Open Access

Differential expression of PD‑L1 and PD‑L2 is associated with the tumor microenvironment of TILs and M2 TAMs and tumor differentiation in non‑small cell lung cancer

  • Authors:
    • Ryota Sumitomo
    • Cheng-Long Huang
    • Masaaki Fujita
    • Hiroyuki Cho
    • Hiroshi Date
  • View Affiliations

  • Published online on: February 15, 2022     https://doi.org/10.3892/or.2022.8284
  • Article Number: 73
  • Copyright: © Sumitomo 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

To improve the treatment strategy of immune‑checkpoint inhibitors for non‑small cell lung cancer (NSCLC), a comprehensive analysis of programmed death‑ligand (PD‑L)1 and PD‑L2 expression is clinically important. The expression of PD‑L1 and PD‑L2 on both tumor cells (TCs) and tumor‑infiltrating immune cells (ICs) was investigated, with respect to tumor‑infiltrating lymphocytes (TILs) and M2 tumor‑associated macrophages (TAMs), which are key components of the tumor microenvironment, in 175 patients with resected NSCLC. The TIL and M2 TAM densities were associated with the expression of PD‑L1 on the two TCs (both P<0.0001) and ICs (both P<0.0001). The TIL and M2 TAM densities were also associated with the expression of PD‑L2 on both TCs (P=0.0494 and P=0.0452, respectively) and ICs (P=0.0048 and P=0.0125, respectively). However, there was no correlation between the percentage of PD‑L1‑positive TCs and the percentage of PD‑L2‑positive TCs (r=0.019; P=0.8049). Meanwhile, tumor differentiation was significantly associated with the PD‑L1 expression on TCs and ICs (P=0.0002 and P<0.0001, respectively). By contrast, tumor differentiation was inversely associated with the PD‑L2 expression on both TCs and ICs (P=0.0260 and P=0.0326, respectively). In conclusion, the combined evaluation of PD‑L1 and PD‑L2 expression could be clinically important in the treatment strategy of immune‑checkpoint inhibitors in patients with NSCLC. In particular, the evaluation of PD‑L2 expression may be necessary for patients with PD‑L1‑negative NSCLC.

Introduction

Non-small cell lung cancer (NSCLC), accounting for ~85% of all cases of lung cancer, remains to be the leading cause of cancer-related mortality worldwide, despite the availability of advanced cytotoxic chemotherapies and molecular-targeted therapies, such as EGFR-tyrosine kinase inhibitors (1,2). However, recently, agents that target the programmed death-1 (PD-1)/programmed death-ligand 1 (PD-L1) axis, such as immune-checkpoint inhibitors, have been widely used as a standard treatment for patients with metastatic NSCLC (35). Pembrolizumab, an anti-PD-1 antibody, has been approved as monotherapy in patients with tumors that have highly upregulated expression of PD-L1 on tumor cells (TCs) (4). This finding made PD-L1 testing a mandatory diagnostic test during treatment planning in patients with NSCLC. Furthermore, effective clinical response to atezolizumab, an anti-PD-L1 antibody, is observed not only in patients with tumors with high PD-L1 expression on TCs, but also in patients with tumors that expressed high levels of PD-L1 on tumor-infiltrating immune cells (ICs) (6). These observations suggest that the PD-L1 expression not only on TCs but also ICs serves an important role in regulating the anti-tumor T cell response. In addition, the PD-L1 expression on TCs and ICs is reported to be affected by microenvironment stimuli, including tumor-infiltrating lymphocytes (TILs) and M2 tumor-associated macrophages (TAMs) (7,8).

On the other hand, recent clinical studies report that PD-L2, another PD-1 ligand, is also widely expressed in numerous types of cancer, including NSCLC (914). Several studies reveal that PD-L2 is also expressed by both TCs and various ICs, depending on the microenvironment stimuli (1517). In addition, experimental studies report that PD-L2-expressing TCs are resistant to treatment with anti-PD-L1 antibody alone and that this resistance is overcome by an anti-PD-1 antibody or in combination with an anti-PD-L2 antibody (18,19). A clinical study reports that clinical response to pembrolizumab in patients with head and neck squamous cell carcinoma may be related partly to blockade of PD-1/PD-L2 interactions (20). However, the clinical significance of the PD-L2 expression in NSCLC is still controversial (7,911).

Taken together, to improve the treatment strategy of immune-checkpoint inhibitors for patients with NSCLC, a comprehensive analysis of the biological mechanisms and clinical significance of PD-L1 and PD-L2 expression was considered to be clinically important. Therefore, a the present study was performed to evaluate the expression of PD-L1 and PD-L2 on both TCs and ICs in patients with NSCLC. In addition, the association between TILs and M2 TAMs, which are key components of the tumor microenvironment (TME), on the expression of PD-L1 and PD-L2 was also analyzed.

Materials and methods

Patients

Consecutive 175 patients with NSCLC, who underwent surgery at the Department of Thoracic Surgery, Kitano Hospital (Osaka, Japan) between November 2011 and December 2014, were included. The present study was approved by the Ethics Committee (approval no. P181200300) and written informed consent was provided from each patient. Pathological staging was determined using the 8th Tumor Node Metastasis (TNM) classification system (21). The histological type and the grade of differentiation of the tumors were determined according to the classification system developed by the World Health Organization (22). The medical records and histopathological diagnosis from the patients were fully documented.

Immunohistochemistry

Immunohistochemical studies were performed to evaluate the TIL distribution by CD3 staining, the M2 TAM distribution by CD163 staining (8,23), PD-L1 expression on TCs and ICs by the Ventana SP263 assay and PD-L2 expression on TCs and ICs, using the Ventana BenchMark GX system (Ventana Medical Systems; Roche Diagnostics), according to the recommended protocol. The following antibodies were used: Rabbit monoclonal anti-human CD3 (clone 2GV6; prediluted; Ventana Medical Systems; Roche Diagnostics), PD-L1 (clone SP263; prediluted; Ventana Medical Systems; Roche Diagnostics) (24) and PD-L2 (cat. no. 18251-1-AP; 1:200; ProteinTech Group, Inc.) and mouse monoclonal anti-human CD163 (clone 760-4437; prediluted; Ventana Medical Systems; Roche Diagnostics). The tissues were fixed in 10% neutral-buffered formalin for 24 h at room temperature. After dehydration in graded ethanol series followed by xylene at room temperature, the tissues were embedded in paraffin at 60°C. Formalin-fixed paraffin-embedded tissue was cut into 4-µm sections and mounted on poly-L-lysine-coated slides. The sections were deparaffinized and rehydrated using EZ Prep (Ventana Medical Systems; Roche Diagnostics) at 75°C. Antigen retrieval was performed using Cell Conditioner 1 (Ventana Medical Systems; Roche Diagnostics) for 64 min at 100°C against CD3, PD-L1 and PD-L2 and 32 min at 100°C against CD163. The sections were then incubated with the specific primary antibody for 16 min at 37°C against CD3, CD163 and PD-L1 and 2 h at 37°C for PD-L2. Subsequently, the sections were treated with the OptiView HQ Linker (Ventana Medical Systems; Roche Diagnostics) for 8 min at 37°C and the OptiView HRP Multimer (Ventana Medical Systems; Roche Diagnostics) for 8 min at 37°C. Finally, counterstaining was performed with Mayer's hematoxylin and Scott's tap water bluing reagent at 37°C.

The evaluation of the stained tissue sections was performed by two investigators (RS and CLH) blinded to the study. The cases with discrepancies were jointly re-evaluated until a consensus was reached. For CD3 and CD163 staining, the five most representative high-power fields (magnification, ×400; 0.0625 mm2) of the tumor stroma were selected. Tumor stroma was defined as the area where tumor stromal cells accounted for >70% of the total cells (25). The number of CD3-positive cells and CD163-positive cells in each area was counted and the average number of fields in each area was calculated. Finally, the CD3-positive cell density in the tumor stroma (TIL density) and the CD163-positive macrophage density in the tumor stroma (M2 TAM density) were defined as the cell number per mm2. PD-L1 and PD-L2 expression was calculated as the percentage of membrane staining on TCs or ICs, respectively, in the overall area of the tumor, regardless of intensity.

Statistical analysis

The statistical significances regarding continuous variables were assessed using either a t-test, ANOVA with Bonferroni/Dunn post hoc test or Pearson's correlation coefficient. Categorical variables were compared using a χ2 test. Statistical analyses were performed using SPSS v23.0 for Windows (IBM Corp.). All P-values were based on the two-sided statistical analysis and P<0.05 was considered to indicate a statistically significant difference.

Results

Distribution and clinical significance of TILs among resected NSCLCs

Immunohistochemistry for CD3 exhibited a membranous and cytoplasmic staining pattern (Fig. 1A, C and E). The TIL density varied among the 175 tumor tissues (mean ± standard deviation, 948.1±890.6; Table I). As the TIL density cut-off (524) demonstrated the highest significance with respect to the percentage of PD-L1-positive TCs and PD-L2-positive TCs, the sample was classified as TIL-high when the TIL density was >524. A total of 71 tumors (40.6%) were classified as TIL-low and 104 tumors (59.4%) were classified as TIL-high. With respect to tumor histology, the TIL density was significantly higher in squamous cell carcinoma compared with that in adenocarcinoma (P=0.0206). In addition, with respect to tumor differentiation, the TIL density was significantly higher in moderately and poorly differentiated tumors compared with that in well-differentiated tumors (P=0.0130).

Table I.

Distributions of TIL and M2 TAM density among NSCLC patients according to clinicopathological characteristics.

Table I.

Distributions of TIL and M2 TAM density among NSCLC patients according to clinicopathological characteristics.

TIL M2 TAM


nlowhighMean ± standard deviationP-valuelowhighMean ± standard deviationP-value
Smoking
  Non-smoker903753981.2±931.30.6139a5931357.7±362.00.3782a
  Smoker853451913.0±849.6 4936408.7±402.4
Tumor status
  T0963712.7±854.20.3270b81205.3±308.80.1526b
T1844044874.8±847.4 5727353.7±378.5
  T2, T3, T48225571048.9±934.9 4339431.4±387.6
Nodal status
  N01376176890.2±884.10.1032a8849346.2±337.10.0165a,c
  N1, N2, N33810281156.5±894.3 2018513.4±495.7
Pathological stage
  0862641.6±884.30.0632b80107.5±102.70.0388b,c
  I1115160842.8±816.2 7140358.2±352.2
  II267191264.0±1190.4 1214517.9±427.0
  III307231145.4±790.0 1713428.2±450.5
Differentiation
  Well351817765.4±717.50.0130b,c278252.5±270.90.0015b,c
  Moderately1054659881.0±864.8 6837363.7±361.7
  Poorly357281331.8±1028.3 1322568.8±467.5
Histology
  Ad1416180857.5±801.00.0206b,c9843336.1±356.20.0036b,c
  Sq279181356.1±1244.2 1017594.2±474.7
  La7161198.6±556.9 07499.9±154.0
Total number of patients17571104948.1±890.6 10867382.5±381.9

a P-value determined using a t-test.

b P-value determined using ANOVA followed by a Bonferroni/Dunn test.

c P<0.05. TIL, tumor-infiltrating lymphocyte; TAM, tumor-associated macrophage; NSCLC, non-small-cell lung cancer; Ad, adenocarcinoma; Sq, squamous cell carcinoma; La, large cell carcinoma.

Distribution and clinical significance of M2 TAMs among resected NSCLCs

Immunohistochemistry for CD163 exhibited a membranous and cytoplasmic staining pattern (Fig. 1G). The M2 TAM density also varied among the 175 tumor tissues (mean ± standard deviation, 382.5±381.9; Table I). There was a weak correlation between TIL and M2 TAM densities (r=0.262; P=0.0004; Fig. 2). The sample was classified as M2 TAM-high when the M2 TAM density was >380 due to the highest significance in the level of C-reactive protein, a marker of the inflammatory response, as previously reported (23). A total of 108 tumors (61.7%) were classified as M2 TAM-low and 67 tumors (38.3%) were classified as M2 TAM-high. The M2 TAM density was also significantly higher in squamous cell carcinoma compared with that in adenocarcinoma (P=0.0036). The M2 TAM density was also significantly higher in poorly differentiated tumors compared with that in well- and moderately differentiated tumors (P=0.0015). Furthermore, the M2 TAM density was significantly higher in node-positive tumors and advanced stage (P=0.0165 and P=0.0388, respectively).

Expression of PD-L1 on TCs and ICs with respect to TILs and M2 TAMs

The percentage of PD-L1-positive TCs varied among the 175 tumor tissues (mean ± standard deviation; 15.6±27.0%; Fig. 1B). PD-L1 expression on TCs was significantly higher in squamous cell carcinoma compared with that in adenocarcinoma (P=0.0001).

The percentage of PD-L1-positive ICs also varied (mean ± standard deviation, 9.4±10.9%; Fig. 1D). PD-L1 expression on ICs was also significantly higher in squamous cell carcinoma compared with that in adenocarcinoma (P=0.0173). Furthermore, PD-L1 expression on ICs was significantly associated with tumor status, nodal status and pathological stage (P=0.0104, P=0.0166 and P=0.0027, respectively).

With respect to TILs, the TIL density was significantly correlated with the percentage of PD-L1-positive TCs (r=0.365; P<0.001). The percentage of PD-L1-positive TCs was significantly higher in the TIL-high group compared with that in the TIL-low group (22.2±30.8% vs. 6.1±16.0%; P<0.0001; Fig. 3A). Furthermore, the TIL density was also significantly correlated with the percentage of PD-L1-positive ICs (r=0.751; P<0.001). The percentage of PD-L1-positive ICs was significantly higher in the TIL-high group compared with that in the TIL-low group (14.4±11.2% vs. 1.9±3.5%; P<0.0001; Fig. 3B).

As previously reported (8), with respect to M2 TAMs, the M2 TAM density was significantly correlated with the percentage of PD-L1-positive TCs (r=0.389; P<0.001). The percentage of PD-L1-positive TCs was significantly higher in the M2 TAM-high group compared with that in the M2 TAM-low group (26.5±32.3% vs. 8.9±20.5%; P<0.0001; Fig. 3C). Furthermore, the M2 TAM density was significantly correlated with the percentage of PD-L1-positive ICs (r=0.375; P<0.001). The percentage of PD-L1-positive ICs was significantly higher in the M2 TAM-high group compared with that in the M2 TAM-low group (14.2±12.0% vs. 6.4±8.8%; P<0.0001; Fig. 3D).

With respect to the combined evaluation of TILs and M2 TAMs, the percentage of PD-L1-positive TCs was significantly the highest in both the TIL-high and M2 TAM-high tumors (Fig. 3E). The percentage of PD-L1-positive ICs was also significantly the highest in both the TIL-high and M2 TAM-high tumors (Fig. 3F).

Expression of PD-L2 on TCs and ICs with respect to TILs and M2 TAMs

The percentage of PD-L2-positive TCs varied among the 175 tumor tissues (mean ± standard deviation, 14.6±22.9%; Fig. 1F) and there were <1% in 70 (40.0%) tumors, 1–49% in 84 (48.0%) tumors and ≥50% in 21 (12.0%) tumors (Table II). The percentage of PD-L2-positive ICs also varied (mean ± standard deviation, 12.5±18.4%; Fig. 1H) and there were <1% in 50 (28.6%) tumors, 1–9% in 69 (39.4%) tumors and ≥10% in 56 (32.0%) tumors (Table II).

Table II.

Distributions of PD-L1 and PD-L2 expressions among NSCLC patients according to clinicopathological characteristics.

Table II.

Distributions of PD-L1 and PD-L2 expressions among NSCLC patients according to clinicopathological characteristics.

PD-L1 expression on TCsPD-L1 expression on ICsPD-L2 expression on TCsPD-L2 expression on ICs




n<1%1-49%≥50%P-value<1%1-9%≥10%P-value<1%1-49%≥50%P-value<1%1-9%≥10%P-value
Smoking
  Non-smoker905025150.5946a2732310.9917a3047130.1596a2435310.7427a
  Smoker85511816 253129 40378 263425
Tumor status
  T096210.1414a5310.0104a,b2700.0218a,b2160.1569a
  T184561513 253821 264414 223428
  T2, T3, T482392617 222238 42337 263422
Nodal status
  N01378433200.0848a4651400.0166a,b5664170.8082a4449440.0852a
  N1, N2, N338171011 61220 14204 62012
Pathological stage
  086200.3067a5300.0027a,b2600.6336a2150.2355a
  I111692517 384330 465015 364134
  II261385 41012 12122 6146
  III301389 5718 10164 61311
Differentiation
  Well3525910.0002a,b12176 <0.0001a,b72260.0260a,b810170.0326a,b
  Moderately105632715 363930 474612 343932
  Poorly3513735 4724 16163 8207
Histology
  Ad1418440170.0001a,b4556400.0173a,b5370180.6431a4259400.2096a
  Sq271539 6615 13113 6714
  La7205 115 430 232
Total number1751014331 526360 708421 506956

a P-value determined using a χ2 test.

b statistical significance <0.05. NSCLC, non-small-cell lung cancer; TCs, tumor cells; ICs, tumor-infiltrating immune cells; Ad, adenocarcinoma; Sq, squamous cell carcinoma; La, large cell carcinoma.

With respect to TILs, the percentage of PD-L2-positive TCs was significantly higher in the TIL-high group compared with that in the TIL-low group (17.4±25.9% vs. 10.5±17.0%; P=0.0494; Fig. 3G). In addition, the TIL density was significantly correlated with the percentage of PD-L2-positive ICs (r=0.226; P=0.003). The percentage of PD-L2-positive ICs was significantly higher in the TIL-high group compared with that in the TIL-low group (15.7±19.7% vs. 7.8±15.3%; P=0.0048; Fig. 3H).

With respect to M2 TAMs, the percentage of PD-L2-positive TCs was significantly higher in the M2 TAM-high group compared with that in the M2 TAM-low group (19.0±27.9% vs. 11.9±18.9%; P=0.0452; Fig. 3I). Furthermore, the percentage of PD-L2-positive ICs was also significantly higher in the M2 TAM-high group compared with that in the M2 TAM-low group (16.9±19.8% vs. 9.8±17.0%; P=0.0125; Fig. 3J).

With respect to the combined evaluation of TILs and M2 TAMs, the percentage of PD-L2-positive TCs was significantly the lowest in both the TIL-low and M2 TAM-low tumors (Fig. 3K). The percentage of PD-L2-positive ICs was significantly lower in both the TIL-low and M2 TAM-low tumors compared with that in both the TIL-high and M2 TAM-high tumors (P=0.0011; Fig. 3L).

Correlations between the expression of PD-L1 and PD-L2 on the TCs and ICs among resected NSCLC

There was no correlation between the percentage of PD-L1-positive TCs and the percentage of PD-L2-positive TCs (r=0.019; P=0.8049; Fig. 4A). On the other hand, the percentage of PD-L1-positive TCs was significantly correlated with the percentage of PD-L1-positive ICs (r=0.396; P<0.0001; Fig. 4B). In addition, the percentage of PD-L2-positive TCs also was significantly correlated with the percentage of PD-L2-positive ICs (r=0.488; P<0.0001; Fig. 4C).

Expression of PD-L1 and PD-L2 with respect to tumor differentiation

PD-L1 expression on TCs was significantly associated with tumor differentiation (P=0.0002; Table II), as previously reported (8). The percentage of PD-L1-positive TCs was 6.0±17.0% in well-differentiated tumors, 13.1±24.2% in moderately differentiated tumors and 32.8±34.9% in poorly differentiated tumors. The percentage of PD-L1-positive TCs was significantly higher in poorly differentiated tumors compared with that in well- and moderately differentiated tumors (P<0.0001 and P=0.0001, respectively; Fig. 5A).

Furthermore, PD-L1 expression on ICs was also significantly associated with tumor differentiation (P<0.0001; Table II), as previously reported (8). The percentage of PD-L1-positive ICs was 5.7±8.6% in well-differentiated tumors, 8.3±10.0% in moderately differentiated tumors and 16.2±12.6% in poorly differentiated tumors. The percentage of PD-L1-positive ICs was significantly higher in poorly differentiated tumors compared with that in well- and moderately differentiated tumors (P<0.0001 and P=0.0001, respectively; Fig. 5B).

On the other hand, PD-L2 expression on TCs was inversely associated with tumor differentiation (P=0.0260; Table II). The percentage of PD-L2-positive TCs was 23.5±25.9% in well-differentiated tumors, 13.4±22.4% in moderately differentiated tumors and 9.2±19.1% in poorly differentiated tumors. The percentage of PD-L2-positive TCs was significantly higher in well-differentiated tumors compared with that in poorly and moderately differentiated tumors (P=0.0088 and P=0.0234, respectively; Fig. 5C).

The PD-L2 expression on ICs was also inversely associated with tumor differentiation (P=0.0326; Table II). The percentage of PD-L2-positive ICs was 19.3±22.5% in well-differentiated tumors, 11.4±17.4% in moderately differentiated tumors and 9.1±15.3% in poorly differentiated tumors. The percentage of PD-L2-positive ICs was significantly higher in well-differentiated tumors compared with that in poorly and moderately differentiated tumors (P=0.0196 and P=0.0269, respectively; Fig. 5D).

Discussion

A comprehensive study on PD-L1 and PD-L2 expression on both TCs and ICs in NSCLC was performed. A recent study reports that PD-L1 has predominant roles in Th1-type immunity whereas PD-L2 is involved in Th2-type immunity (26). In addition, to elucidate the biological mechanisms of their regulation, TILs and M2 TAMs, which are key components of the TME and associated with tumor progression, were investigated. The evaluation of PD-L1 and PD-L2 on both TCs and ICs is clinically important and immunohistochemistry is an appropriate method for the design of the present study. A previous study reports that the Ventana PD-L1 (SP-263) assay is clinically useful for PD-L1 staining on both TCs and ICs (24). In addition, the PD-L2 expression using the Ventana system also exhibited a clear staining on both TCs and ICs in the present study.

Consequently, the present study revealed that the TIL density was strongly associated with the PD-L1 expression on both TCs and ICs. On the other hand, PD-L2 was widely expressed not only on TCs, but also on ICs in NSCLC. In addition, the TIL density was also associated with PD-L2 expression on both TCs and ICs. Initially, CD8+ or CD4+ T cells and NK cells are known to induce PD-L1 expression by producing interferon (IFN)-γ (27,28). TILs have been reported to be an important cause of PD-L1 expression on ICs, such as lymphatic endothelial cells, macrophages and monocytes (2931). Numerous clinical studies have also revealed that TILs are associated with PD-L1 expression in human cancer, including NSCLC (7,32,33). In addition, previous studies report that TILs are also associated with PD-L2 expression in human cancer (33,34).

Based on the physiological or pathological situation, macrophages can be polarized into various phenotypes with different biological properties, such as tumor-inhibiting M1 macrophages and tumor-promoting M2 macrophages (35,36). During tumor progression, Th2-derived cytokines originating from TCs and stromal cells can induce the production of M2 TAMs in the TME, which can promote tumor cell proliferation (37). In fact, the M2 TAM density was associated with nodal status and pathological stage in the present study. Thus, M2 TAM-high tumors have more aggressive potential in NSCLC (23).

On the other hand, our previous study found that the M2 TAM density was strongly associated with PD-L1 expression on both TCs and ICs (8). In addition, the present study demonstrated that the M2 TAM density was also associated with the PD-L2 expression on both TCs and ICs. Experimental studies report that TCs can induce M2 TAMs with increased expression of PD-L1 (38,39). It is also known that PD-L1, induced by IFN-γ from TAMs, promoted the progression of lung cancer (40). Recent studies show that other signals derived from macrophages, such as TNF-α, VEGF and CXCL8, can induce PD-L1 expression (4143). In addition, previous studies report that macrophages can induce not only PD-L1 expression, but also PD-L2 expression (44,45).

From these findings, the TIL and M2 TAM densities were associated with the expression of PD-L1 and PD-L2 on TCs and ICs. In the present study, the TIL density was significantly associated with the preoperative serum albumin level (r=0.269; P<0.001; Fig. S1A) and the preoperative peripheral blood lymphocyte count (r=0.209; P=0.006; Fig. S1B). Therefore, TILs are considered to be a host-related factor. By contrast, M2 TAMs are considered to be a tumor-related factor (23). Thus, such complex crosstalk in the TME, including TILs and M2 TAMs, could affect the expression of PD-L1 and PD-L2 on TCs and ICs in NSCLC (46).

However, the present study demonstrated the additional finding of no correlation between PD-L1 expression on TCs and PD-L2 expression on TCs, despite the possible same regulations by TILs and M2 TAMs. Several studies also report a high frequency of discordance between PD-L1 and PD-L2 expression in human cancer (47,48). By contrast, there were correlations between PD-L1 expression on TCs and PD-L1 expression on ICs and between PD-L2 expression on TCs and PD-L2 expression on ICs in the present study.

The present study revealed that tumor differentiation was strongly associated with PD-L1 expression on TCs and ICs. The percentages of PD-L1-positive TCs and PD-L1-positive ICs were higher in poorly differentiated tumors compared with that in well- and moderately differentiated tumors. A meta-analysis on PD-L1 expression in lung cancer also reports the same results (49). In addition, an experimental study reveals that PD-L1 could upregulate the β-catenin signaling pathway to induce epithelial-mesenchymal transition (50), which is associated with tumor differentiation in lung cancer (51,52). By contrast, tumor differentiation was inversely associated with PD-L2 expression on TCs and ICs in the present study. The percentages of PD-L2-positive TCs and PD-L2-positive ICs were higher in well-differentiated tumors compared with that in poorly and moderately differentiated tumors.

Therefore, the combined evaluation of PD-L1 and PD-L2 expression could be considered clinically important in the treatment strategy of immune-checkpoint inhibitors in patients with NSCLC. In particular, the evaluation of PD-L2 expression may be necessary for patients with PD-L1-negative NSCLC. Patients with PD-L2-positive NSCLC could be treated with anti-PD-1 antibodies, such as Pembrolizumab, and combined treatment with anti-PD-L2 antibodies in the future (1820). In fact, in the present study, immune-checkpoint inhibitors were only used in 7 cases of PD-L1-positive tumors at the time of disease recurrence, whereas 56 cases had recurrence following surgery. Further clinical studies are required for patients with PD-L2-positive NSCLC. In addition, the present study was performed using a relatively small number of patients at one institution. Therefore, a further study using more cases is required to elucidate the clinical significance of PD-L2 expression, especially with respect to the treatment strategy of immune-checkpoint inhibitors. Furthermore, the present study was evaluated only by immunohistochemistry and a further study to investigate their gene copy numbers may be needed (53).

In conclusion, PD-L1 and PD-L2 expression on TCs and ICs was associated with TILs and M2 TAMs in NSCLC. However, there was no correlation between PD-L1 and PD-L2 expression on TCs. Meanwhile, PD-L1 expression on TCs and ICs was associated with tumor differentiation, while PD-L2 expression on TCs and ICs was inversely associated with tumor differentiation. The combined evaluation of PD-L1 and PD-L2 expression could be considered clinically important in the treatment strategy of immune-checkpoint inhibitors in patients with NSCLC. In particular, the evaluation of PD-L2 expression may be necessary for patients with PD-L1-negative NSCLC.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

RS, CLH and HD designed the study. RS, CLH and MF designed and performed the experiments. RS, CLH and HC collected the data. RS and CLH analyzed and interpreted the data and wrote the manuscript. RS and CLH confirm the authenticity of all the raw data. All authors have read and approved the final version of the manuscript for publication.

Ethics approval and consent to participate

The current study was approved by the Institutional Ethics Committee of the Kitano Hospital (approval no. P181200300) and written informed consent was provided from each patient. The research was conducted in compliance with the principles outlined in the Declaration of Helsinki.

Patient consent for publication

Written informed consent for publication of patient data/accompanying images was obtained.

Competing interests

The authors declare that they have no competing interests.

References

1 

Ettinger DS, Akerley W, Bepler G, Blum MG, Chang A, Cheney RT, Chirieac LR, D'Amico TA, Demmy TL, Ganti AK, et al: Non-small cell lung cancer. J Natl Compr Canc Netw. 8:740–801. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Hsu WH, Yang JC, Mok TS and Loong HH: Overview of current systemic management of EGFR-mutant NSCLC. Ann Oncol. 29 (Suppl_1):i3–i9. 2018. View Article : Google Scholar : PubMed/NCBI

3 

Borghaei H, Paz-Ares L, Horn L, Spigel DR, Steins M, Ready NE, Chow LQ, Vokes EE, Felip E, Holgado E, et al: Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N Engl J Med. 373:1627–1639. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Herbst RS, Baas P, Kim DW, Felip E, Perez-Gracia JL and Han JY: Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): A randomized controlled trial. Lancet. 387:1540–1550. 2016. View Article : Google Scholar : PubMed/NCBI

5 

Rittmeyer A, Barlesi F, Waterkamp D, Park K, Ciardiello F, von Pawel J, Gadgeel SM, Hida T and Kowalski DM: Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): A phase 3, open-label, multicenter randomized controlled trial. Lancet. 389:255–265. 2017. View Article : Google Scholar : PubMed/NCBI

6 

Kowanetz M, Zou W, Gettinger SN, Koeppen H, Kockx M, Schmid P, Kadel EE, Wistuba I, Chaft J, Rizvi NA, et al: Differential regulation of PD-L1 expression by immune and tumor cells in NSCLC and the response to treatment with atezolizumab (anti-PD-L1). Proc Natl Acad Sci USA. 115:E10119–E10126. 2018. View Article : Google Scholar : PubMed/NCBI

7 

Kim MY, Koh J, Kim S, Go H, Jeon YK and Chung DH: Clinicopathological analysis of PD-L1 and PD-L2 expression in pulmonary squamous cell carcinoma: Comparison with tumor-infiltrating T cells and the status of oncogenic drivers. Lung Cancer. 88:24–33. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Sumitomo R, Hirai T, Fujita M, Murakami H, Otake Y and Huang C: 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

9 

Shinchi Y, Komohara Y, Yonemitsu K, Sato K, Ohnishi K, Saito Y, Fujiwara Y, Mori T, Shiraishi K, Ikeda K and Suzuki M: Accurate expression of PD-L1/L2 in lung adenocarcinoma cells: A retrospective study by double immunohistochemistry. Cancer Sci. 110:2711–2721. 2019. View Article : Google Scholar : PubMed/NCBI

10 

Matsubara T, Takada K, Azuma K, Takamori S, Toyokawa G, Haro A, Osoegawa A, Tagawa T, Kawahara A, Akiba J, et al: A clinicopathological and prognostic analysis of PD-L2 expression in surgically resected primary lung squamous cell carcinoma. Ann Surg Oncol. 26:1925–1933. 2019. View Article : Google Scholar : PubMed/NCBI

11 

Takamori S, Takada K, Azuma K, Jogo T, Shimokawa M, Toyokawa G, Hirai F, Tagawa T, Kawahara A, Akiba J, et al: Prognostic impact of programmed death-ligand 2 expression in primary lung adenocarcinoma patients. Ann Surg Oncol. 26:1916–1924. 2019. View Article : Google Scholar : PubMed/NCBI

12 

Baptista MZ, Sarian LO, Derchain SFM, Pinto GA and Vassallo J: Prognostic significance of PD-L1 and PD-L2 in breast cancer. Hum Pathol. 47:78–84. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Zhao SG, Lehrer J, Chang SL, Das R, Erho N, Liu Y, Sjostrom M, Den RB, Freedland SJ, Klein EA, et al: The immune landscape of prostate cancer and nomination of PD-L2 as a potential therapeutic target. J Natl Cancer Inst. 111:301–310. 2019. View Article : Google Scholar : PubMed/NCBI

14 

Okadome K, Baba Y, Nomoto D, Yagi T, Kalikawe R, Harada K, Hiyoshi Y, Nagai Y, Ishimoto T, Iwatsuki M, et al: Prognostic and clinical impact of PD-L2 and PD-L1 expression in a cohort of 437 oesophageal cancers. Br J Cancer. 122:1535–1543. 2020. View Article : Google Scholar : PubMed/NCBI

15 

Okazaki T and Honjo T: PD-1 and PD-1 ligands: From discovery to clinical application. Int Immunol. 19:813–824. 2007. View Article : Google Scholar : PubMed/NCBI

16 

Rozali EN, Hato SV, Robinson BW, Lake RA and Lesterhuis WJ: Programmed death ligand 2 in cancer-induced immune suppression. Clin Dev Immunol. 2021:6563402012.PubMed/NCBI

17 

Zhong X, Tumang JR, Gao W, Bai C and Rothstein TL: PD-L2 expression extends beyond dendritic cells/macrophages to B1 cells enriched for V(H)11/V(H)12 and phosphatidylcholine binding. Eur J Immunol. 37:2405–2410. 2007. View Article : Google Scholar : PubMed/NCBI

18 

Tanegashima T, Togashi Y, Azuma K, Kawahara A, Ikeguchi K, Sugiyama D, Kinoshita F, Akiba J, Kashiwagi E, Takeuchi A, et al: Immune suppression by PD-L2 against spontaneous and treatment-related antitumor immunity. Clin Cancer Res. 25:4808–4819. 2019. View Article : Google Scholar : PubMed/NCBI

19 

Umezu D, Okada N, Sakoda Y, Adachi K, Ojima T, Yamaue H, Eto M and Tamada K: Inhibitory functions of PD-L1 and PD-L2 in the regulation of anti-tumor immunity in murine tumor microenvironment. Cancer Immunol Immunother. 68:201–211. 2019. View Article : Google Scholar : PubMed/NCBI

20 

Yearley JH, Gibson C, Yu N, Moon C, Murphy E, Juco J, Lunceford J, Cheng J, Chow LQM, Seiwert TY, et al: PD-L2 Expression in Human Tumors: Relevance to Anti-PD-1 Therapy in Cancer. Clin Cancer Res. 23:3158–3167. 2017. View Article : Google Scholar : PubMed/NCBI

21 

Amin MB, Edge S and Greene F: AJCC Cancer Staging Manual. 8th edition. Springer; New York: 2017, View Article : Google Scholar

22 

Travis WD, Brambilla E, Burke AP, Marx A and Nicholson AG: WHO Classification of Tumours of the Lung, Pleura, Thymus and Heart. 4th edition. International Agency for Research on Cancer; Lyon, France: 2015

23 

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

24 

Tsao MS, Kerr KM, Kockx M, Beasley M, Borczuk AC, Botling J, Budendorf L, Chirieac L, Chen G, Chou T, et al: PD-L1 immunohistochemistry comparability study in real-life clinical samples: Results of Blueprint print phase 2 project. J Thorac Oncol. 13:1302–1311. 2018. View Article : Google Scholar : PubMed/NCBI

25 

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

26 

Tanaka R, Ichimura Y, Kubota N, Saito A, Nakamura Y, Ishitsuka Y, Watanabe R, Fujisawa Y, Mizuno S, Takahashi S, et al: Differential involvement of programmed cell death ligands in skin immune responses. J Invest Dermatol. 142:145–154.e8. 2022. View Article : Google Scholar : PubMed/NCBI

27 

Sanmamed MF and Chen L: Inducible expression of B7-H1 (PD-L1) and its selective role in tumor site immune modulation. Cancer J. 20:256–261. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Chen J, Feng Y, Lu L, Wang H, Dai L, Li Y and Zhang P: Interferon-γ-induced PD-L1 surface expression on human oral squamous carcinoma via PKD2 signal pathway. Immunobiology. 217:385–393. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Lane RS, Femel J, Breazeale AP, Loo CP, Thibault G, Kaempf A, Mori M, Tsujikawa T, Chang YH and Lund AW: IFNγ-activated dermal lymphatic vessels inhibit cytotoxic T cells in melanoma and inflamed skin. J Exp Med. 215:3057–3074. 2018. View Article : Google Scholar : PubMed/NCBI

30 

Qian J, Wang C, Wang B, Yang J, Wang Y, Luo F, Xu J, Zhao C, Liu R and Chu Y: The IFN-γ/PD-L1 axis between T cells and tumor microenvironment: Hints for glioma anti-PD-1/PD-L1 therapy. J Neuroinflammation. 15:2902018. View Article : Google Scholar : PubMed/NCBI

31 

Chen S, Crabill GA, Pritchard TS, McMiller TL, Wei P, Pardoll DM, Pan F and Topalian SL: Mechanisms regulating PD-L1 expression on tumor and immune cells. J Immunother Cancer. 7:3052019. View Article : Google Scholar : PubMed/NCBI

32 

Arrieta O, Montes-Servin E, Hernandez-Martinez J, Cardona AF, Cases-Ruiz E, Crispin JC, Motola D, Flores-Estrada D and Barrera L: Expression of PD-1/PD-L1 and PD-L2 in peripheral T-cells from non-small cell lung cancer patients. Oncotarget. 8:101994–102005. 2017. View Article : Google Scholar : PubMed/NCBI

33 

Kitsou M, Ayiomamitis GD and Zaravinos A: High expression of immune checkpoints is associated with the TIL load, mutation rate and patient survival in colorectal cancer. Int J Oncol. 57:237–248. 2020. View Article : Google Scholar : PubMed/NCBI

34 

Zhang Y, Xu J, Hua J, Liu J, Liang C, Meng Q, Wei M, Zhang B and Yu X: A PD-L2-based immune marker signature helps to predict survival in resected pancreatic ductal adenocarcinoma. J Immunotherapy Cancer. 7:2332019. View Article : Google Scholar : PubMed/NCBI

35 

Mei J, Xiao Z, Guo C, Pu Q, Ma L, Liu C, Lin F, Liao H, You Z and Liu L: Prognostic impact of tumor-associated macrophage infiltration in non-small cell lung cancer: A systemic review and meta-analysis. Oncotarget. 7:34217–34228. 2016. View Article : Google Scholar : PubMed/NCBI

36 

Jackute J, Zemaitis M, Pranys D, Sitkauskiene B, Miliauskas S, Vaitkiene S and Sakalaukas R: Distribution of M1 and M2 macrophages in tumor islets and stroma in relation to prognosis of non-small cell lung cancer. BMC Immunol. 19:32018. View Article : Google Scholar : PubMed/NCBI

37 

Mantovani A, Sozzani S, Locati M, Allavena P and Sica A: Macrophage polarization: Tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Trends Immunol. 23:549–555. 2002. View Article : Google Scholar : PubMed/NCBI

38 

Gabrusiewicz K, Li X, Wei J, Hashimoto Y, Marisetty AL, Ott M, Wang F, Hawke D, Yu J, Healy LM, et al: Glioblastoma stem cell-derived exosomes induce M2 macrophages and PD-L1 expression on human monocytes. Oncoimmunology. 7:e14129092018. View Article : Google Scholar : PubMed/NCBI

39 

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

40 

Zhang X, Zeng Y, Qu Q, Zhu J, Liu Z, Ning W, Zeng H, Zhang N, Du W, Chen C and Huang JA: PD-L1 induced by IFN-γ from tumor-associated macrophages via the JAK/STAT3 and PI3K/AKT signalling pathways promoted progression of lung cancer. Int J Clin Oncol. 22:1026–1033. 2017. View Article : Google Scholar : PubMed/NCBI

41 

Tsukamoto M, Imai K, Ishimoto T, Komohara Y, Yamashita Y, Nakagawa S, Umezaki N, Yamao T, Kitano Y, Miyata T, et al: PD-L1 expression enhancement by infiltrating macrophage-derived tumor necrosis factor-α leads to poor pancreatic cancer prognosis. Cancer Sci. 110:310–320. 2019.PubMed/NCBI

42 

Lai YS, Wahyuningtyas R, Aui SP and Chang KT: Autocrine VEGF signalling on M2 macrophages regulates PD-L1 expression for immunomodulation of T cells. J Cell Mol Med. 23:1257–1267. 2019.PubMed/NCBI

43 

Lin C, He H, Liu H, Li R, Chen Y, Qi Y, Jiang Q, Chen L, Zhang P, Zhang H, et al: Tumor-associated macrophage-derived CXCL8 determines immune evasion through autonomous PD-L1 expression in gastric cancer. Gut. 68:1764–1773. 2019. View Article : Google Scholar : PubMed/NCBI

44 

Horlad H, Ma C, Yano H, Pan C, Ohnishi K, Fujiwara Y, Endo S, Kikukawa Y, Okuno Y, Matsuoka M, et al: An IL-27/Stat3 axis induces expression of programmed death ligands (PD-L1/2) on infiltrating macrophages in lymphoma. Cancer Sci. 107:1696–1704. 2016. View Article : Google Scholar : PubMed/NCBI

45 

Cai X, Yuan F, Zhu J, Yang J, Tang C, Cong Z and Ma C: Glioma-associated stromal cells stimulate glioma malignancy by regulating the tumor immune microenvironment. Front Oncol. 11:6729282021. View Article : Google Scholar : PubMed/NCBI

46 

Wang J, Li D, Cang H and Guo B: Crosstalk between cancer and immune cells: Role of tumor-associated macrophages in the tumor microenvironment. Cancer Med. 8:4709–4721. 2019. View Article : Google Scholar : PubMed/NCBI

47 

Menguy S, Prochazkova-Carlotti M, Beylot-Barry M, Saltel F, Vergier B, Merlio J and Pham-Ledard A: PD-L1 and PD-L2 are differentially expressed by macrophages or tumor cells in primary cutaneous diffuse large B-cell lymphoma, Leg type. Am J Surg Pathol. 42:326–334. 2018. View Article : Google Scholar : PubMed/NCBI

48 

Pinato DJ, Vallipuram A, Evans JS, Wong C, Zhang H, Brown M, Dina RE, Trivedi P, Akarca AU, Marafioti T, et al: Programmed cell death ligand expression drives immune tolerogenesis across the diverse subtypes of neuroendocrine tumors. Neuroendocrinology. 111:465–474. 2021. View Article : Google Scholar : PubMed/NCBI

49 

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 Ling Cancer Res. 8:429–449. 2019. View Article : Google Scholar : PubMed/NCBI

50 

Yu W, Hua Y, Qiu H, Hao J, Zou Z, Li Z, Hu S, Guo P, Chen M, Sui S, et al: PD-L1 promotes tumor growth and progression by activating WIP and β-catenin signaling pathways and predicts poor prognosis in lung cancer. Cell Death Dis. 11:5062020. View Article : Google Scholar : PubMed/NCBI

51 

Sato M, Shames DS and Hasegawa Y: Emerging evidence of epithelial-to-mesenchymal transition in lung carcinogenesis. Respirology. 17:1048–1059. 2012. View Article : Google Scholar : PubMed/NCBI

52 

Brabletz S, Schuhwerk H, Brabletz T and Stemmler MP: Dynamic EMT: A multi-tool for tumor progression. EMBO J. 40:e1086472021. View Article : Google Scholar : PubMed/NCBI

53 

Inoue Y, Yoshimura K, Nishimoto K, Inui N, Karayama M, Yasui H, Hozumi H, Suzuki Y, Furuhashi K, Fujisawa T, et al: Evaluation of programmed death ligand 1 (PD-L1) gene amplification and response to nivolumab monotherapy in non-small cell lung cancer. JAMA Netw Open. 3:e20118182020. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2022
Volume 47 Issue 4

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Sumitomo R, Huang C, Fujita M, Cho H and Date H: Differential expression of PD‑L1 and PD‑L2 is associated with the tumor microenvironment of TILs and M2 TAMs and tumor differentiation in non‑small cell lung cancer. Oncol Rep 47: 73, 2022
APA
Sumitomo, R., Huang, C., Fujita, M., Cho, H., & Date, H. (2022). Differential expression of PD‑L1 and PD‑L2 is associated with the tumor microenvironment of TILs and M2 TAMs and tumor differentiation in non‑small cell lung cancer. Oncology Reports, 47, 73. https://doi.org/10.3892/or.2022.8284
MLA
Sumitomo, R., Huang, C., Fujita, M., Cho, H., Date, H."Differential expression of PD‑L1 and PD‑L2 is associated with the tumor microenvironment of TILs and M2 TAMs and tumor differentiation in non‑small cell lung cancer". Oncology Reports 47.4 (2022): 73.
Chicago
Sumitomo, R., Huang, C., Fujita, M., Cho, H., Date, H."Differential expression of PD‑L1 and PD‑L2 is associated with the tumor microenvironment of TILs and M2 TAMs and tumor differentiation in non‑small cell lung cancer". Oncology Reports 47, no. 4 (2022): 73. https://doi.org/10.3892/or.2022.8284