Neoplastic cancer cells and cancer stroma (including infiltrating immune cells) determine the biology and prognosis of cancer. Various types of adaptive and innate immune cells are known to infiltrate the cancer stroma. However, the patterns and spatial distribution of immune cell infiltration as well as its association with tumor histology remain poorly understood. To address these issues, we comprehensively analyzed the infiltrating immune cells present in lung adenocarcinoma. The principal types of both adaptive and innate infiltrating immune cells were immunohistochemically evaluated in the predominant histologic components of 111 lung adenocarcinomas. The same analysis was also carried out on 143 samples of histologic subtypes making up more than 20% of tumors. As a result, plasma cells and B cells with interfollicular distribution were almost exclusively observed in invasive histologic subtypes, while an increased number of mast cells were observed in noninvasive histologic subtypes. Cluster analysis revealed four distinct immunosubtypes (CD8, mast cell, macrophage/dendritic cell, and plasma cell subtypes) based on the infiltrating immune cell profiles. These immunosubtypes correlated with histologic subtypes, and univariate and multivariate analyses identified the plasma cell subtype as an independent negative prognostic factor. These plasma cells may be one of the major producers of the immunosuppressive cytokine IL35 in cancer stroma. Cancer Immunol Res; 4(3); 234–47. ©2016 AACR.

Cancer tissue is composed of neoplastic cancer cells and nonneoplastic cancer stroma; the latter includes blood vessels, lymphatic vessels, cancer-associated fibroblasts, extracellular matrix, and infiltrating immune cells (1, 2). Cancer stroma is induced and maintained by cancer cells, and there are reciprocal interactions between cancer cells and cancer stroma in the course of the initiation, progression, and metastasis of cancer (1–3). Accumulating evidence indicates that the phenotype of cancer is defined not only by the biologic nature of cancer cells but also by the stromal component, including infiltrating immune cells (4). Recent successes in blockade of the immune inhibitory axis using monoclonal antibodies against programmed death-1 (PD-1) and its ligand PD-L1 have attracted further interest to the roles played by infiltrating immune cells (5).

So far, only a few studies have addressed the complex nature of immune cell infiltration in cancer stroma. Studies have been carried out in which immunohistochemistry-based detection of infiltrating immune cells was combined with cluster analysis in breast and ovarian cancer (6, 7). However, the range of infiltrating immune cells analyzed in these studies was not fully comprehensive. Improved completeness was achieved in studies using flow cytometric analysis (8, 9) and gene expression–based approaches, including ESTIMATE and CIBERSORT (10–13), but histologic information on the distribution of infiltrating immune cells in cancer tissue is lost when using these approaches (14). A mass spectrometry–based approach (15) can combine the analysis of numerous antigens with histology of tumors, but the technology is not fully established. Therefore, the diversity and the landscape of infiltrating immune cells in cancer stroma are still largely undetermined.

Lung adenocarcinoma accounts for more than 40% of total lung cancers and is one of the leading causes of cancer-related death worldwide (16). Infiltrating immune cells have been intensively studied in non–small cell lung carcinomas (NSCLC), which include both adenocarcinoma and squamous cell carcinoma. These studies have shown that lung NSCLCs with increased numbers of T cells, B cells, and S100+ dendritic cells (DC) are associated with better prognosis (17, 18), whereas those with increased numbers of regulatory T cells (Treg) and stromal macrophages (19–21) are associated with poorer prognosis. Several studies have indicated that the prognostic values of infiltrating immune cells differ between lung adenocarcinoma and squamous cell carcinoma; for example, the numbers of T cells, B cells, and macrophages showed no prognostic value in lung adenocarcinoma (19). However, only a few studies have noted these differences to date. Therefore, the prognostic values of infiltrating immune cells and their patterns of infiltration in lung adenocarcinoma are still poorly understood.

Another intriguing but challenging aspect of lung adenocarcinoma lies in the diversity and heterogeneity of histologic findings within one tumor. The recently revised 2015 World Health Organization (WHO) classification (16) and the International Association for the Study of Lung Cancer, American Thoracic Society, and European Respiratory Society (IASLC/ATS/ERS) classification of lung adenocarcinomas (22) have defined the terms for histologic subtypes. These include the noninvasive lepidic subtype and other invasive histologic subtypes (papillary, acinar, micropapillary, and solid subtypes). Using these terms, adenocarcinoma is classified into adenocarcinoma in situ (AIS), which is purely lepidic and noninvasive; minimally invasive adenocarcinoma (MIA); and invasive adenocarcinoma classified by the predominant histologic subtype: lepidic, papillary, acinar, micropapillary, and solid adenocarcinoma. However, the relationships between these histologic subtypes and the patterns of immune cell infiltration are currently unknown.

Here, we report the results of comprehensive immunohistochemistry-based profiling of infiltrating immune cells in lung adenocarcinoma, covering the principal types of both adaptive and innate immune cells. By carrying out cluster analysis of the infiltrating immune cells, we identified four distinct immunosubtypes of lung adenocarcinoma [CD8, mast cell, macrophage/DC (MØ/DC), and plasma cell subtypes]. These immunosubtypes correlated with histologic subtypes, and univariate and multivariate analyses identified the plasma cell subtype as an independent negative prognostic factor. These results strongly indicate the importance of comprehensive analyses of infiltrating immune cells and comparison of these data with spatial information, including tumor histology.

Patients

One hundred and fourteen patients with lung adenocarcinoma who underwent lobectomy (111 cases) and partial resection (3 cases) at Keio University Hospital (Tokyo, Japan) between 2003 and 2005 were enrolled in this study. No patient underwent neoadjuvant therapy before surgery. Cases with positive surgical margins (two cases) and one stage IIIb case were excluded from the clinicopathologic analysis but were enrolled in the histopathologic analysis. Clinical data, including age, sex, smoking history, and follow-up data, were obtained by reviewing medical records and are shown in Table 1. The median follow-up period was 85 months (range, 3–108 months). The study was approved by the ethics committees of Keio University School of Medicine.

Table 1.

Univariate analysis of clinicopathologic factors and infiltrating immune cells

All casesStage I
VariablesN5-year DFSPN5-year DFSP
Age, y 
 <65 57 75% 0.767  n.d.  
 ≥65 54 72%     
Gender 
 Male 57 67% 0.211  n.d.  
 Female 54 79%     
Ever smoker 
 No 63 81% 0.015  n.d.  
 Yes 48 64%     
Stage 
 I 93 83% <0.001*  n.d.  
 II, IIIA 18 33%     
Tumor size 
 ≤3.0 cm 96 77% <0.001* 70 86% 0.163 
 >3.0 cm 15 53%  23 73%  
Pleural invasion 
 Absent 84 82% <0.001* 76 89% <0.001* 
 Present 27 46%  17 58%  
Lymph node metastasis 
 Absent 93 82% <0.001*  n.d.  
 Present 18 33%     
Lymphatic invasion 
 Absent 78 87% <0.001* 77 89% 0.0011* 
 Present 33 42%  16 56%  
Vascular invasion 
 Absent 94 81% <0.001* 84 87% <0.001* 
 Present 17 34%  44%  
Histology subtype by WHO classification       
 AIS/MIA/lepidic 36 96% <0.001* 24 100% 0.011 
 Papillary/acinar/micropapillary/solid 75 62%  69 77%  
Lymphoid follicle, per LPF 
 <4 77 76% 0.344 60 85% 0.433 
 ≥4 68 66%  33 78%  
CD3+ T cell, cells/HPF 
 <150 78 76% 0.070 66 88% 0.030 
 ≥150 33 66%  27 69%  
CD8/CD3 ratio (high) 
 <0.6 91 90% 0.076 74 89% 0.432 
 ≥0.6 20 70%  19 82%  
CD8/CD3 ratio (low) 
 >0.4 99 73% 0.270 86 83% 0.413 
 ≤0.4 12 74%  83%  
Foxp3+CD3+ Treg, cells/HPF 
 <10 66 82% 0.006* 59 87% 0.080 
 ≥10 45 60%  34 75%  
Total mast cell, cells/HPF 
 <30 70 62% <0.001* 53 74% 0.0022* 
 ≥30 41 94%  40 97%  
Stromal mast cell, cells/HPF 
 <10 93 73% 0.595 75 84% 0.515 
 ≥10 18 76%  18 76%  
Interfollicular plasma cell, cells/HPF 
 <30 78 86% <0.001* 70 92% <0.001* 
 ≥30 33 45%  23 56%  
Parafollicular plasma cell, cells/HPF 
 <25 89 79% <0.001* 77 87% <0.001* 
 ≥25 22 50%  16 62%  
Interfollicular B cell, cells/HPF 
 <30 81 83% <0.001* 71 91% <0.001* 
 ≥30 30 49%  22 58%  
Macrophage, cells/HPF 
 <40 62 81% 0.048 51 91% 0.013 
 ≥40 49 64%  42 72%  
CD11c+ DC 
 Score 1, 2 62 90% <0.001* 56 96% <0.001* 
 Score 3, 4 49 53%  37 63%  
S100+ DC, cells/HPF 
 <30 83 76% 0.211 72 82% 0.782 
 ≥30 28 66%  21 84%  
Plasmacytoid DC, cells/HPF 
 <10 78 75% 0.261 66 87% 0.040 
 ≥10 33 67%  27 73%  
Neutrophil, cells/HPF 
 <10 90 80% 0.0025* 79 86% 0.035 
 ≥10 21 47%  14 64%  
All casesStage I
VariablesN5-year DFSPN5-year DFSP
Age, y 
 <65 57 75% 0.767  n.d.  
 ≥65 54 72%     
Gender 
 Male 57 67% 0.211  n.d.  
 Female 54 79%     
Ever smoker 
 No 63 81% 0.015  n.d.  
 Yes 48 64%     
Stage 
 I 93 83% <0.001*  n.d.  
 II, IIIA 18 33%     
Tumor size 
 ≤3.0 cm 96 77% <0.001* 70 86% 0.163 
 >3.0 cm 15 53%  23 73%  
Pleural invasion 
 Absent 84 82% <0.001* 76 89% <0.001* 
 Present 27 46%  17 58%  
Lymph node metastasis 
 Absent 93 82% <0.001*  n.d.  
 Present 18 33%     
Lymphatic invasion 
 Absent 78 87% <0.001* 77 89% 0.0011* 
 Present 33 42%  16 56%  
Vascular invasion 
 Absent 94 81% <0.001* 84 87% <0.001* 
 Present 17 34%  44%  
Histology subtype by WHO classification       
 AIS/MIA/lepidic 36 96% <0.001* 24 100% 0.011 
 Papillary/acinar/micropapillary/solid 75 62%  69 77%  
Lymphoid follicle, per LPF 
 <4 77 76% 0.344 60 85% 0.433 
 ≥4 68 66%  33 78%  
CD3+ T cell, cells/HPF 
 <150 78 76% 0.070 66 88% 0.030 
 ≥150 33 66%  27 69%  
CD8/CD3 ratio (high) 
 <0.6 91 90% 0.076 74 89% 0.432 
 ≥0.6 20 70%  19 82%  
CD8/CD3 ratio (low) 
 >0.4 99 73% 0.270 86 83% 0.413 
 ≤0.4 12 74%  83%  
Foxp3+CD3+ Treg, cells/HPF 
 <10 66 82% 0.006* 59 87% 0.080 
 ≥10 45 60%  34 75%  
Total mast cell, cells/HPF 
 <30 70 62% <0.001* 53 74% 0.0022* 
 ≥30 41 94%  40 97%  
Stromal mast cell, cells/HPF 
 <10 93 73% 0.595 75 84% 0.515 
 ≥10 18 76%  18 76%  
Interfollicular plasma cell, cells/HPF 
 <30 78 86% <0.001* 70 92% <0.001* 
 ≥30 33 45%  23 56%  
Parafollicular plasma cell, cells/HPF 
 <25 89 79% <0.001* 77 87% <0.001* 
 ≥25 22 50%  16 62%  
Interfollicular B cell, cells/HPF 
 <30 81 83% <0.001* 71 91% <0.001* 
 ≥30 30 49%  22 58%  
Macrophage, cells/HPF 
 <40 62 81% 0.048 51 91% 0.013 
 ≥40 49 64%  42 72%  
CD11c+ DC 
 Score 1, 2 62 90% <0.001* 56 96% <0.001* 
 Score 3, 4 49 53%  37 63%  
S100+ DC, cells/HPF 
 <30 83 76% 0.211 72 82% 0.782 
 ≥30 28 66%  21 84%  
Plasmacytoid DC, cells/HPF 
 <10 78 75% 0.261 66 87% 0.040 
 ≥10 33 67%  27 73%  
Neutrophil, cells/HPF 
 <10 90 80% 0.0025* 79 86% 0.035 
 ≥10 21 47%  14 64%  

NOTE: *, P < 0.01.

Abbreviations: AIS, adenocarcinoma in situ; DFS, disease-free survival; HPF, high-power field; LPS, low-power field; MIA, minimally invasive adenocarcinoma; n.d., not determined.

Histopathologic analysis

All cases were reviewed by three pathologists (Y. Kurebayashi, K. Emoto, and Y. Hayashi). Identification of the histologic subtypes of lung adenocarcinoma was based on the 2015 WHO classification and the IASLC/ATC/ERS classification (16, 22). Tumor grade [grade 1 to 3 (G1–3)] was determined based on the extent of structural and nuclear atypia. We considered the lepidic subtype to be low-grade, papillary (Pap) and acinar (Aci) subtypes to be moderate-grade, and micropapillary (MP) and solid subtypes to be high-grade histologic subtypes. Furthermore, we distinguished the papillary/acinar subtype with a G3 component (Pap/Aci G3*) from that without a G3 component (Pap/Aci G1/2), and considered the former to be a high-grade histologic subtype. Tumor staging was based on the 7th edition of the TNM classification of the International Union Against Cancer (IUAC; ref. 23). Lymphatic invasion and vessel invasion were also recorded if present. Slide images were imported using Nanozoomer-XR (Hamamatsu Photonics).

Immunohistochemistry

Immunohistochemical analysis was performed on 4-μm-thick, formalin-fixed, paraffin-embedded tissue sections. Each section was deparaffinized in xylene, rehydrated, and incubated in 0.03% H2O2 in 95% methanol for 10 minutes. The antibodies used in this study and the heat-induced epitope retrieval methods (HIER) for each antigen are listed in Supplementary Table S1. HIER was performed at 120°C for 5 minutes using an autoclave. Staining was performed using a horseradish peroxidase system (ImmPRESS, Vector Laboratories) or an alkaline phosphatase system (N-Histofine Simple Stain, Nichirei Bioscience) according to the instructions provided. The chromogens used in this study included DAB, First Red (Nichirei bioscience), Histogreen (AbCys), and Vector SG (Vector Laboratories). For double/triple staining using primary antibodies from the same species, inactivation of the primary antibody and linked enzymes used in the previous step was performed as described previously (24). Briefly, slides after the first staining were washed in phosphate-buffered saline for 5 minutes, autoclaved at 110°C for 10 minutes in the epitope retrieval solution for the second antigen, and the second staining was carried out. The same protocol was repeated to carry out triple staining. After staining, sections were counterstained with hematoxylin, dehydrated with alcohol, penetrated by xylene, and permanently mounted.

Evaluation of infiltrating immune cells

Numbers of each type of infiltrating immune cell were counted in at least three different high-power fields (HPF), and the averages were recorded. To evaluate DC infiltration, a grading system ranging from grade 1 to 5 was used, based on the densities in cancer stroma. If a tumor contained several different histologic subtypes, infiltrating immune cells were evaluated separately for each different histologic subtype that made up more than 20% of a tumor (total histologic subtype dataset, N = 143). In addition, the representative data for infiltrating immune cells in each tumor were defined as those evaluated in the predominant histologic subtype of each tumor (predominant histologic subtype dataset, N = 111); this dataset was used for comparison with survival data.

For analysis of heterogeneity in infiltrating immune cells in each tumor, the numbers of each type of infiltrating immune cells were counted in randomly selected 5 HPFs of described lesions and 10 HPFs of total tumor area, and relative standard deviations (RSD) were calculated.

Cutoff values for the comparison with histologic subtypes were determined by reference of median and 80% tail for lymphoid follicles, CD3+ T cells, total mast cells, macrophages, CD11c+ DCs, S100+ DCs, and CD123+ plasmacytoid DCs (pDC). Cutoff value of the CD8/CD3 ratio was determined by reference of 20% and 80% tail. Cutoff values of Foxp3+CD3+ T cells, interfollicular B cells, parafollicular plasma cells, and interfollicular plasma cells were determined by reference of their bimodal distribution and 80% tail. Because of the scarcity of stromal neutrophils, cutoff value of neutrophils was determined arbitrary. Median was used as cutoff values of IgG+ and IgA+ plasma cells.

Statistical analysis

The correlation matrix was computed using the Pearson correlation coefficient. Univariate analysis was performed using the log-rank test and multivariate analysis was performed using the Cox proportional hazards model. The forward stepwise method was used to select the factors analyzed in the Cox proportional hazard model. Survival curves were calculated using the Kaplan–Meier method. For cluster analysis, the original dataset was standardized using the following formula (25),

formula

where X is the original value and Min (X) and Max (X) are the minimum and maximum values of the variables. Hierarchical clustering based on the Euclidean distance was performed, and a heat map was generated using MultiExperimental Viewer (MeV) v4.9 (26). P values between unpaired and two paired samples were calculated using the Mann–Whitney U test and the Wilcoxon signed-rank test, respectively. A 1% or lower value was considered to be statistically significant and denoted by an asterisk.

Profiles of infiltrating adaptive immune cells

In this study, lymphoid follicles, CD3+ T cells, the CD8/CD3 ratio, Foxp3+CD3+ Tregs, CD20+ B cells, and CD79α+p63+ plasma cells were analyzed as indicators of infiltrating adaptive immune cells. In lung adenocarcinoma, CD20+ B cells were mainly located in ectopically formed lymphoid follicles, whereas CD3+ T cells were distributed in association with B-cell follicles or randomly distributed in the cancer stroma (Fig. 1A). Germinal center reactions in lymphoid follicles were relatively rare in lung adenocarcinoma. These lymphoid follicles were also characterized by a network of CD21+ follicular DCs (FDC) in B-cell areas, as previously observed (27), irrespective of whether germinal center reactions occurred (Fig. 1B). CD3+ T cells included variable numbers of CD8+ cytotoxic T cells and Foxp3+ Tregs; Fig. 1C).

Figure 1.

Localization of adaptive immune cells and histopathologic findings in lung adenocarcinoma. A, double immunohistochemical staining for CD3 (brown) and CD20 (green), indicating the follicular distribution of B cells and both the follicular and interfollicular distribution of T cells. Scale bar, 250 μm. B, double immunohistochemical staining for CD21 (brown) and CD20 (green). Scale bar, 100 μm. C, triple immunohistochemical staining for Foxp3 (red), CD8 (brown), and CD3 (green). Arrowheads indicate Foxp3+CD3+ Tregs, most of which are CD8. Scale bar, 25 μm. D, double immunohistochemical staining for p63 (brown) and CD20 (green), indicating the parafollicular distribution of plasma cells. Scale bar, 100 μm. E, double immunohistochemical staining for p63 (brown) and CD20 (green), indicating the interfollicular distribution of plasma cells. Scale bar, 100 μm. F, triple immunohistochemical staining for IRF4 (red), p63 (brown), and CD79α (green). Arrowheads indicate CD79α+p63lo cells (most of which express IRF4) with intermediate features between those of B cells and plasma cells. Scale bar, 25 μm. G, IRF4 positivity among B-cell and plasma cell subsets (F, follicular; IF, interfollicular). H, number of lymphoid follicles in different histologic subtypes (LPF, low-power field). I, number of CD3+ T cells in different histologic subtypes. J, CD8/CD3 ratio in different histologic subtypes. K, number of Foxp3+CD3+ Tregs in different histologic subtypes. L, number of interfollicular B cells in different histologic subtypes. M, number of parafollicular plasma cells in different histologic subtypes. N, number of interfollicular plasma cells in different histologic subtypes. Normal, non-tumor lung tissue (n = 13); AIS, adenocarcinoma in situ (n = 8); Lepidic, lepidic subtype (n = 31); Pap/Aci, papillary/acinar subtype (n = 61); Pap/Aci G3*, papillary/acinar subtype with G3 component (n = 21), Solid/MP, solid/micropapillary subtype (n = 19).

Figure 1.

Localization of adaptive immune cells and histopathologic findings in lung adenocarcinoma. A, double immunohistochemical staining for CD3 (brown) and CD20 (green), indicating the follicular distribution of B cells and both the follicular and interfollicular distribution of T cells. Scale bar, 250 μm. B, double immunohistochemical staining for CD21 (brown) and CD20 (green). Scale bar, 100 μm. C, triple immunohistochemical staining for Foxp3 (red), CD8 (brown), and CD3 (green). Arrowheads indicate Foxp3+CD3+ Tregs, most of which are CD8. Scale bar, 25 μm. D, double immunohistochemical staining for p63 (brown) and CD20 (green), indicating the parafollicular distribution of plasma cells. Scale bar, 100 μm. E, double immunohistochemical staining for p63 (brown) and CD20 (green), indicating the interfollicular distribution of plasma cells. Scale bar, 100 μm. F, triple immunohistochemical staining for IRF4 (red), p63 (brown), and CD79α (green). Arrowheads indicate CD79α+p63lo cells (most of which express IRF4) with intermediate features between those of B cells and plasma cells. Scale bar, 25 μm. G, IRF4 positivity among B-cell and plasma cell subsets (F, follicular; IF, interfollicular). H, number of lymphoid follicles in different histologic subtypes (LPF, low-power field). I, number of CD3+ T cells in different histologic subtypes. J, CD8/CD3 ratio in different histologic subtypes. K, number of Foxp3+CD3+ Tregs in different histologic subtypes. L, number of interfollicular B cells in different histologic subtypes. M, number of parafollicular plasma cells in different histologic subtypes. N, number of interfollicular plasma cells in different histologic subtypes. Normal, non-tumor lung tissue (n = 13); AIS, adenocarcinoma in situ (n = 8); Lepidic, lepidic subtype (n = 31); Pap/Aci, papillary/acinar subtype (n = 61); Pap/Aci G3*, papillary/acinar subtype with G3 component (n = 21), Solid/MP, solid/micropapillary subtype (n = 19).

Close modal

Although CD20+ B cells were mainly located in lymphoid follicles, in some cases they were diffusely distributed between the follicles, and sometimes the structures of lymphoid follicles were obscure. These interfollicular B cells were often intermingled with plasma cells (Fig. 1D and E). Intriguingly, the biologic and prognostic roles of plasma cells are largely unknown in human cancers, including lung adenocarcinoma. In lung adenocarcinoma, plasma cells were found in association with lymphoid follicles (parafollicular plasma cells; Fig. 1D) or diffusely distributed between lymphoid follicles (interfollicular plasma cells; Fig. 1E). Both of these plasma cell types may result from in situ differentiation from B cells in cancer stroma, because B cells and plasma cells were often intermingled, and large numbers of plasmablasts with transitional phenotypes between B cells and plasma cells (variable p63 expression with a CD79α+ phenotype) were observed (Fig. 1F). These plasma cells and plasmablasts infiltrating the tumor stroma expressed IRF4, a finding that is compatible with previous observations on plasma cell differentiation (Fig. 1F and G; ref. 28).

Next, we compared the extent of adaptive immune cell infiltration with the histologic subtypes of lung adenocarcinoma. The background information and clinical data of patients involved in this study are shown in Table 1. When lepidic, papillary/acinar, micropapillary, solid, and invasive mucinous subtypes were separately evaluated, 70 cases out of 114 (61.4%) contained two histologic subtypes and 6 cases out of 114 (5.3%) contained three histologic subtypes. To analyze the patterns of immune cell infiltration in different histologic subtypes in the same tumor, infiltrating immune cells were independently analyzed in each histologic subtype making up more than 20% of the total tumor volume, and the acquired data were added to the analysis (total histologic subtype dataset, N = 143). Lymphoid follicles were most frequently observed in G1/2 papillary/acinar subtypes, but their numbers decreased in high-grade histologic subtypes (Fig. 1H). The numbers of extrafollicular CD3+ T cells and Foxp3+ Tregs were higher in invasive histologic subtypes, whereas the CD8/CD3 ratio was higher in noninvasive histologic subtypes with relatively few infiltrating T cells (Fig. 1I–K). Infiltrating interfollicular B cells and plasma cells were mainly observed in invasive histologic subtypes, and their numbers were particularly high in papillary/acinar subtypes with a G3 component (Fig. 1L–N). These observations were also statistically significant (Supplementary Fig. S1A).

Profiles of infiltrating innate immune cells

Tryptase+ mast cells, chymase+ stromal mast cells, CD68+CD11c stromal macrophages, CD11c+ DCs, S100+ DCs, CD123+ pDCs, and CD15+ neutrophils were analyzed as indicators of infiltrating innate immune cells (Fig. 2A–F). Although CD11c is a well-established DC marker, most studies use S100 as a DC marker for immunohistochemical analysis. Because a recent report showed different distributions of CD11c+ DCs and S100+ DCs in colorectal carcinoma (29), we examined both populations in this study. In lung adenocarcinoma, S100+ DCs mainly resided between the tumor cells, as previously reported (30), whereas CD11c+ DCs mainly resided in cancer stroma (Fig. 2C and D). CD56+ natural killer (NK) cells were very rare in lung adenocarcinoma, as previously reported (ref. 19; data not shown), and we did not examine them further in this study.

Figure 2.

Localization of innate immune cells and histopathologic findings in lung adenocarcinoma. A, double immunohistochemical staining for tryptase (red) and CD3 (brown). Scale bar, 50 μm. B, double immunohistochemical staining for chymase (red) and CD3 (brown). Scale bar, 50 μm. C, double immunohistochemical staining for CD11c (brown) and CD68 (green), indicating the stromal distribution of CD11c+ DCs. Scale bar, 50 μm. D, immunohistochemical staining for S100, indicating epithelial distribution of S100+ DCs. Scale bar, 50 μm. E, immunohistochemical staining for CD123. Scale bar, 50 μm. F, immunohistochemical staining for CD15. Scale bar, 50 μm. G, number of tryptase+ total mast cells in different histologic subtypes. H, number of chymase+ stromal mast cells in different histologic subtypes. I, number of CD68+CD11c macrophages in different histologic subtypes. J, infiltration grade of CD11c+ DCs in different histologic subtypes. K, number of S100+ DCs in different histologic subtypes. L, number of CD123+ pDCs in different histologic subtypes. M, number of CD15+ neutrophils in different histologic subtypes. Lepidic, lepidic subtype; Solid/MP, solid/micropapillary subtype. Normal, non-tumor lung tissue (n = 13); AIS, adenocarcinoma in situ (n = 8); Lepidic, lepidic subtype (n = 31); Pap/Aci, papillary/acinar subtype (n = 61); Pap/Aci G3*, papillary/acinar subtype with G3 component (n = 21); Solid/MP, solid/micropapillary subtype (n = 19).

Figure 2.

Localization of innate immune cells and histopathologic findings in lung adenocarcinoma. A, double immunohistochemical staining for tryptase (red) and CD3 (brown). Scale bar, 50 μm. B, double immunohistochemical staining for chymase (red) and CD3 (brown). Scale bar, 50 μm. C, double immunohistochemical staining for CD11c (brown) and CD68 (green), indicating the stromal distribution of CD11c+ DCs. Scale bar, 50 μm. D, immunohistochemical staining for S100, indicating epithelial distribution of S100+ DCs. Scale bar, 50 μm. E, immunohistochemical staining for CD123. Scale bar, 50 μm. F, immunohistochemical staining for CD15. Scale bar, 50 μm. G, number of tryptase+ total mast cells in different histologic subtypes. H, number of chymase+ stromal mast cells in different histologic subtypes. I, number of CD68+CD11c macrophages in different histologic subtypes. J, infiltration grade of CD11c+ DCs in different histologic subtypes. K, number of S100+ DCs in different histologic subtypes. L, number of CD123+ pDCs in different histologic subtypes. M, number of CD15+ neutrophils in different histologic subtypes. Lepidic, lepidic subtype; Solid/MP, solid/micropapillary subtype. Normal, non-tumor lung tissue (n = 13); AIS, adenocarcinoma in situ (n = 8); Lepidic, lepidic subtype (n = 31); Pap/Aci, papillary/acinar subtype (n = 61); Pap/Aci G3*, papillary/acinar subtype with G3 component (n = 21); Solid/MP, solid/micropapillary subtype (n = 19).

Close modal

Among the innate arm of infiltrating immune cells, increased numbers of tryptase+ total mast cells and chymase+ stromal mast cells were observed in noninvasive histologic subtypes (Fig. 2G and H), whereas the numbers of other types of infiltrating innate immune cells mainly increased in invasive histologic subtypes (Fig. 2I–M). In particular, the numbers of infiltrating CD11c+ DCs, pDCs, and neutrophils increased in high-grade invasive histologic subtypes. These observations were also statistically significant (Supplementary Fig. S1B). Neutrophils were mainly found in blood and lymphatic vessels, and only a small number of neutrophils were observed in cancer stroma in a majority of cases (Fig. 2M).

Prognostic value of infiltrating immune cells

As shown in Table 1, univariate analysis indicated that the infiltration of Tregs, interfollicular B cells, plasma cells, CD11c+ DCs, and neutrophils was significantly associated with poorer prognosis when analyzed for all cases. When analyzed for stage I cases, only interfollicular B cells, plasma cells, and CD11c+ DCs were prognostically significant (Table 1 and Supplementary Figs. S2 and S3). In contrast, the infiltration of total mast cells was significantly associated with better prognosis both in total cases and stage I cases. Among the infiltrating immune cells of prognostic significance in univariate analysis (Table 1), multivariate analysis showed that the infiltration of interfollicular plasma cells serves as an independent negative prognostic factor, along with two histopathologic factors (T factor and lymph node metastasis; Supplementary Table S2). Total mast cells were selected as one of the variables in multivariate analysis but did not reach statistical significance (Supplementary Table S2).

Immunosubtypes identified by cluster analysis

The data from comprehensive evaluation of infiltrating immune cells allowed further multivariate analysis and characterization of the stromal component of lung adenocarcinoma. First, we established the correlation matrix of all infiltrating immune cell types analyzed in this study (Fig. 3A). Strong correlations (r > 0.7) were observed among the CD3+ T-cell subsets and among the B cell and plasma cell subsets (Supplementary Table S3). Among these infiltrating immune cell types with high correlation coefficients, we chose total CD3+ T cells and interfollicular plasma cells as representatives and used these two types of cells in the following cluster analysis to minimize the effects of similar variables with high correlativity. Weaker positive correlations were observed among other infiltrating immune cells other than total mast cells and CD8/CD3 ratios, both of which showed negative correlations with other infiltrating immune cell types. However, there was no correlation between total mast cells and CD8/CD3 ratios (Fig. 3A).

Figure 3.

Cluster analysis identified distinct subtypes of lung adenocarcinoma with characteristic patterns of infiltrating immune cells. A, correlation matrix including all immunologic parameters. B, cluster analysis of infiltrating immune cell data from samples of histologic subtypes making up more than 20% of tumors (total histologic subtype dataset, N = 143). Four distinct immunosubtypes were indicated. C, cluster analysis of infiltrating immune cells evaluated in the predominant histologic subtypes of lung adenocarcinomas (predominant histologic subtype dataset, N = 111). D, fraction of histologic subtypes in each immunosubtype identified in B. IM, invasive mucinous adenocarcinoma. E, fraction of histologic subtypes in different immunosubtypes clustered in C. F, changes in the numbers of indicated infiltrating immune cells in the lepidic legion (Lep) and in the adjacent papillary/acinar legion (P/A) in a single tumor, n = 16. G, RSD of the numbers of indicated infiltrating immune cells in total tumor area (Total), lepidic subtype (Lepidic), and papillary/acinar subtype (Pap/Aci) in a single tumor, n = 16. *, P < 0.01. H, numbers of indicated infiltrating immune cells in different lepidic lesions. N, non-tumor lung tissue (n = 13); A, adenocarcinoma in situ (n = 8); M, microinvasive adenocarcinoma (n = 9); L, lepidic adenocarcinoma (n = 22).

Figure 3.

Cluster analysis identified distinct subtypes of lung adenocarcinoma with characteristic patterns of infiltrating immune cells. A, correlation matrix including all immunologic parameters. B, cluster analysis of infiltrating immune cell data from samples of histologic subtypes making up more than 20% of tumors (total histologic subtype dataset, N = 143). Four distinct immunosubtypes were indicated. C, cluster analysis of infiltrating immune cells evaluated in the predominant histologic subtypes of lung adenocarcinomas (predominant histologic subtype dataset, N = 111). D, fraction of histologic subtypes in each immunosubtype identified in B. IM, invasive mucinous adenocarcinoma. E, fraction of histologic subtypes in different immunosubtypes clustered in C. F, changes in the numbers of indicated infiltrating immune cells in the lepidic legion (Lep) and in the adjacent papillary/acinar legion (P/A) in a single tumor, n = 16. G, RSD of the numbers of indicated infiltrating immune cells in total tumor area (Total), lepidic subtype (Lepidic), and papillary/acinar subtype (Pap/Aci) in a single tumor, n = 16. *, P < 0.01. H, numbers of indicated infiltrating immune cells in different lepidic lesions. N, non-tumor lung tissue (n = 13); A, adenocarcinoma in situ (n = 8); M, microinvasive adenocarcinoma (n = 9); L, lepidic adenocarcinoma (n = 22).

Close modal

Next, we conducted a cluster analysis of infiltrating immune cell types for the total histologic subtype dataset (N = 143). As shown in Fig. 3B, cases were clustered into four immunosubtypes: those characterized by high CD8/CD3 ratios with low stromal cellularity (CD8 subtype), those with increased mast cell numbers (mast cell subtype), those with increased macrophage (MØ)/DC infiltration with variable amounts of T-cell infiltration (MØ/DC subtype), and those with increased plasma cell infiltration in addition to increased macrophage/DC infiltration and variable increases in other infiltrating immune cells (T cells, Tregs, pDCs, and neutrophils; plasma cell subtype). Similar results were obtained when cluster analysis was conducted of infiltrating immune cells using the predominant histologic subtype dataset (n = 111; Fig. 3C). In this analysis, the MØ/DC subtype could be further divided into two subgroups, MØ/DC-1 and MØ/DC-2, with the latter subgroup containing higher numbers of CD11c+ DCs. Notably, these four immunosubtypes were associated with the histologic subtypes: the CD8 subtype and mast cell subtype were associated with AIS and lepidic subtypes, the MØ/DC subtype was associated with the G1/2 papillary/acinar subtype, and the plasma cell subtype was associated with high-grade histologic subtypes (Fig. 3D and E).

Within one tumor, the stromal immune cell infiltration patterns were heterogeneous, reflecting histologic heterogeneity in lung adenocarcinoma. Among the 76 cases containing two or more histologic subtypes, 40 cases (53%) contained lepidic and papillary/acinar subtypes. In these cases, increased numbers of T cells, macrophages, and DCs and decreased CD8/CD3 ratios were observed in the papillary/acinar subtypes compared with the adjacent lepidic subtype (Fig. 3F). The RSD of the number of infiltrating immune cells was significantly lower in each histologic component than those observed in total tumor area (Fig. 3G), indicating that patterns of infiltrating immune cells were more homogeneous in each histologic lesion compared with the heterogeneity observed in the total tumor area. We also compared the infiltrating immune cell types of different lepidic components (i.e., AIS, MIA, and lepidic adenocarcinoma), but no apparent differences were observed (Fig. 3H).

Plasma cell subtype as an independent prognostic factor

Among these immunosubtypes, the plasma cell subtype had the poorest prognosis (Fig. 4A). The MØ/DC-2 subtype exhibited a worse prognosis than that of MØ/DC-1. The CD8 and mast cell subtypes showed excellent prognosis after surgery. There was a significant overlap between lung adenocarcinoma cases with plasma cell infiltration (≥30 cells/HPF) and plasma cell subtype lung adenocarcinoma (Fig. 4B), and multivariate analysis revealed that the plasma cell immunosubtype was also an independent negative prognostic factor (Supplementary Table S4).

Figure 4.

Immunosubtypes of lung adenocarcinoma and their prognostic properties. A, Kaplan–Meier curves for disease-free survival (DFS) in patients stratified by immunosubtypes clustered in Fig. 3C. B, Venn diagram of cases with plasma cell infiltration (≥30 cells/HPF) and cases with plasma cell subtype stroma (J, Jaccard index). C, Kaplan–Meier curves for DFS in patients stratified by histologic subtypes. D, Kaplan–Meier curves for DFS in patients stratified by histologic subtypes and tumor grade. The fraction of immunosubtypes in each stratified population is shown in the pie charts. AIS/MIA, n = 17; Lepidic, n = 8; Pap/Aci G1/G2, n = 54; Pap/Aci G3*, n = 16; MP/solid, n = 13. E, Kaplan–Meier curves for DFS stratified by plasma cell subtype or non–plasma cell subtype. Pap/Aci G1/G2 (stage I), n = 49. F, Kaplan–Meier curves for DFS in indicated populations stratified by plasma cell infiltration.

Figure 4.

Immunosubtypes of lung adenocarcinoma and their prognostic properties. A, Kaplan–Meier curves for disease-free survival (DFS) in patients stratified by immunosubtypes clustered in Fig. 3C. B, Venn diagram of cases with plasma cell infiltration (≥30 cells/HPF) and cases with plasma cell subtype stroma (J, Jaccard index). C, Kaplan–Meier curves for DFS in patients stratified by histologic subtypes. D, Kaplan–Meier curves for DFS in patients stratified by histologic subtypes and tumor grade. The fraction of immunosubtypes in each stratified population is shown in the pie charts. AIS/MIA, n = 17; Lepidic, n = 8; Pap/Aci G1/G2, n = 54; Pap/Aci G3*, n = 16; MP/solid, n = 13. E, Kaplan–Meier curves for DFS stratified by plasma cell subtype or non–plasma cell subtype. Pap/Aci G1/G2 (stage I), n = 49. F, Kaplan–Meier curves for DFS in indicated populations stratified by plasma cell infiltration.

Close modal

The plasma cell subset was distributed among the invasive histologic subtypes, but whether its prognostic value is equally significant for each histologic subtype is currently unclear. In this study, histologic subtyping based on the WHO and IASLC/ATS/ERS classifications clearly divided the cases into distinct prognostic subgroups (Fig. 4C). In addition, the existence of the G3 component further classified papillary/acinar adenocarcinoma into two subgroups with distinct prognoses (Fig. 4D). Among these subgroups, G1/2 papillary/acinar adenocarcinoma still accounted for nearly half of all cases (n = 54). The patterns of immune cell infiltration were the most heterogeneous in this subgroup, about one-quarter of which contained plasma cell subtype stroma (Fig. 4D). Notably, analysis of G1/2 papillary/acinar adenocarcinomas based on membership of the plasma cell subtype successfully extracted cases with a higher risk of recurrence (Fig. 4E). In contrast, belonging to the plasma cell subtype was not prognostically significant for papillary/acinar adenocarcinoma with a G3 component nor for solid/micropapillary adenocarcinoma (Fig. 4E). Similar results were observed when the above populations were stratified by plasma cell infiltration (≥30 cells/HPF; Fig. 4F and Supplementary Fig. S4). In addition, stratification by plasma cell infiltration indicated significant prognostic ability with respect to stage I G1/2 papillary/acinar adenocarcinomas (Fig. 4F). Stratification by other infiltrating immune cells both in G1/2 papillary/acinar adenocarcinomas and in high-grade adenocarcinomas (papillary/acinar carcinomas with G3 component + solid/micropapillary adenocarcinoma) did not reach statistical significance, although macrophage and CD11c+ DC showed borderline significance in G1/2 papillary/acinar adenocarcinoma (Supplementary Table S5). Therefore, the plasma cell infiltration is the exclusive independent negative prognostic factor in G1/2 papillary/acinar adenocarcinoma.

Plasma cells as a major source of IL35 in cancer stroma

We further characterized the phenotype of plasma cells present in cancer stroma. More than 50% of tumor-infiltrating plasma cells produced IgG with variable proportions of IgA, IgM, and IgG4 production (Fig. 5A and Supplementary Fig. S5A). The fraction of IgA+, IgG4+, and IgM+ cells among infiltrating plasma cells neither correlated with any types of infiltrating immune cells nor was prognostically significant (Fig. 5B–D).

Figure 5.

Plasma cells in cancer stroma may produce regulatory cytokine IL35 and coexist with APRIL-producing myeloid cells. A, fraction of IgM-, IgG-, IgG4-, and IgA-positive plasma cells in cases with ≥30 plasma cells/HPF (n = 27). B, correlation matrix including the fraction of IgG4-, IgA-, and IgM-expressing plasma cells, and other immunologic parameters in cases with ≥30 plasma cells/HPF (n = 27). C, Kaplan–Meier curves for disease-free survival stratified by IgG4 positivity in cases with ≥30 plasma cells/HPF. IgG4+ < 5.4%, n = 14; ≥5.4%, n = 13. D, Kaplan–Meier curves for disease-free survival stratified by IgA positivity in cases with ≥30 plasma cells/HPF. IgA+ < 22.8%, n = 14; ≥22.8%, n = 13. E, double immunohistochemical staining for EBI3 (red) and CD20, p63, or CD138 (blue). Scale bars, 50 μm. F, EBI3 positivity among the indicated populations (n = 10). G, Ki67 positivity among the indicated populations (F, follicular; IF, interfollicular). H, double immunohistochemical staining for APRIL (red) and p63 (brown). Scale bar, 50 μm. I, number of APRIL+ cells in cancer stroma. Plasma cell subset, n = 15; non–plasma cell subset, n = 15. J, APRIL positivity among indicated populations (n = 15).

Figure 5.

Plasma cells in cancer stroma may produce regulatory cytokine IL35 and coexist with APRIL-producing myeloid cells. A, fraction of IgM-, IgG-, IgG4-, and IgA-positive plasma cells in cases with ≥30 plasma cells/HPF (n = 27). B, correlation matrix including the fraction of IgG4-, IgA-, and IgM-expressing plasma cells, and other immunologic parameters in cases with ≥30 plasma cells/HPF (n = 27). C, Kaplan–Meier curves for disease-free survival stratified by IgG4 positivity in cases with ≥30 plasma cells/HPF. IgG4+ < 5.4%, n = 14; ≥5.4%, n = 13. D, Kaplan–Meier curves for disease-free survival stratified by IgA positivity in cases with ≥30 plasma cells/HPF. IgA+ < 22.8%, n = 14; ≥22.8%, n = 13. E, double immunohistochemical staining for EBI3 (red) and CD20, p63, or CD138 (blue). Scale bars, 50 μm. F, EBI3 positivity among the indicated populations (n = 10). G, Ki67 positivity among the indicated populations (F, follicular; IF, interfollicular). H, double immunohistochemical staining for APRIL (red) and p63 (brown). Scale bar, 50 μm. I, number of APRIL+ cells in cancer stroma. Plasma cell subset, n = 15; non–plasma cell subset, n = 15. J, APRIL positivity among indicated populations (n = 15).

Close modal

Plasma cells can play a regulatory role in the immune system by producing IL35 (31), which is a heterodimer composed of p35 and EBI3. The expression of EBI3 is the rate-limiting step of IL35 production, although p35 is stably expressed in activated B and plasma cells. IL35-producing cells in tumor stroma have been detected through immunohistochemical detection of EBI3 (32), but the exact source of IL35 is still unknown. We found that these EBI3-expressing stromal cells were mainly p63+ or CD138+ plasma cells, and CD20+ B cells did not express EBI3 (Fig. 5E and F). Consistent with the previous study in which EBI3 expression increased in the course of B-cell differentiation into plasma cells, mature CD138+ plasma cells had the highest positivity for EBI3 (Fig. 5F). These observations indicate that plasma cells are one of the major sources of IL35 in cancer stroma, although we cannot exclude the possibility that some plasma cell–derived EBI3 is also utilized for IL27 formation, because activated B cells and plasma cells express a small amount of p28 (31). Studies have shown that IL35 is also produced by regulatory T cells; however, no expression of EBI3 was apparent in other lymphoid cells (Fig. 5E).

Another study has revealed that chemotherapy-induced expression of IgA and PD-L1 in plasma cells confers an immunosuppressive phenotype (33). However, all cases were chemotherapy-naïve in the current study and PD-L1 expression was neither evident in IgA+ nor IgA plasma cells (Supplementary Fig. S5B).

Myeloid cells produce APRIL in plasma cell–rich stroma

To obtain insights into the mechanism of plasma cell accumulation in cancer stroma, we analyzed the proliferation capacity of each B-cell/plasma cell lineage using Ki67 as a marker for proliferation. Ki67+ cells were sporadically observed among CD20+ interfollicular B cells; however, only a few CD79α+p63lo plasmablasts and p63high plasma cells were Ki67 positive (Fig. 5G), indicating that the accumulation of plasma cells in tumor stroma mainly resulted from the prolonged survival of plasma cells, rather than from the active proliferation of interfollicular B cells and plasmablasts. The survival of plasma cells requires B-cell activating factor (BAFF) and a proliferation-inducing ligand (APRIL) secreted by different kinds of cells depending on the context (34, 35). In lung adenocarcinoma, APRIL-producing cells were also interspersed with stromal plasma cells (Fig. 5H), and increased numbers of APRIL-producing cells were observed in cases with increased plasma cell infiltration (Fig. 5I). Most of these APRIL+ cells were CD15+ neutrophils and some were CD68+ macrophages or CD11c+ DCs (Fig. 5J), indicating that this mixed source of APRIL, including neutrophils, macrophages, and DCs, could sustain plasma cell–rich cancer stroma.

The most striking result from the current comprehensive profiling of infiltrating immune cells was that immune cell infiltration in lung adenocarcinoma can be clustered into four distinct immunosubtypes: CD8, mast cell, MØ/DC, and plasma cell subtypes. We expanded on previous immunohistochemistry-based analyses (6, 7, 19) to cover the principal components of both adaptive and innate immune cells and combined this traditional approach with cluster analysis to extract the characteristic patterns of immune cell infiltration in cancer stroma. In this way, we could identify the previously overlooked plasma cells and CD11c+ DCs as the characteristic components of immunosubtypes. Our data also allowed comparison of the patterns of immune cell infiltration with the histologic findings of cancer tissues, and the immunosubtypes identified in this study associated with histologic subtypes of lung adenocarcinoma (16, 22). CD8, mast, MØ/DC, and plasma cell subtypes were associated with low-grade to high-grade histologic subtypes, in this order, indicating that patterns of immune cell infiltration reflect the nature of each histologic subtype. The patterns of immune cell infiltration were also heterogeneous in a single tumor, reflecting heterogeneity in the histologic subtypes of lung adenocarcinoma. Therefore, the development of these patterns of infiltrating immune cells is a very localized event.

The immunosubtypes also reflected the prognosis of lung adenocarcinoma: plasma cell–subtype lung adenocarcinoma had the poorest prognosis, whereas the CD8 and mast cell subtypes had excellent prognoses. Importantly, the plasma cell subtype was identified as an independent prognostic factor in addition to other conventional histopathologic factors. Additionally, most cases belonging to the plasma cell subtype could be determined simply by evaluating the infiltration of plasma cells in cancer stroma without requiring comprehensive immunohistochemistry and cluster analysis. The presence of plasma cells in cancer stroma was also found to be an independent prognostic factor that could classify papillary/acinar adenocarcinoma without a G3 component into populations with distinctly different prognoses. Therefore, the evaluation of infiltrating immune cells in cancer stroma can provide additional prognostic information on top of that derived from routine histopathologic examinations.

Ectopic lymphoid follicle formation in cancer stroma and after plasma cell differentiation have been regarded as indicators of anticancer immune reactions (36); however, recent studies using mouse models have revealed the supportive roles of B cells and plasma cells in the development of cancer. For example, the activation of Fcγ receptors by B cell–derived immunoglobulin deposition in cancer stroma confers protumorigenic phenotypes on myeloid cells including macrophages (37). In addition, B cells and plasma cells can produce the immunosuppressive cytokines IL10 and IL35 and dampen the effector activity of T cells and anticancer immunity (38). Chemotherapy-induced, TGFβ-dependent, expression of IgA, IL10, and PD-L1 by murine plasma cells are crucial for their immunosuppressive activity during immunogenic chemotherapy (33). In human cancers, however, plasma cells are poorly defined infiltrating immune cells. Plasma cell infiltration is reportedly more frequently observed in lung squamous cell carcinoma than in adenocarcinoma (17), and the presence of higher numbers of IgG4+ plasma cells is associated with better prognosis in lung squamous cell carcinoma (39). Shalapour and colleagues (33) also observed the increased number of IgA+ plasma cells in therapy-resistant and metastatic prostate adenocarcinomas compared with early-stage prostate adenocarcinomas.

Here, we observed plasma cell–rich stroma in about one-quarter of lung adenocarcinomas, which was related to high-grade histologic subtypes. The fraction of IgG4+ or IgA+ plasma cells varied among the cases with increased plasma cell infiltration (≥30 cells/HPF), which may reflect different cytokine/chemokine milieus in each lung adenocarcinoma; however, the fraction of neither IgG4- nor IgA-producing plasma cells influenced the prognosis. We did not observe the expression of PD-L1 among stromal lymphocytes, including plasma cells, which may partly be because all cases included in the current study were neoadjuvant-naïve. Therefore, it would be interesting to analyze in future studies how the population of infiltrating immune cells and the expression of immunosuppressive molecules change in human cancers through chemotherapy.

The existence of cells producing the immunosuppressive cytokine IL35 in human cancer stroma was noted previously through the immunohistochemical detection of EBI3 (32). In the current study, we demonstrated that these EBI3-expressing stromal cells were mainly plasma cells. Although EBI3 also forms IL27 together with p28, the amount of p28 expression is relatively small. Furthermore, p35 is stably expressed in activated B and plasma cells, and there is a positive correlation between the expression of EBI3 and IL35 heterodimer production in these cells (31). Therefore, our observations indicate that the plasma cell is one of the major sources of IL35 in cancer stroma, although we cannot exclude the possibility that a small amount of this plasma cell–derived EBI3 is also utilized for IL27 production. Murine tumor models show that IL35 promotes tumor development by suppressing antitumor immunity (38). It is likely that plasma cell–derived IL35 has a similar regulatory function in human lung adenocarcinoma and could therefore be an attractive target for treatment of the disease.

We also observed that plasma cells often intermingle with B cells without germinal center reactions in lymphoid follicles, mimicking the extrafollicular differentiation of plasma cells that is induced by chronic inflammation (40). Therefore, these plasma cells mainly derive from the in situ differentiation of B cells in cancer stroma, rather than the homing of differentiated plasma cells from regional lymph nodes. These plasma cells are known to require a suitable microenvironment for their prolonged survival. In this study, we observed the co-infiltration of DCs and APRIL-producing neutrophils together with plasma cell infiltration, suggesting their possible roles in facilitating the differentiation and survival of plasma cells in cancer stroma (41).

One recent study using gene expression–based approaches characterizes plasma cells and neutrophils as positive and negative prognostic factors, respectively, in several cancer types, including lung adenocarcinoma (11). However, in the current study, plasma cell infiltration was a stronger negative prognostic factor than neutrophils in multivariate analysis. We also found that neutrophil infiltration is often increased in plasma cell immunosubtypes. Furthermore, increased infiltration of plasma cells and neutrophils was observed in no more than 30% of lung adenocarcinoma cases in the current study, and it may be difficult to predict the clinical course of lung adenocarcinoma by evaluating only the neutrophil/plasma cell ratio (11). The differences between the results of the two studies may derive from several differences in the methods to evaluate infiltrating immune cells. In the current study, increased infiltration of neutrophils was often observed around necrosis and near to obstructive pneumonitis, as well as in blood or lymphatic vessels, which we carefully excluded from analysis, but may be included in gene expression–based analysis. Therefore, comparison with histologic information is important in the analysis of infiltrating immune cells, especially in the evaluation of stromal neutrophils. More comprehensive studies of infiltrating immune cells, comparing histology-based and gene expression–based approaches, are needed.

Among the types of infiltrating immune cells analyzed in this study, only mast cells were found by univariate analysis to be associated with better prognosis. Some studies have shown that mast cell infiltration correlates with angiogenesis and poorer prognosis in early-stage lung adenocarcinoma (42, 43), whereas others have shown that the prognosis of lung adenocarcinoma is not influenced by the extent of mast cell infiltration (44, 45). Although the results of these studies are quite different with respect to the prognostic roles of mast cells, both previous studies and the current study consistently showed that mast cell infiltration is more intensive in well-differentiated tumors and low-grade histologic subtypes than in poorly differentiated and high-grade subtypes (44, 45). The findings on the prognostic values of mast cells in this study may, therefore, be attributed to the different measuring method used for mast cells; previous studies counted mast cells in regions with the highest mast cell densities (42–44), whereas we used the number of mast cells in the predominant histologic subtype (to enable valid comparisons with other types of infiltrating immune cells). Considering the conflicting associations of mast cell infiltration with prognostic values observed in previous studies, the evaluation of mast cell infiltration in the dominant histologic subtype may yield increased prognostic and clinical values.

The distribution of DCs in cancer stroma has been variously evaluated as that of immature S100+ or CD1a+ DCs (30, 46) or that of mature DC-Lamp+ DCs (36), all of which represent only subsets of tumor-infiltrating DCs. Recently, an antibody to CD11c that is effective in formalin-fixed, paraffin-embedded sections has enabled the total population of conventional DCs to be studied, and different distributions of S100+ immature DCs and CD11c+ DCs have been identified (29, 47). In the current study, we also noted that, whereas S100+ DCs were distributed among the carcinoma cells, as previously described (30), CD11c+ DCs were mainly located in the stromal component of lung adenocarcinoma. Considering the relatively low positivity of mature markers for CD11c+ DCs in previous studies (48), DCs located in cancer stroma likely possess an intermediate phenotype between those of immature and mature DCs under the influence of the chronically inflamed, but simultaneously regulatory, microenvironment of cancer stroma.

In conclusion, we showed that lung adenocarcinoma can be clustered into four distinct immunosubtypes (CD8, mast cell, MØ/DC, and plasma cell subtypes) based on the immunohistochemistry-based profiling of infiltrating immune cells, and these immunosubtypes brought additional prognostic value to those of conventional pathologic examinations. Consequently, comprehensive immunohistochemistry-based profiling of infiltrating immune cells has potential as a powerful method for analyzing the cancer stroma of various cancer tissues.

No potential conflicts of interest were disclosed.

Conception and design: Y. Kurebayashi, M. Sakamoto

Development of methodology: Y. Kurebayashi, K. Emoto, H. Asamura, M. Sakamoto

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): Y. Kurebayashi, K. Emoto, Y. Hayashi, H. Asamura, M. Sakamoto

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): Y. Kurebayashi, Y. Hayashi, T. Ohtsuka, M. Sakamoto

Writing, review, and/or revision of the manuscript: Y. Kurebayashi, T. Ohtsuka, M. Sakamoto

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases):

Study supervision: I. Kamiyama, M. Sakamoto

This work was in part supported by a Grant-in-Aid for Scientific Research (B; 26293081 to M. Sakamoto) from the Japan Society for the Promotion of Science.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Hanahan
D
,
Coussens
LM
. 
Accessories to the crime: functions of cells recruited to the tumor microenvironment
.
Cancer Cell
2012
;
21
:
309
22
.
2.
Mantovani
A
,
Allavena
P
,
Sica
A
,
Balkwill
F
. 
Cancer-related inflammation
.
Nature
2008
;
454
:
436
44
.
3.
Swann
JB
,
Smyth
MJ
. 
Immune surveillance of tumors
.
J Clin Invest
2007
;
117
:
1137
46
.
4.
Galon
J
,
Pages
F
,
Marincola
FM
,
Angell
HK
,
Thurin
M
,
Lugli
A
, et al
Cancer classification using the Immunoscore: a worldwide task force
.
J Transl Med
2012
;
10
:
205
.
5.
Okazaki
T
,
Chikuma
S
,
Iwai
Y
,
Fagarasan
S
,
Honjo
T
. 
A rheostat for immune responses: the unique properties of PD-1 and their advantages for clinical application
.
Nat Immunol
2013
;
14
:
1212
8
.
6.
Liu
M
,
Matsumura
N
,
Mandai
M
,
Li
K
,
Yagi
H
,
Baba
T
, et al
Classification using hierarchical clustering of tumor-infiltrating immune cells identifies poor prognostic ovarian cancers with high levels of COX expression
.
Mod Pathol
2009
;
22
:
373
84
.
7.
Garcia-Martinez
E
,
Gil
GL
,
Benito
AC
,
Gonzalez-Billalabeitia
E
,
Conesa
MA
,
Garcia Garcia
T
, et al
Tumor-infiltrating immune cell profiles and their change after neoadjuvant chemotherapy predict response and prognosis of breast cancer
.
Breast Cancer Res
2014
;
16
:
488
.
8.
Ganesan
AP
,
Johansson
M
,
Ruffell
B
,
Yagui-Beltran
A
,
Lau
J
,
Jablons
DM
, et al
Tumor-infiltrating regulatory T cells inhibit endogenous cytotoxic T cell responses to lung adenocarcinoma
.
J Immunol
2013
;
191
:
2009
17
.
9.
Ruffell
B
,
Au
A
,
Rugo
HS
,
Esserman
LJ
,
Hwang
ES
,
Coussens
LM
. 
Leukocyte composition of human breast cancer
.
Proc Natl Acad Sci U S A
2012
;
109
:
2796
801
.
10.
Bindea
G
,
Mlecnik
B
,
Tosolini
M
,
Kirilovsky
A
,
Waldner
M
,
Obenauf
AC
, et al
Spatiotemporal dynamics of intratumoral immune cells reveal the immune landscape in human cancer
.
Immunity
2013
;
39
:
782
95
.
11.
Gentles
AJ
,
Newman
AM
,
Liu
CL
,
Bratman
SV
,
Feng
W
,
Kim
D
, et al
The prognostic landscape of genes and infiltrating immune cells across human cancers
.
Nat Med
2015
;
21
:
938
45
.
12.
Yoshihara
K
,
Shahmoradgoli
M
,
Martinez
E
,
Vegesna
R
,
Kim
H
,
Torres-Garcia
W
, et al
Inferring tumour purity and stromal and immune cell admixture from expression data
.
Nat Commun
2013
;
4
:
2612
.
13.
Newman
AM
,
Liu
CL
,
Green
MR
,
Gentles
AJ
,
Feng
W
,
Xu
Y
, et al
Robust enumeration of cell subsets from tissue expression profiles
.
Nat Methods
2015
;
12
:
453
7
.
14.
Alizadeh
AA
,
Aranda
V
,
Bardelli
A
,
Blanpain
C
,
Bock
C
,
Borowski
C
, et al
Toward understanding and exploiting tumor heterogeneity
.
Nat Med
2015
;
21
:
846
53
.
15.
Angelo
M
,
Bendall
SC
,
Finck
R
,
Hale
MB
,
Hitzman
C
,
Borowsky
AD
, et al
Multiplexed ion beam imaging of human breast tumors
.
Nat Med
2014
;
20
:
436
42
.
16.
Travis
WD
,
Brambilla
E
,
Burke
AP
,
Marx
A
,
Nicholson
AG
. 
WHO classification of tumors of the lung, pleura, thymus and heart
. 4th ed.
France
:
International Agency for Research on Cancer
; 
2015
.
17.
Al-Shibli
KI
,
Donnem
T
,
Al-Saad
S
,
Persson
M
,
Bremnes
RM
,
Busund
LT
. 
Prognostic effect of epithelial and stromal lymphocyte infiltration in non-small cell lung cancer
.
Clin Cancer Res
2008
;
14
:
5220
7
.
18.
Inoshima
N
,
Nakanishi
Y
,
Minami
T
,
Izumi
M
,
Takayama
K
,
Yoshino
I
, et al
The influence of dendritic cell infiltration and vascular endothelial growth factor expression on the prognosis of non-small cell lung cancer
.
Clin Cancer Res
2002
;
8
:
3480
6
.
19.
Suzuki
K
,
Kadota
K
,
Sima
CS
,
Nitadori
J
,
Rusch
VW
,
Travis
WD
, et al
Clinical impact of immune microenvironment in stage I lung adenocarcinoma: tumor interleukin-12 receptor beta2 (IL-12Rbeta2), IL-7R, and stromal FoxP3/CD3 ratio are independent predictors of recurrence
.
J Clin Oncol
2013
;
31
:
490
8
.
20.
Petersen
RP
,
Campa
MJ
,
Sperlazza
J
,
Conlon
D
,
Joshi
MB
,
Harpole
DH
 Jr
, et al
Tumor infiltrating Foxp3+ regulatory T-cells are associated with recurrence in pathologic stage I NSCLC patients
.
Cancer
2006
;
107
:
2866
72
.
21.
Welsh
TJ
,
Green
RH
,
Richardson
D
,
Waller
DA
,
O'Byrne
KJ
,
Bradding
P
. 
Macrophage and mast-cell invasion of tumor cell islets confers a marked survival advantage in non-small-cell lung cancer
.
J Clin Oncol
2005
;
23
:
8959
67
.
22.
Travis
WD
,
Brambilla
E
,
Noguchi
M
,
Nicholson
AG
,
Geisinger
KR
,
Yatabe
Y
, et al
International association for the study of lung cancer/american thoracic society/european respiratory society international multidisciplinary classification of lung adenocarcinoma
.
J Thorac Oncol
2011
;
6
:
244
85
.
23.
Leslie
HS
,
Mary
KG
,
Christian
W
. 
The TNM classification of malignant tumours
. 7th ed. Hoboken, NJ:
Wiley
-
Blackwell
; 
2009
.
24.
Nichirei Biosciences
. 
Technical report 1: IHC triple staining method. Technical information
.
[cited 2015 2015/5/23]; Available from
: https://www.nichirei.co.jp/bio/english/tech_info/report001.html
25.
Glenn
WM
,
Martha
CC
. 
A study of standardization of variables in cluster analysis
.
J Classification
1988
;
5
:
181
204
.
26.
Saeed
AI
,
Sharov
V
,
White
J
,
Li
J
,
Liang
W
,
Bhagabati
N
, et al
TM4: a free, open-source system for microarray data management and analysis
.
Biotechniques
2003
;
34
:
374
8
.
27.
Germain
C
,
Gnjatic
S
,
Tamzalit
F
,
Knockaert
S
,
Remark
R
,
Goc
J
, et al
Presence of B cells in tertiary lymphoid structures is associated with a protective immunity in patients with lung cancer
.
Am J Respir Crit Care Med
2014
;
189
:
832
44
.
28.
Ochiai
K
,
Maienschein-Cline
M
,
Simonetti
G
,
Chen
J
,
Rosenthal
R
,
Brink
R
, et al
Transcriptional regulation of germinal center B and plasma cell fates by dynamical control of IRF4
.
Immunity
2013
;
38
:
918
29
.
29.
Nagorsen
D
,
Voigt
S
,
Berg
E
,
Stein
H
,
Thiel
E
,
Loddenkemper
C
. 
Tumor-infiltrating macrophages and dendritic cells in human colorectal cancer: relation to local regulatory T cells, systemic T-cell response against tumor-associated antigens and survival
.
J Transl Med
2007
;
5
:
62
.
30.
Kurabayashi
A
,
Furihata
M
,
Matsumoto
M
,
Hayashi
H
,
Ohtsuki
Y
. 
Distribution of tumor-infiltrating dendritic cells in human non-small cell lung carcinoma in relation to apoptosis
.
Pathol Int
2004
;
54
:
302
10
.
31.
Shen
P
,
Roch
T
,
Lampropoulou
V
,
O'Connor
RA
,
Stervbo
U
,
Hilgenberg
E
, et al
IL-35-producing B cells are critical regulators of immunity during autoimmune and infectious diseases
.
Nature
2014
;
507
:
366
70
.
32.
Wang
Z
,
Liu
JQ
,
Liu
Z
,
Shen
R
,
Zhang
G
,
Xu
J
, et al
Tumor-derived IL-35 promotes tumor growth by enhancing myeloid cell accumulation and angiogenesis
.
J Immunol
2013
;
190
:
2415
23
.
33.
Shalapour
S
,
Font-Burgada
J
,
Di Caro
G
,
Zhong
Z
,
Sanchez-Lopez
E
,
Dhar
D
, et al
Immunosuppressive plasma cells impede T-cell-dependent immunogenic chemotherapy
.
Nature
2015
;
521
:
94
8
.
34.
Mohr
E
,
Serre
K
,
Manz
RA
,
Cunningham
AF
,
Khan
M
,
Hardie
DL
, et al
Dendritic cells and monocyte/macrophages that create the IL-6/APRIL-rich lymph node microenvironments where plasmablasts mature
.
J Immunol
2009
;
182
:
2113
23
.
35.
Mhawech-Fauceglia
P
,
Kaya
G
,
Sauter
G
,
McKee
T
,
Donze
O
,
Schwaller
J
, et al
The source of APRIL up-regulation in human solid tumor lesions
.
J Leukoc Biol
2006
;
80
:
697
704
.
36.
Dieu-Nosjean
MC
,
Antoine
M
,
Danel
C
,
Heudes
D
,
Wislez
M
,
Poulot
V
, et al
Long-term survival for patients with non-small-cell lung cancer with intratumoral lymphoid structures
.
J Clin Oncol
2008
;
26
:
4410
7
.
37.
Andreu
P
,
Johansson
M
,
Affara
NI
,
Pucci
F
,
Tan
T
,
Junankar
S
, et al
FcRgamma activation regulates inflammation-associated squamous carcinogenesis
.
Cancer Cell
2010
;
17
:
121
34
.
38.
Collison
LW
,
Delgoffe
GM
,
Guy
CS
,
Vignali
KM
,
Chaturvedi
V
,
Fairweather
D
, et al
The composition and signaling of the IL-35 receptor are unconventional
.
Nat Immunol
2012
;
13
:
290
9
.
39.
Fujimoto
M
,
Yoshizawa
A
,
Sumiyoshi
S
,
Sonobe
M
,
Kobayashi
M
,
Koyanagi
I
, et al
Stromal plasma cells expressing immunoglobulin G4 subclass in non-small cell lung cancer
.
Hum Pathol
2013
;
44
:
1569
76
.
40.
MacLennan
IC
,
Toellner
KM
,
Cunningham
AF
,
Serre
K
,
Sze
DM
,
Zuniga
E
, et al
Extrafollicular antibody responses
.
Immunol Rev
2003
;
194
:
8
18
.
41.
Vinuesa
CG
,
Chang
PP
. 
Innate B cell helpers reveal novel types of antibody responses
.
Nat Immunol
2013
;
14
:
119
26
.
42.
Imada
A
,
Shijubo
N
,
Kojima
H
,
Abe
S
. 
Mast cells correlate with angiogenesis and poor outcome in stage I lung adenocarcinoma
.
Eur Respir J
2000
;
15
:
1087
93
.
43.
Takanami
I
,
Takeuchi
K
,
Naruke
M
. 
Mast cell density is associated with angiogenesis and poor prognosis in pulmonary adenocarcinoma
.
Cancer
2000
;
88
:
2686
92
.
44.
Nagata
M
,
Shijubo
N
,
Walls
AF
,
Ichimiya
S
,
Abe
S
,
Sato
N
. 
Chymase-positive mast cells in small sized adenocarcinoma of the lung
.
Virchows Arch
2003
;
443
:
565
73
.
45.
Carlini
MJ
,
Dalurzo
MC
,
Lastiri
JM
,
Smith
DE
,
Vasallo
BC
,
Puricelli
LI
, et al
Mast cell phenotypes and microvessels in non-small cell lung cancer and its prognostic significance
.
Hum Pathol
2010
;
41
:
697
705
.
46.
Al-Shibli
K
,
Al-Saad
S
,
Donnem
T
,
Persson
M
,
Bremnes
RM
,
Busund
LT
. 
The prognostic value of intraepithelial and stromal innate immune system cells in non-small cell lung carcinoma
.
Histopathology
2009
;
55
:
301
12
.
47.
Perez
L
,
Shurin
MR
,
Collins
B
,
Kogan
D
,
Tourkova
IL
,
Shurin
GV
. 
Comparative analysis of CD1a, S-100, CD83, and CD11c human dendritic cells in normal, premalignant, and malignant tissues
.
Histol Histopathol
2005
;
20
:
1165
72
.
48.
Perrot
I
,
Blanchard
D
,
Freymond
N
,
Isaac
S
,
Guibert
B
,
Pacheco
Y
, et al
Dendritic cells infiltrating human non-small cell lung cancer are blocked at immature stage
.
J Immunol
2007
;
178
:
2763
9
.