T cells, leading to infiltration and distant metastasis of cancer cells (28).

The present study focused on the temporal-spatial distribution and quantitative expression of CD68+ and CD206+ TAMs in two intra-tumor areas, and CD105 and PD-L1 in the TME, to investigate the heterogenic molecular profile of the TME, in an attempt to provide a guide for improving the individual treatment strategy for patients with NSCLC.

Materials and methods

Patients and samples

A total of 92 paraffin-embedded NSCLC samples were collected from Zhejiang Cancer Hospital (Hangzhou, China) between April 2008 and January 2014. Using these samples, 6 tissue microarrays (TMAs) were constructed, as previously described (29). For construction, a 3-mm core was taken from each representative tumor tissue. The study protocol was approved by The Ethics Committee of Zhejiang Cancer Hospital (Hangzhou, China) and the patients provided written informed consent regarding the use of their tissues. The main patient inclusion criteria were as follows: i) Histologically confirmed primary NSCLC; ii) patient underwent curative radical surgery. Patients who had received other anticancer treatment prior to surgery were excluded. Tumor staging was based on the 8th Tumor-Node-Metastasis classification system of the American Joint Committee on Cancer staging criteria (30). The last follow-up date was April 2017, at which point all patients had died. Overall survival (OS) time was defined as the interval from the date of surgery to the date of death. Disease-free survival (DFS) time was defined as the interval from the date of surgery to the date of disease progression.

Immunohistochemistry (IHC) and multiplexed immunofluorescence

IHC staining for macrophages marked by CD68 and CD206, and tumor neo-vessels marked by CD105 and PD-L1, was performed. In brief, TMA slides were treated by deparaffinization in xylene, hydration with graded alcohols and subjected to antigen retrieval (98°C, 20 min). The slides were then placed in 3% hydrogen peroxide for 10 min at room temperature to inactivate endogenous peroxidases. After washing three times in PBS, the slides were blocked with 2% BSA (cat. no. B2064; Sigma-Aldrich; Merck KGaA) for 30 min at room temperature, followed by incubation with primary antibodies against CD68 (1:200 dilution; cat. no. ab125212; Abcam), CD206 (1:200; cat. no. 60143–1-Ig; ProteinTech Group, Inc.), CD105 (1:500; cat. no. ab2529; Abcam) and PD-L1 (1:100; cat. no. ab205921; Abcam) at 4°C overnight. After washing in PBS, the slides were incubated with secondary antibody (1:200; Goat Anti-Rabbit IgG (horseradish peroxidase, cat. no. ab150077; Abcam) for 60 min at 37°C. The slides were visualized using Dako REAL EnVision™ (DAB; cat. no. PW017; Sangon Biotech, Co., Ltd.) and counterstained with haematoxylin for 2 min at room temperature.

Multiplexed immunofluorescence staining for CD68 and CD105 was also performed. The primary antibodies against CD68 and CD105 were mixed. Similar to IHC, after the secondary immunofluorescent antibody (1:2,000; Goat Anti-Rabbit IgG and Goat Anti-Mouse IgG (horseradish peroxidase), cat. nos. ab6721 and ab6789; Abcam) was incubated with the slides (37°C, 60 min), all slides were covered by Fluoroshield containing DAPI (Abcam) for 10 min at room temperature to identify nuclei.

Quantification of IHC and immunofluorescent staining

All slides were examined under an Olympus BX51 fluorescence microscope equipped with an Olympus DP72 camera (Olympus Corporation). Positive staining was indicated by brownish granules. The macrophages marked by CD68 and CD206 were counted in three high-power fields selected at the tumor islets and tumor stroma, and the mean number of CD68+ and CD206+ cells in these three fields was documented. Tumor islets were defined as areas where tumor cells accounted for >70% of the total cells, and tumor stroma as areas where tumor stromal cells accounted for >70% of the total cells (12). Tumor neo-vessels marked by CD105 and tumor cells expressing PD-L1 were counted in six high-power fields selected at the tumor site and the mean cell counts were documented. Pathologists defined positively expressed cells according to the Hue, Saturation, Intensity (HSI) color selection system (H=0-30; S=0-255; I=0-255), specified cell size and filtered non-specific positive color rendering noise <50 pixels, after which the software automatically counted. Positive cell counting was completed using Image-Pro Plus 6.0 software (Media Cybernetics, Inc). The cut-off value according to the median of each group was used to determine the density of infiltrating macrophages, tumor neo-vessel density and PD-L1 expression. Two independent pathologists who were blinded to the clinicopathological characteristics of all tissue specimens participated in the data evaluation.

Statistical analysis

Statistical analyses were performed with SPSS 25.0 (IBM Corp.) and R 4.2.2 (R Development Core Team). For categorical data, the χ2 test was performed. Spearman's rank correlation analysis was used to analyze the correlation between macrophages, tumor neo-vessels and PD-L1 expression. Differences in the CD68+ and CD206+ TAMs among the groups were analyzed by the Mann-Whitney U-test. The Kaplan-Meier method was used to estimate the survival curve for OS time, and the log-rank or two-stage tests were used to assess the differences in survival between groups. The Cox regression model was used to assess the influence of the binary factors in univariate and multivariate analyses. The factors with P<0.2 in the univariate analysis were included in the multivariate analysis, and the relationship between TME-related markers and prognosis was examined. A two-tailed P<0.05 was considered to indicate statistical significance.

Results

Major clinicopathological features of the 92 NSCLC cases and heterogenic TME in TMAs

Among the 92 NSCLC cases included in the present study, 71 (77.2%) were male and 21 (22.8%) were female, and they were aged 39–75 (median, 61) years. With regards to staging, there was 1 (1.1%) case of IA, 9 (9.8%) cases of IB, 7 (7.6%) of IIA, 23 (25%) of IIB, 44 (47.8%) of IIIA and 8 (8.7%) of IIIB. A total of 58 patients underwent adjuvant therapy, 40 of which received adjuvant chemotherapy, 12 received adjuvant radiotherapy and chemotherapy, 5 received adjuvant radiotherapy and 1 received tyrosine kinase inhibitor treatment. A total of 16 patients did not receive any adjuvant therapy and the therapy regimen of the remaining 18 patients was unknown. The two-stage test demonstrated that there was no significant difference in OS and DFS times between the treatment and no-treatment groups (median OS, 30 vs. 23 months, respectively; P=0.571; median DFS, 11 vs. 13 months, respectively; P=0.844; Fig. S1). There were 73 known progression or recurrence events, and the treatments of 51 patients were known. Among these 51 patients, 30 (58.8%) received two or more combined treatments, 19 (37.3%) received one treatment strategy and 2 (3.9%) did not receive any treatment. These treatments had no impact on the OS time between single treatment, multiple treatments and no-treatment (median OS, 33 vs. 36 vs. 6 months, respectively; P=0.289; Fig. S1). Other major clinicopathological characteristics data available, such as smoking status, tumor size, histology and differentiation, are presented in Tables I and II.

Table I.

Relationship between CD68+ TAMs, CD206+ TAMs and the clinicopathological features of patients with non-small cell lung cancer.

Table I.

Relationship between CD68+ TAMs, CD206+ TAMs and the clinicopathological features of patients with non-small cell lung cancer.

CD68+ TAMsaCD206+ TAMs


Tumor isletsTumor stromaTumor isletsTumor stroma




Clinicopathological featureTotal number of patientsLow n=46High n=45P-valueLow n=46High n=45P-valueLow n=47High n=45P-valueLow n=46High n=46P-value
Sex 0.193 0.759 0.390 0.804
  Female218 (38.1)13 (61.9) 10 (47.6)11 (52.4) 9 (42.9)12 (57.1) 11 (52.4)10 (47.6)
  Male7138 (54.3)32 (45.7) 36 (51.4)34 (48.6) 38 (53.5)33 (46.5) 35 (49.3)36 (50.7)
Age, years 0.908 0.760 0.982 0.294
  ≤604121 (51.2)20 (48.8) 20 (48.8)21 (51.2) 21 (51.2)20 (48.8) 23 (56.1)18 (43.9)
  >605125 (50.0)25 (50.0) 26 (52.0)24 (48.0) 26 (51.0)25 (49.0) 23 (45.1)28 (54.9)
Smoking status 0.320 0.691 0.552 1.000
  Never smoker2611 (42.3)15 (57.7) 14 (53.8)12 (46.2) 12 (46.2)14 (53.8) 13 (50.0)13 (50.0)
  Current/former6635 (53.8)30 (46.2) 32 (49.2)33 (50.8) 35 (53.0)31 (47.0) 33 (50.0)33 (50.0)
Histology 0.607 0.107 0.549 1.000
  Adenocarcinoma5428 (52.8)25 (47.2) 23 (43.4)30 (56.6) 29 (53.7)25 (46.3) 27 (50.0)27 (50.0)
  Non-adenocarcinoma3818 (50.5)20 (52.6) 23 (60.5)15 (39.5) 18 (47.4)20 (52.6) 19 (50.0)19 (50.0)
Tumor size, cm 0.423 0.606 0.879 0.381
  ≤56028 (47.5)31 (52.5) 31 (52.5)28 (47.5) 31 (51.7)29 (48.3) 32 (53.3)28 (46.7)
  >53218 (56.3)14 (43.8) 15 (46.9)17 (53.1) 16 (50.0%16 (50.0) 14 (43.8)18 (56.3)
Differentiationb 0.413 0.144 0.371 0.631
  Low165 (33.3)10 (66.7) 6 (40.0)9 (60.0) 11 (68.8)5 (31.3) 10 (62.5)6 (37.5)
  Moderate3418 (52.9)16 (47.1) 13 (38.2)21 (61.8) 17 (50.0)17 (50.0) 17 (50.0)17 (50.0)
  High3116 (51.6)15 (48.4) 19 (61.3)12 (38.7) 15 (48.4)16 (51.6) 15 (48.4)16 (51.6)
Lymph node metastasis 0.603 0.941 0.038 0.026
  Negative3014 (46.7)16 (53.3) 15 (50.0)15 (50.0) 20 (66.7)10 (33.3) 20 (66.7)26 (41.9)
  Positive6232 (52.5)29 (47.5) 31 (50.8)30 (49.2) 27 (43.5)35 (56.5) 10 (33.3)36 (58.1)
Stage 0.072 0.168 0.820 0.209
  I, II4225 (61.0)16 (39.0) 24 (58.5)17 (41.5) 22 (52.4)20 (47.6) 24 (57.1)18 (42.9)
  III5021 (42.0)29 (58.0) 22 (44.0)28 (56.0) 25 (50.0)25 (50.0) 22 (44.0)28 (56.0)

a 91 cases were detected. Values are expressed as n (%). TAM, tumor-associated macrophages.

b 81 cases recorded differentiation.

Table II.

Relationship between CD105, PD-L1 and the clinicopathological features of non-small cell lung cancer.

Table II.

Relationship between CD105, PD-L1 and the clinicopathological features of non-small cell lung cancer.

CD105-positive cellsPD-L1-positive cells


Clinicopathological featureTotal number of patientsLow n=46High n=46P-valueLow n=46High n=46P-value
Sex 0.804 0.082
  Female2111 (52.4)10 (47.6) 14 (66.7)7 (33.3)
  Male7135 (49.3)36 (50.7) 32 (45.1)39 (54.9)
Age, years 0.294 0.529
  ≤604118 (43.9)23 (56.1) 19 (46.3)22 (53.7)
  >605128 (54.9)23 (45.1) 27 (52.9)24 (47.1)
Smoking status 0.643 0.552
  Never smoked2612 (46.2)14 (53.8) 16 (61.5)10 (38.5)
  Current/former smoker6634 (51.5)32 (48.5) 30 (45.5)36 (47.0)
Histology 0.672 0.165
  Adenocarcinoma5426 (48.1)28 (51.9) 25 (46.3)29 (54.5)
  Non-adenocarcinoma3820 (52.6)18 (47.4) 21 (55.3)17 (44.7)
Tumor size, cm 0.381 0.662
  ≤56028 (46.7)32 (53.5) 31 (51.7)29 (48.3)
  >53218 (56.3)14 (43.8) 15 (46.9)17 (53.1)
Differentiationa 0.691 0.399
  Low169 (56.3)7 (43.8) 7 (43.8)9 (56.3)
  Moderate3415 (44.1)19 (55.9) 16 (47.1)18 (52.9)
  High3116 (51.6)15 (48.4) 19 (61.3)12 (38.7)
Lymph node metastasis 0.656 0.656
  Negative3014 (46.7)16 (53.3) 14 (46.7)16 (53.3)
  Positive6232 (51.6)30 (48.4) 32 (51.6)30 (48.4)
Stage 0.675 0.209
  I, II4220 (47.6)22 (52.4) 24 (57.1)18 (42.9)
  III5026 (52.0)24 (48.0) 22 (44.0)28 (56.0)

{ label (or @symbol) needed for fn[@id='tfn3-ol-25-3-13717'] } Values are expressed as n (%). PD-L1, programmed cell death-ligand 1.

a 81 cases recorded differentiation.

IHC analysis, including distribution and quantitative measurement of CD68+ TAMs, CD206+ TAMs, CD105 and PD-L1, was performed on all TMAs. In certain cases, only few CD68+ and CD206+ TAMs were present compared with other cases with high expression (‘expression’ here and throughout means the density of cells with positive expression) (Fig. 1A and B). The quantitative data demonstrated that in tumor islets, the mean numbers of CD68+ and CD206+ TAMs were 146 (median, 131; range, 8–348) and 70 (median, 52; range, 2–220), respectively (P<0.001). In tumor stroma, the mean numbers of CD68+ and CD206+ TAMs were 186 (median, 169; range, 23–412) and 103 (median, 81; range, 7–358), respectively (P<0.001). Therefore, the quantity of CD68+ TAMs was higher than that of CD206+ TAMs in tumor islets and stroma. The CD68+ and CD206+ TAMs also had a higher distribution in tumor stroma than in tumor islets (both P<0.0001). In addition, significant correlations were found between the distributions of CD68+ and CD206+ TAMs in each area (tumor islets, ρ=0.5179; tumor stroma, ρ=0.5081; both P<0.0001; Fig. 1C). According to the median of CD68+ and CD206+ TAMs in the tumor islets and stroma as the cut-off, the patients were divided into the low-density or high-density group. The survival analysis demonstrated that patients with low expression of CD68+ and CD206+ TAMs in tumor islets and stroma had a favorable prognosis (Fig. 1D).

CD105 and PD-L1 staining were mainly located in the cytoplasm or on the cell membrane of the tumor stroma. Tumor neo-vessels and PD-L1 were also heterogeneously expressed in the TME, in certain cases with low expression and other cases with high expression. The quantitative density of CD105+ cells in tumor tissues was 19–368 (median, 156). The quantitative density of PD-L1+ cells in tumor tissues was 9–493 (median, 103) (Fig. 2A and B). According to the median density of cells with positive CD105 and PD-L1 expression in tumor tissues as the cut-off, patients were divided into the low-density or high-density group. The survival analysis demonstrated that the CD105 density had no association with OS time in patients with NSCLC, whereas patients with high PD-L1 expression had a higher risk of death (Fig. 2C).

Correlations between CD68+ and CD206+ TAMs, tumor neo-vessels, PD-L1 expression and clinicopathological features

The quantitative data of CD68+ and CD206+ TAM density are provided in Table I. For CD68+ and CD206+ TAMs in tumor islets and stroma, the cut-off to classify low and high subgroups. Subgroups were as follows: in the tumor islets, less than 131 is the low CD68+TAM expression group, more than or equal to 131 is the high CD68+TAM expression group, less than 52 is the low CD206+TAM expression group, more than or equal to 52 is the high CD206+TAM expression group; in tumor stroma, less than 169 is the low CD68+TAM expression group, more than or equal to 169 is the high CD68+TAM expression group, less than 81 is the low CD206+TAM expression group, more than or equal to 81 is the high CD206+TAM expression group. For tumor neo-vessels and PD-L1 in tumor tissues, the cut-off to classify low and high subgroups was 156 and 103, respectively. In the low and high CD68+ TAM subgroups, the high tumor neo-vessel density cases were 20 (43.5%) and 26 (56.5%), respectively. In the low and high PD-L1 expression subgroups, high densities of CD68+ TAMs were observed in 18 (40.9%) and 26 cases (59.1%), respectively. In the low and high CD206+ TAM subgroups, 21 (45.7%) and 25 (54.3%) cases had high tumor neo-vessel density, respectively. In the low and high PD-L1 expression subgroups, there were 15 (33.3%) and 30 (66.7%) cases with high CD68+ TAM density, respectively. Of note, tumor neo-vessels, CD68+ TAMs and PD-L1 expression were not significantly associated with any of the clinicopathological characteristics, which indicated that these key components of the TME were independent of clinical features, including tumor size, tumor histological type, degree of differentiation, lymph node metastasis and tumor staging (Tables I and II).

In addition, CD68+ TAMs were mostly localized with tumor neo-vascularization, and the quantitative analysis demonstrated that CD68+ TAMs and CD105 had similar trends in expression (ρ=0.2401; P=0.021; Fig. 3), while there was no obvious correlation between CD206+ TAMs and CD105 (ρ=0.109; P>0.05; data not shown). Furthermore, the expression of CD68+ TAMs was significantly correlated to the expression of PD-L1 in tumor tissues (ρ=0.332; P=0.030; data not shown) and there was a significant correlation between CD206+ TAMs and PD-L1 (ρ=0.428; P=0.038; data not shown).

Prognostic significance of tumor stromal features in NSCLC

Univariate analyses demonstrated that clinical factors, such as tumor differentiation, were associated with OS time (P<0.05). PD-L1 and CD68+ TAM densities in tumor islets and stroma were also negatively associated with DFS time (both P<0.05), but there was no statistically significant independent predictor of DFS time in NSCLC (Table SI). PD-L1 and CD68+ TAM densities in tumor islets and stroma, as well as CD206+ TAM density in tumor stroma, were negatively associated with OS time (P<0.05 for all). Furthermore, tumor size, differentiation degree, high density of CD68+ TAMs in tumor islets and PD-L1 expression were statistically significant independent predictors of a poor prognosis in NSCLC (P<0.05 for all; Table III).

Table III.

Univariate and multivariate analyses of the clinicopathological factors for overall survival time of patients with non-small cell lung carcinoma.

Table III.

Univariate and multivariate analyses of the clinicopathological factors for overall survival time of patients with non-small cell lung carcinoma.

Univariate analysisMultivariate analysis


Clinicopathological factorHR(95% CI)P-valueHR(95% CI)P-value
Sex (male vs. female)0.9800.572–1.6780.941
Age, years (>60 vs. ≤60)1.2690.804–2.0050.306
Smoking status (smoker vs. never-smoker)1.1620.698–1.9320.5640.5050.160–1.5930.244
Histological type (adenocarinoma vs. non-adenocarinoma)0.7340.462–1.1680.1920.4440.167–1.1830.104
Tumor size, cm (>5 vs. ≤5)1.3730.850–2.2180.1951.9451.089–3.4750.025
Differentiation 0.045 0.033
  Low vs. moderate2.1181.087–4.1270.0280.5950.252–1.4060.237
  Low vs. high1.7521.013–3.0300.0450.3360.146–0.7740.010
Lymph node metastasis (positive vs. negative)0.9090.694–1.1900.4860.8100.384–1.7100.581
Stage (III vs. I, II)1.1230.706–1.7850.6240.7850.374–1.6450.521
CD105 expression (high vs. low)1.1060.698–1.7530.6671.0020.998–1.0050.301
PD-L1 expression (high vs. low)1.6851.066–2.6630.0251.0031.001–1.0110.010
CD68+ TAMs in tumor islets (high vs. low)1.6661.051–2.6410.0301.0061.001–1.0110.031
CD206+ TAMs in tumor islets (high vs. low)1.5800.996–2.5060.0520.9990.991–1.0070.750
CD68+ TAMs in tumor stroma (high vs. low)1.9161.202–3.0550.0061.0000.995–1.0050.941
CD206+ TAMs in tumor stroma (high vs. low)1.7411.091–2.7760.0200.9990.993–1.0050.725

[i] CI, confidence interval; HR, hazard ratio; PD-L1, programmed cell death-ligand 1; TAMs, tumor-associated macrophages.

The aforementioned key components were explored collectively to reveal the association between TME and NSCLC prognosis. For the combined group, taking the median values for tumor neo-vessels, macrophages and PD-L1 as the cut-off, the patients could be divided into groups. The groups were assigned as follows: Group 1, all components were expressed at a low level; group 2, one or two of the components was expressed at a high level; group 3, all components were expressed at a high level. The combined analysis indicated that the OS rate of group 3 was worse than that of groups 1 and 2 (Fig. 4).

Discussion

A number of conflicting results have been reported regarding the prognostic significance of TAMs, tumor neo-vessels and PD-L1/PD-1 expression in NSCLC from clinical practice (3135). The reasons for the inconsistent reports may be related to the choice of markers, as well as differences in statistical power and evaluation methods. In addition, these factors have not been observed to be adequately reliable as a single biomarker to evaluate the prognosis of patients with NSCLC (36,37). To the best of our knowledge, the present study was the first to compare TAM distribution detecting CD68+ and CD206+ in two intra-tumor areas, and to compare the distribution of TAMs, tumor neo-vessels and PD-L1 in NSCLC.

The results demonstrated that the number of CD68+ and CD206+ TAMs was higher in the tumor stroma but lower in tumor islets, and there was a correlation between the distribution of CD68+ and CD206+ TAMs in tumor islets and stroma. Furthermore, the mean numbers of CD68+ TAMs in each location of the tumor islets and stroma were significantly higher than those of CD206+ TAMs. Univariate analysis demonstrated that a large number of CD68+ and CD206+ TAMs in the tumor stroma were associated with a shorter OS time, which is consistent with the results of Li et al (12) and Dai et al (11). The former study also demonstrated that the tumor stroma is the most suitable intra-tumor area for evaluating the relationship between TAMs and prognosis of NSCLC cases. The present study also found that CD206+ stromal TAMs, CD105 and PD-L1 had a relationship with prognosis. When the number of positive cells in each section (tumor islets and tumor stroma) was summed, and CD68+ or CD206+ TAMs were combined with the other two key components, the high density of the three components may indicate a worse prognosis within 30 months of surgery.

New blood vessels support rapid tumor tissue growth, providing nutrients and oxygen to thriving tumor cells (38). However, in the present study, tumor neo-vessel density was not significantly correlated with the prognosis of patients with NSCLC, but there was a significant correlation between CD68+ and CD105. These results suggested that macrophages have a significant role in tumor neo-vessels in the process of cancer invasion and metastasis. TAMs are considered to be ‘angiogenesis switches’ and a key factor leading to a proangiogenic environment (26,39).

In the present study, it was observed that PD-L1 expression was correlated with the prognosis of NSCLC and may be used as an independent prognostic factor for patients with NSCLC. PD-L1 mediates immunosuppressive signals and certain studies suggest that PD-L1 upregulation is associated with longer survival time in early NSCLC (40), breast carcinoma (41), gastric cancer (42) and colorectal cancer (43). However, another study has indicated that there is no association between PD-L1 expression and OS (44). A number of previous studies have reported that high PD-L1 expression is associated with poor prognosis in NSCLC (4547). In these studies, the definition of PD-L1+ or high density was different, leading to difficulties in concluding on the relationship between PD-L1 expression and NSCLC prognosis. Evidence suggests that PD-L1 upregulation is an adaptive mechanism and may be a response of tumor cells to host immune pressure (48). It is also understood that PD-L1 expression is related to the endogenous immune response, such as tumor-infiltrating lymphocytes in NSCLC and indoleamine 2,3-dioxygenase-1 expression by dendritic cells (49). It may therefore be suggested that any possible prognostic significance is not directly related to a single immune signal but to the overall balance between the host's antitumor immune response and tumor-mediated immunosuppression.

The present study demonstrated that the expression of PD-L1 in cancer cells was correlated with the density of CD68+ and CD206+ TAMs. M2 type macrophages have a weak antigen-presenting capacity and suppress T-cell immune responses by releasing immunosuppressive factors, such as TGF-β and IL-10 (50). In the hypoxic TME, the expression of certain immunosuppressive factors, such as prostaglandin E2 (PGE-2) and IL-10, not only inhibits the activation of M1 macrophages, but also converts the generated M1 type to the M2 type (51).

In conclusion, the distribution of stromal macrophages and M2 type TAMs are important factors affecting other key components in the TME. The present study demonstrated that the different immunological molecular profiles of the TME were associated with the prognosis of patients with NSCLC. There are also certain limitations to the present study. The number of cases was small and treatments of patients after postoperative recurrent-metastasis is incomplete. Further studies with a larger sample size are required to be conducted to gain a deeper understanding and explanation of this mechanism. To the best of our knowledge, the present study was the first to provide a multi-component combined prognostic survival analysis of different types of macrophages in different regions with tumor neo-vessels and PD-L1, and the combined analysis of key components may improve the prediction of the prognosis.

Supplementary Material

Supporting Data
Supporting Data

Acknowledgements

Not applicable.

Funding

This research was supported by the grants of The National Natural Science Foundation of China (grant no. 81703018) and The Zhejiang Medical and Health Science and Technology Project (grant nos. 2020KY466 and 2022RC110), both to MF.

Availability of data and materials

The datasets generated and/or analyzed during the current study are not publicly available due to restrictions applied by The Cancer Hospital of the University of Chinese Academy (Zhejiang Cancer Hospital, Hangzhou, China) but are available from the corresponding author on reasonable request.

Authors' contributions

Conception and design: MF and XL. Administrative support: MF and XL. Provision of study materials or patients: MF and XL. Collection and assembly of data: MF, QH and HY. Data analysis and interpretation: MF, QH, HY, GC and SY. Manuscript writing: HY and QH. HY and MF confirm the authenticity of all the raw data. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

All procedures performed in the present study involving human participants were in accordance with the Declaration of Helsinki (as revised in 2013). The study was approved by the Institutional Ethics committee of Zhejiang Cancer Hospital (Hangzhou, China; approval no. IRB-2021-111). Written informed consent was obtained from each patient.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2020. CA Cancer J Clin. 70:7–30. 2020. View Article : Google Scholar : PubMed/NCBI

2 

Liu X, Shen SL and Wang YP: Current ststus of comprehensive interventional therapy for advanced non-small cell lung cancer. Zhonghua Fei Bu Ji Bing Za Zhi (Dian Zi Ban). 10:486–489. 2017.(In Chinese).

3 

Paget S: The distribution of secondary growths in cancer of the breast. 1889. Cancer Metastasis Rev. 8:98–101. 1989.PubMed/NCBI

4 

Catalano V, Turdo A, Di Franco S, Dieli F, Todaro M and Stassi G: Tumor and its microenvironment: A synergistic interplay. Semin Cancer Biol. 23:522–532. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Mosser DM and Edwards JP: Exploring the full spectrum of macrophage activation. Nat Rev Immunol. 8:958–969. 2008. View Article : Google Scholar : PubMed/NCBI

6 

Schmieder A, Michel J, Schonhaar K, Goerdt S and Schledzewski K: Differentiation and gene expression profile of tumor-associated macrophages. Semin Cancer Biol. 22:289–297. 2012. View Article : Google Scholar : PubMed/NCBI

7 

Pollard JW: Tumour-educated macrophages promote tumour progression and metastasis. Nat Rev Cancer. 4:71–78. 2004. View Article : Google Scholar : PubMed/NCBI

8 

Joshi N, Walter JM and Misharin AV: Alveolar macrophages. Cell Immunol. 330:86–90. 2018. View Article : Google Scholar : PubMed/NCBI

9 

Falini B, Flenghi L, Pileri S, Bigerna B, Durkop H, Eitelbach F, Thiele J, Pacini R and Cavaliere A: PG-M1: A new monoclonal antibody directed against a fixative-resistant epitope on the macrophage-restricted form of the CD68 molecule. Am J Pathol. 142:1359–1372. 1993.PubMed/NCBI

10 

Gordon S: Alternative activation of macrophages. Nat Rev Immunol. 3:23–35. 2003. View Article : Google Scholar : PubMed/NCBI

11 

Dai F, Liu L, Che G, Yu N, Pu Q, Zhang S, Ma J, Ma L and You Z: The number and microlocalization of tumor-associated immune cells are associated with patient's survival time in non-small cell lung cancer. BMC Cancer. 10:2202010. View Article : Google Scholar : PubMed/NCBI

12 

Li Z, Maeda D, Yoshida M, Umakoshi M, Nanjo H, Shiraishi K, Saito M, Kohno T, Konno H, Saito H, et al: The intratumoral distribution influences the prognostic impact of CD68- and CD204-positive macrophages in non-small cell lung cancer. Lung Cancer. 123:127–135. 2018. View Article : Google Scholar : PubMed/NCBI

13 

Komohara Y, Jinushi M and Takeya M: Clinical significance of macrophage heterogeneity in human malignant tumors. Cancer Sci. 105:1–8. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Sumitomo R, Hirai T, Fujita M, Murakami H, Otake Y and Huang CL: M2 tumor-associated macrophages promote tumor progression in non-small-cell lung cancer. Exp Ther Med. 18:4490–4498. 2019.PubMed/NCBI

15 

Hicklin DJ and Ellis LM: Role of the vascular endothelial growth factor pathway in tumor growth and angiogenesis. J Clin Oncol. 23:1011–1027. 2005. View Article : Google Scholar : PubMed/NCBI

16 

Lin CY, Siow TY, Lin MH, Hsu YH, Tung YY, Jang T, Recht L and Chang C: Visualization of rodent brain tumor angiogenesis and effects of antiangiogenic treatment using 3D ΔR2-µMRA. Angiogenesis. 16:785–793. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Sajib S, Zahra FT, Lionakis MS, German NA and Mikelis CM: Mechanisms of angiogenesis in microbe-regulated inflammatory and neoplastic conditions. Angiogenesis. 21:1–14. 2018. View Article : Google Scholar : PubMed/NCBI

18 

Li S, Xu HX, Wu CT, Wang WQ, Jin W, Gao HL, Li H, Zhang SR, Xu JZ, Qi ZH, et al: Angiogenesis in pancreatic cancer: Current research status and clinical implications. Angiogenesis. 22:15–36. 2019. View Article : Google Scholar : PubMed/NCBI

19 

Weidner N, Folkman J, Pozza F, Bevilacqua P, Allred EN, Moore DH, Meli S and Gasparini G: Tumor angiogenesis: A new significant and independent prognostic indicator in early-stage breast carcinoma. J Natl Cancer Inst. 84:1875–1887. 1992. View Article : Google Scholar : PubMed/NCBI

20 

Akagi K, Ikeda Y, Sumiyoshi Y, Kimura Y, Kinoshita J, Miyazaki M and Abe T: Estimation of angiogenesis with anti-CD105 immunostaining in the process of colorectal cancer development. Surgery. 131:S109–S113. 2002. View Article : Google Scholar : PubMed/NCBI

21 

Wu TH, Li YY, Wu TL, Chang JWC, Chou WC, Hsieh LL, Chen JR and Yeh KY: Culture supernatants of different colon cancer cell lines induce specific phenotype switching and functional alteration of THP-1 cells. Cell Immunol. 290:107–115. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Yeo EJ, Cassetta L, Qian BZ, Lewkowich I, Li JF, Stefater JA III, Smith A, Wiechmann LS, Wang Y, Pollard JW and Lang RA: Myeloid WNT7b mediates the angiogenic switch and metastasis in breast cancer. Cancer Res. 74:2962–2973. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Ohba K, Miyata Y, Kanda S, Koga S, Hayashi T and Kanetake H: Expression of urokinase-type plasminogen activator, urokinase-type plasminogen activator receptor and plasminogen activator inhibitors in patients with renal cell carcinoma: Correlation with tumor associated macrophage and prognosis. J Urol. 174:461–465. 2005. View Article : Google Scholar : PubMed/NCBI

24 

Allavena P, Sica A, Solinas G, Porta C and Mantovani A: The inflammatory micro-environment in tumor progression: The role of tumor-associated macrophages. Crit Rev Oncol Hematol. 66:1–9. 2008. View Article : Google Scholar : PubMed/NCBI

25 

Han B, Li K, Zhao Y, Li B, Cheng Y, Zhou J, Lu Y, Shi Y, Wang Z, Jiang L, et al: Anlotinib as a third-line therapy in patients with refractory advanced non-small-cell lung cancer: A multicentre, randomised phase II trial (ALTER0302). Br J Cancer. 118:654–661. 2018. View Article : Google Scholar : PubMed/NCBI

26 

Murdoch C, Muthana M, Coffelt SB and Lewis CE: The role of myeloid cells in the promotion of tumour angiogenesis. Nat Rev Cancer. 8:618–631. 2008. View Article : Google Scholar : PubMed/NCBI

27 

Anagnostou VK and Brahmer JR: Cancer immunotherapy: A future paradigm shift in the treatment of non-small cell lung cancer. Clin Cancer Res. 21:976–984. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Noman MZ, Desantis G, Janji B, Hasmim M, Karray S, Dessen P, Bronte V and Chouaib S: PD-L1 is a novel direct target of HIF-1α, and its blockade under hypoxia enhanced MDSC-mediated T cell activation. J Exp Med. 211:781–790. 2014. View Article : Google Scholar : PubMed/NCBI

29 

Dancau AM, Simon R, Mirlacher M and Sauter G: Tissue microarrays. Methods Mol Biol. 576:49–60. 2010. View Article : Google Scholar : PubMed/NCBI

30 

Goldstraw P, Chansky K, Crowley J, Rami-Porta R, Asamura H, Eberhardt WRR, Nicholson AG, Groome P, Mitchell A, Bolejack V, et al: The IASLC lung cancer staging project: Proposals for revision of the TNM stage groupings in the forthcoming (Eighth) edition of the TNM Classification for lung cancer. J Thorac Oncol. 11:39–51. 2016. View Article : Google Scholar : PubMed/NCBI

31 

Zhang QW, Liu L, Gong CY, Shi HS, Zeng YH, Wang XZ, Zhao YW and Wei YQ: Prognostic significance of tumor-associated macrophages in solid tumor: A meta-analysis of the literature. PLoS One. 7:e509462012. View Article : Google Scholar : PubMed/NCBI

32 

Du ZY, Shi MH, Ji CH and Yu Y: Serum pleiotrophin could be an early indicator for diagnosis and prognosis of non-small cell lung cancer. Asian Pac J Cancer Prev. 16:1421–1425. 2015. View Article : Google Scholar : PubMed/NCBI

33 

Keskin S, Kutluk AC and Tas F: Prognostic and predictive role of angiogenic markers in non- small cell lung cancer. Asian Pac J Cancer Prev. 20:733–736. 2019. View Article : Google Scholar : PubMed/NCBI

34 

Huang W, Ran R, Shao B and Li H: Prognostic and clinicopathological value of PD-L1 expression in primary breast cancer: A meta-analysis. Breast Cancer Res Treat. 178:17–33. 2019. View Article : Google Scholar : PubMed/NCBI

35 

Yagi T, Baba Y, Ishimoto T, Iwatsuki M, Miyamoto Y, Yoshida N, Watanabe M and Baba H: PD-L1 Expression, tumor-infiltrating lymphocytes, and clinical outcome in patients with surgically resected esophageal cancer. Ann Surg. 269:471–478. 2019. View Article : Google Scholar : PubMed/NCBI

36 

Patel SP and Kurzrock R: PD-L1 expression as a predictive biomarker in cancer immunotherapy. Mol Cancer Ther. 14:847–856. 2015. View Article : Google Scholar : PubMed/NCBI

37 

Brahmer J, Reckamp KL, Baas P, Crinò L, Eberhardt WEE, Poddubskaya E, Antonia S, Pluzanski A, Vokes EE, Holgado E, et al: Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung cancer. New Eng J Med. 373:123–135. 2015. View Article : Google Scholar : PubMed/NCBI

38 

Muz B, de la Puente P, Azab F and Azab AK: The role of hypoxia in cancer progression, angiogenesis, metastasis, and resistance to therapy. Hypoxia (Auckl). 3:83–92. 2015. View Article : Google Scholar : PubMed/NCBI

39 

Mazzieri R, Pucci F, Moi D, Zonari E, Ranghetti A, Berti A, Politi LS, Gentner B, Brown JL, Naldini L and De Palma M: Targeting the ANG2/TIE2 axis inhibits tumor growth and metastasis by impairing angiogenesis and disabling rebounds of proangiogenic myeloid cells. Cancer Cell. 19:512–526. 2011. View Article : Google Scholar : PubMed/NCBI

40 

Cooper WA, Tran T, Vilain RE, Madore J, Selinger CI, Kohonen-Corish M, Yip P, Yu B, O'Toole SA, McCaughan BC, et al: PD-L1 expression is a favorable prognostic factor in early stage non-small cell carcinoma. Lung Cancer. 89:181–188. 2015. View Article : Google Scholar : PubMed/NCBI

41 

Schalper KA, Velcheti V, Carvajal D, Wimberly H, Brown J, Pusztai L and Rimm DL: In situ tumor PD-L1 mRNA expression is associated with increased TILs and better outcome in breast carcinomas. Clin Cancer Res. 20:2773–2782. 2014. View Article : Google Scholar : PubMed/NCBI

42 

Zheng Z, Bu Z, Liu X, Zhang L, Li Z, Wu A, Wu X, Cheng X, Xing X, Du H, et al: Level of circulating PD-L1 expression in patients with advanced gastric cancer and its clinical implications. Chin J Cancer Res. 26:104–111. 2014.PubMed/NCBI

43 

Droeser RA, Hirt C, Viehl CT, Frey DM, Nebiker C, Huber X, Zlobec I, Eppenberger-Castori S, Tzankov A, Rosso R, et al: Clinical impact of programmed cell death ligand 1 expression in colorectal cancer. Eur J Cancer. 49:2233–2242. 2013. View Article : Google Scholar : PubMed/NCBI

44 

Sorensen SF, Zhou W, Dolled-Filhart M and Georgsen J: 1328ppd-L1 expression and survival among advanced non-small cell lung cancer (Nsclc) patients treated with chemotherapy. Ann Oncol. 25 (Suppl 4):iv467. 2014. View Article : Google Scholar : PubMed/NCBI

45 

Li H, Xu Y, Wan B, Song Y, Zhan P, Hu Y, Zhang Q, Zhang F, Liu H, Li T, 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. 8:429–449. 2019. View Article : Google Scholar : PubMed/NCBI

46 

Keller MD, Neppl C, Irmak Y, Hall SR, Schmid RA, Langer R and Berezowska S: Adverse prognostic value of PD-L1 expression in primary resected pulmonary squamous cell carcinomas and paired mediastinal lymph node metastases. Mod Pathol. 31:101–110. 2018. View Article : Google Scholar : PubMed/NCBI

47 

Igawa S, Sato Y, Ryuge S, Ichinoe M, Katono K, Hiyoshi Y, Otani S, Nagashio R, Nakashima H, Katagiri M, et al: Impact of PD-L1 expression in patients with surgically resected non-small-cell lung cancer. Oncology. 92:283–290. 2017. View Article : Google Scholar : PubMed/NCBI

48 

Taube JM, Anders RA, Young GD, Xu H, Sharma R, McMiller TL, Chen S, Klein AP, Pardoll DM, Topalian SL and Chen L: Colocalization of inflammatory response with B7-h1 expression in human melanocytic lesions supports an adaptive resistance mechanism of immune escape. Sci Transl Med. 4:127ra1372012. View Article : Google Scholar : PubMed/NCBI

49 

Mandarano M, Bellezza G, Belladonna ML, Van den Eynde BJ, Chiari R, Vannucci J, Mondanelli G, Ludovini V, Ferri I, Bianconi F, et al: Assessment of TILs, IDO-1, and PD-L1 in resected non-small cell lung cancer: An immunohistochemical study with clinicopathological and prognostic implications. Virchows Arch. 474:159–168. 2019. View Article : Google Scholar : PubMed/NCBI

50 

Sumitomo R, Hirai T, Fujita M, Murakami H, Otake Y and Huang CL: PD-L1 expression on tumor-infiltrating immune cells is highly associated with M2 TAM and aggressive malignant potential in patients with resected non-small cell lung cancer. Lung Cancer. 136:136–144. 2019. View Article : Google Scholar : PubMed/NCBI

51 

Wen ZF, Liu H, Gao R, Zhou M, Ma J, Zhang Y, Zhao J, Chen Y, Zhang T, Huang F, et al: Tumor cell-released autophagosomes (TRAPs) promote immunosuppression through induction of M2-like macrophages with increased expression of PD-L1. J Immunother Cancer. 6:1512018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2023
Volume 25 Issue 3

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ying H, Hang Q, Cheng G, Yang S, Lai X and Fang M: Impact of the immune molecular profile of the tumor microenvironment on the prognosis of NSCLC. Oncol Lett 25: 131, 2023
APA
Ying, H., Hang, Q., Cheng, G., Yang, S., Lai, X., & Fang, M. (2023). Impact of the immune molecular profile of the tumor microenvironment on the prognosis of NSCLC. Oncology Letters, 25, 131. https://doi.org/10.3892/ol.2023.13717
MLA
Ying, H., Hang, Q., Cheng, G., Yang, S., Lai, X., Fang, M."Impact of the immune molecular profile of the tumor microenvironment on the prognosis of NSCLC". Oncology Letters 25.3 (2023): 131.
Chicago
Ying, H., Hang, Q., Cheng, G., Yang, S., Lai, X., Fang, M."Impact of the immune molecular profile of the tumor microenvironment on the prognosis of NSCLC". Oncology Letters 25, no. 3 (2023): 131. https://doi.org/10.3892/ol.2023.13717