Immune Cell Composition in Human Non-small Cell Lung Cancer

Branislava Stankovic, Heidi Anine Korsmo Bjørhovde, Renate Skarshaug, Henrik Aamodt, Astri Frafjord, Elisabeth Müller, Clara Hammarström, Kahsai Beraki, Espen S Bækkevold, Per Reidar Woldbæk, Åslaug Helland, Odd Terje Brustugun, Inger Øynebråten, Alexandre Corthay, Branislava Stankovic, Heidi Anine Korsmo Bjørhovde, Renate Skarshaug, Henrik Aamodt, Astri Frafjord, Elisabeth Müller, Clara Hammarström, Kahsai Beraki, Espen S Bækkevold, Per Reidar Woldbæk, Åslaug Helland, Odd Terje Brustugun, Inger Øynebråten, Alexandre Corthay

Abstract

Non-small cell lung cancer (NSCLC) is the leading cause of cancer-related death in the world. Immunological analysis of the tumor microenvironment (immunoscore) shows great promise for improved prognosis and prediction of response to immunotherapy. However, the exact immune cell composition in NSCLC remains unclear. Here, we used flow cytometry to characterize the immune infiltrate in NSCLC tumors, non-cancerous lung tissue, regional lymph node, and blood. The cellular identity of >95% of all CD45+ immune cells was determined. Thirteen distinct immune cell types were identified in NSCLC tumors. T cells dominated the lung cancer landscape (on average 47% of all CD45+ immune cells). CD4+ T cells were the most abundant T cell population (26%), closely followed by CD8+ T cells (22%). Double negative CD4-CD8- T cells represented a small fraction (1.4%). CD19+ B cells were the second most common immune cell type in NSCLC tumors (16%), and four different B cell sub-populations were identified. Macrophages and natural killer (NK) cells composed 4.7 and 4.5% of the immune cell infiltrate, respectively. Three types of dendritic cells (DCs) were identified (plasmacytoid DCs, CD1c+ DCs, and CD141+ DCs) which together represented 2.1% of all immune cells. Among granulocytes, neutrophils were frequent (8.6%) with a high patient-to-patient variability, while mast cells (1.4%), basophils (0.4%), and eosinophils (0.3%) were less common. Across the cohort of patients, only B cells showed a significantly higher representation in NSCLC tumors compared to the distal lung. In contrast, the percentages of macrophages and NK cells were lower in tumors than in non-cancerous lung tissue. Furthermore, the fraction of macrophages with high HLA-DR expression levels was higher in NSCLC tumors relative to distal lung tissue. To make the method readily accessible, antibody panels and flow cytometry gating strategy used to identify the various immune cells are described in detail. This work should represent a useful resource for the immunomonitoring of patients with NSCLC.

Keywords: NSCLC; flow cytometry; human lung cancer; immunomonitoring; immunoscore; tumor-infiltrating immune cells.

Figures

Figure 1
Figure 1
NSCLC tumors are infiltrated by various quantities of CD45+ leukocytes. (A–F) Data obtained by flow cytometry analysis of 57 patients, 32 with adenocarcinoma and 25 with squamous cell carcinoma. (A) The size of the events (FSC-A) was used to exclude cellular debris and define all cells. (B) Single cells were identified using FSC-A and FSC-H and doublets were gated out. (C) Live cells are negative for propidium iodide (PI) stain. (D) Live leukocytes were defined as CD45+PI− cells. (E) and (F) show leukocytes (CD45+PI−) as percent of all live cells identified by flow cytometry of (E) adenocarcinoma and (F) squamous cell carcinoma. Each symbol in the graphs (E,F) represents data from one patient. Statistical calculations were performed with non-parametric Kruskal-Wallis analysis and Dunn's post-test, comparing tumor to distal lung and lymph node (LN). Red dots in graphs (E) and (F) show patient samples also analyzed by immunohistochemistry. (G–J) Immunohistochemistry analysis of tissue sections from tumors of four NSCLC patients. HE staining (blue) and anti-CD45 staining of immune cells (brown) at 100× magnifications. The area shown in the pictures is representative of the inflammation in the tumor as a whole. (G) Adenocarcinoma tumor showing high infiltration of CD45+ cells (91% of all cells in flow cytometry). (H) Squamous cell carcinoma tumor with high infiltration of CD45+ cells (96% of all cells in flow cytometry). (I) Low infiltration of CD45+ cells in squamous cell carcinoma (45% of all cells in flow cytometry). (J) Low infiltration of CD45+ cells in adenocarcinoma tumor (52% of all cells in flow cytometry).
Figure 2
Figure 2
Flow cytometry analysis of T cells in NSCLC tumor tissue. (A) Gate for nucleated cells based on the size and complexity of the event (FCS-A and SSC-A, respectively). (B) Nucleated cells were further plotted in FSC-A and FSC-H to gate single cells and exclude doublets. (C) From the single cell gate, live leukocytes were defined as CD45+PI−. (D) A lymphocyte gate was made based on FSC-A and SSC-H. (E) CD19+ B cells were excluded from the lymphocyte population, and the CD19−CD3+ population was defined as T cells. (F) T cells were further divided in CD4+, CD8+, and CD4−CD8− populations. Each subset was examined for the naive phenotype CD45RA+CD45RO− and the effector/memory phenotype CD45RA−CD45RO+. (G) Naive/memory phenotyping of CD8+ T cells. (H) Naive/memory phenotyping of CD4−CD8− T cells. (I) Naive/memory phenotyping of CD4+ T cells. The percentages presented in the figure are average values of all NSCLC patients analyzed for T cells (n = 30; 15 adenocarcinoma, 14 squamous cell carcinoma, one large cell carcinoma). Percentages were calculated from the total number of live leukocytes (CD45+PI− population). DN, double negative T cells.
Figure 3
Figure 3
Percentage of T cells in NSCLC tumors of different histological types. Data for adenocarcinoma and squamous cell carcinoma are presented. The gating strategy is described in Figure 2. Percentages of (A) All CD3+ T cells, (B) CD3+CD4+ T cells, (C) CD3+CD8+ T cells, and (D) CD3+CD4−CD8− T cells, i.e., double negative T cells. The percentages of T cells were calculated from the total number of live leukocytes (CD45+PI−) in the sample. Each symbol represents data for one patient (n = 42; 25 adenocarcinoma, 17 squamous cell carcinoma), and mean values are indicated with blue lines. Statistical calculations were performed with non-parametric Kruskal-Wallis analysis and Dunn's post-test, and no significant differences were observed between the groups.
Figure 4
Figure 4
Flow cytometry analysis of B cell sub-populations in NSCLC tumors. (A) The FSC-A and SSC-A plot was used to identify nucleated cells. (B) In the FSC-A and FSC-H plot single cells were gated and doublets were excluded. (C) Live leukocytes were defined as CD45+PI−. (D) A lymphocyte gate was set in a FSC-A and SSC-H plot. (E) CD14+ macrophages were excluded. (F) B cell gate defining all B cells as CD19+ and CD3−. (G–I) Determination of B cell sub-populations. (G) Characterization of IgM−IgD− B cells. Two populations were identified: CD27+CD38++ plasma cells and CD27+CD38+/− cells (H) A IgD/IgM plot was used to identify an IgM+IgD− B cell sub-population and to define IgM−IgD− and IgM+IgD+ gates. (I) Further characterization of IgM+IgD+ B cells: naive B cells are defined as CD27−CD38+/−. The percentage numbers presented in the figure are average values of all NSCLC patients analyzed for the indicated B cell sub-populations (n = 23; 12 adenocarcinoma, 11 squamous cell carcinoma). For each patient, the percentages of B cells and their sub-populations were calculated from the total number of live leukocytes.
Figure 5
Figure 5
Percentage of all CD19+ B cells and B-cell sub-populations in different tissues from NSCLC patients. (A,B) Comparison of percentages of CD19+ B cells in patients diagnosed with (A) adenocarcinoma (n = 33) and (B) squamous cell carcinoma (n = 23). (C,D) Percentage of CD27+CD38+/− B cells in tissues of patients diagnosed with (C) adenocarcinoma (n = 12) and (D) squamous cell carcinoma (n = 11). (E) Presence of plasma cells in adenocarcinoma (n = 12) and (F) in squamous cell carcinoma (n = 11). (G) Percentage of IgM+IgD− B cells in adenocarcinoma (n = 12) and (H) squamous cell carcinoma (n = 11). (I) Percentage of naïve B cells in adenocarcinoma (n = 12) and (J) squamous cell carcinoma (n = 11). The cells were gated as indicated in Figure 4. Each symbol represents data from one patient as percentage of the total number of live leukocytes (CD45+PI−) and blue lines indicate mean values. Statistical analysis was performed using non-parametric Kruskal-Wallis analysis and Dunn's post-test comparing tumor, distal lung, and lymph node (LN).
Figure 6
Figure 6
Flow cytometry analysis of macrophages and DCs in NSCLC tumors. (A) FSC-A and SSC-A were used to gate nucleated cells. (B) FSC-A and FSC-H were used to gate single cells and exclude doublets. (C) Live leukocytes were defined as CD45+PI−. (E) Exclusion of CD19+ B cells. (D) Gate for HLA-DR+ and CD11c+ cells. (F) Macrophages were defined as CD14+HLA-DR+ cells. HLA-DR expression on macrophages was considered either high or low as shown in the plot. (G) Myeloid DCs were defined as CD11c+CD14−. (H) Plasmacytoid DCs were defined as HLA-DR+CD123+ and also (I) CD11c− and CD14−. (J) Two subsets of myeloid DCs were identified: CD141+ DCs and CD1c+ DCs. A double negative (DN) population was also observed. The percentages of the cell populations shown in the figure were calculated from the total number of live leukocytes and represent average values from 30 patients (16 adenocarcinoma, 13 squamous cell carcinoma and one large cell carcinoma).
Figure 7
Figure 7
Macrophages and DN myeloid cells are less abundant in NSCLC tumors than in distal lung. (A) Percentages of macrophages/monocytes in adenocarcinoma patients (n = 18), (B) squamous cell carcinoma patients (n = 14), and (C) NSCLC (n = 33). (D) Percentages of DN myeloid cells (presumably DCs and/or macrophages) in adenocarcinoma (n = 16), (E) squamous cell carcinoma (n = 13), and (F) NSCLC (n = 29). Cells were gated as shown in Figure 6. The percentages were calculated from the total number of live leukocytes (CD45+PI−). Each symbol represents data from one patient. Mean values are indicated by blue lines. Statistical calculations were performed with non-parametric Kruskal-Wallis analysis and Dunn's post-test comparing tumor, distal lung, and lymph node (LN).
Figure 8
Figure 8
Macrophages in NSCLC tumors express high levels of HLA-DR. (A) Macrophages and monocytes were divided into two populations based on the expression level of HLA-DR on the surface: those with high and those with low HLA-DR expression as indicated. (B) Percentages of macrophages expressing high levels of HLA-DR in patients with adenocarcinoma (n = 11) and squamous cell carcinoma (n = 9). Cells were gated as shown in Figure 6. The percentages were calculated from the total number of macrophages/monocytes defined as CD14+HLA-DR+ cells. Each symbol represents data from one patient. Mean values are indicated by blue lines. Statistical calculations were performed with non-parametric Kruskal-Wallis analysis and Dunn's post-test comparing tumor, distal lung, and lymph node (LN).
Figure 9
Figure 9
Flow cytometry analysis of NK cells in NSCLC tumors. (A) A FSC-A and SSC-A plot was used to identify nucleated cells. (B) Single cells were gated and doublets were excluded. (C) Live leukocytes were defined as CD45+PI− cells. (D) A lymphocyte gate was set based on the size and granularity of the cells. (E) CD19+ B cells were excluded. (F) CD14+ macrophages were excluded. (G) NK cells were defined as CD3−CD56+. (H) Two NK cell subsets were identified: CD16+ and CD16− NK cells. The percentages of all the populations were calculated from the total number of live leukocytes, and average values from 19 patients are presented (11 adenocarcinoma, eight squamous cell carcinoma).
Figure 10
Figure 10
The percentage of NK cells is lower in NSCLC tumors compared to distal lung. (A–C) Percentages of all CD56+ NK cells in different tissues of patients diagnosed with (A) adenocarcinoma (n = 11), (B) squamous cell carcinoma (n = 8), and (C) NSCLC (n = 19). (D–F) Percentages of CD56+CD16+ NK cells in (D) adenocarcinoma, (E) squamous cell carcinoma and (F) NSCLC. (G–I) Percentages of CD56+CD16− NK cells in (G) adenocarcinoma, (H) squamous cell carcinoma, and (I) NSCLC. The gating strategy is presented in Figure 9. Each symbol represents data from one patient, as percentage of all live leukocytes (CD45+PI−). Mean values are indicated with blue lines. Statistical calculations were performed with non-parametric Kruskal-Wallis analysis and Dunn's post-test comparing tumor, distal lung and lymph node (LN).
Figure 11
Figure 11
Flow cytometry analysis of granulocytes in NSCLC tumors. (A) A FSC-A and SSC-A plot was used to identify nucleated cells. (B) Single cells were gated and doublets were excluded. (C) Live leukocytes were defined as CD45+PI− cells. (D) CD3+ T cells and CD19+ B cells were excluded. (E) CD14+ macrophages were excluded. (F) CD11b was used to separate granulocytes: CD11b− cells include mast cells and basophils, whereas CD11b+ cells include neutrophils and eosinophils. (G) In the population of CD49d+ cells, basophils were distinguished from mast cells by use of CD117. (H) Mast cells and basophils are both defined as FcεR1+ cells. (I) From the CD11b+ leukocyte population, neutrophils and eosinophils were defined as CD15+ cells. (J) In the CD15+ population, neutrophils are CD49d− and eosinophils CD49d+. The percentages indicated for each granulocyte population were calculated from the total number of live leukocytes for 18 NSCLC patients (11 adenocarcinoma and seven squamous cell carcinoma).
Figure 12
Figure 12
Granulocyte infiltration in NSCLC tumors. (A,B) Mast cell infiltration in (A) adenocarcinoma (n = 11) and (B) squamous cell carcinoma (n = 7). (C,D) Neutrophil infiltration in (C) adenocarcinoma (n = 11) and (D) squamous cell carcinoma (n = 7). Each symbol represents data from one patient as percentage of all live leukocytes (CD45+PI−). The gating strategy is described in Figure 11. Mean values are indicated with blue lines. Statistical calculations were performed with non-parametric Kruskal-Wallis analysis and Dunn's post-test comparing tumor, distal lung, and lymph node (LN).
Figure 13
Figure 13
Immune cell composition in NSCLC. (A) The bar graph shows the immune cell composition in adenocarcinoma, squamous cell carcinoma, distal lung, regional lymph node, and PBMCs in the cohort of NSCLC patients (n = 68). Colors in the bars represent mean values based on data collected for the respective cell populations in the indicated tissue. Distal lung represents pooled data from non-cancerous lung tissue obtained from patients with adenocarcinoma and squamous cell carcinoma. Stars indicate significant differences in percentages of cells between tumor and distal lung calculated by Kruskal-Wallis analysis and Dunn's post-test. (B) The immune cell composition in NSCLC tumors illustrated as a pie chart.

References

    1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2017. CA Cancer J Clin. (2017) 67:7 10.3322/caac.21387
    1. Goldstraw P, Ball D, Jett JR, Le Chevalier T, Lim E, Nicholson AG, et al. . Non-small-cell lung cancer. Lancet (2011) 378:1727. 10.1016/S0140-6736(10)62101-0
    1. Travis WD, Brambilla E, Noguchi M, Nicholson AG, Geisinger K, Yatabe Y, et al. . International Association for the Study of Lung Cancer/American Thoracic Society/European Respiratory Society: international multidisciplinary classification of lung adenocarcinoma: executive summary. Proc Am Thorac Soc. (2011) 8:381. 10.1513/pats.201107-042ST
    1. Chen Z, Fillmore CM, Hammerman PS, Kim CF, Wong K-K. Non-small-cell lung cancers: a heterogeneous set of diseases. Nat Rev Cancer (2014) 14:535. 10.1038/nrc3775
    1. Mirsadraee S, Oswal D, Alizadeh Y, Caulo A, van Beek E, Jr. The 7th lung cancer TNM classification and staging system: review of the changes and implications. World J Radiol. (2012) 4:128. 10.4329/wjr.v4.i4.128
    1. 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. 10.1200/JCO.2005.01.4910
    1. Zhang L, Conejo-Garcia JR, Katsaros D, Gimotty PA, Massobrio M, Regnani G, et al. . Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. N Engl J Med. (2003) 348:203. 10.1056/NEJMoa020177
    1. Galon J, Pagès 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. 10.1186/1479-5876-10-205
    1. Naito Y, Saito K, Shiiba K, Ohuchi A, Saigenji K, Nagura H, et al. . CD8+ T cells infiltrated within cancer cell nests as a prognostic factor in human colorectal cancer. Cancer Res. (1998) 58:3491.
    1. Mahmoud SM, Paish EC, Powe DG, Macmillan RD, Grainge MJ, Lee AH, et al. . Tumor-infiltrating CD8+ lymphocytes predict clinical outcome in breast cancer. J Clin Oncol. (2011) 29:1949. 10.1200/JCO.2010.30.5037
    1. Kilic A, Landreneau RJ, Luketich JD, Pennathur A, Schuchert MJ. Density of tumor-infiltrating lymphocytes correlates with disease recurrence and survival in patients with large non-small-cell lung cancer tumors. J Surg Res. (2011) 167:207. 10.1016/j.jss.2009.08.029
    1. Horne ZD, Jack R, Gray ZT, Siegfried JM, Wilson DO, Yousem SA, et al. . Increased levels of tumor-infiltrating lymphocytes are associated with improved recurrence-free survival in stage 1A non-small-cell lung cancer. J Surg Res. (2011) 171:1. 10.1016/j.jss.2011.03.068
    1. Chen X, Wan J, Liu J, Xie W, Diao X, Xu J, et al. . Increased IL-17-producing cells correlate with poor survival and lymphangiogenesis in NSCLC patients. Lung Cancer (2010) 69:348. 10.1016/j.lungcan.2009.11.013
    1. Domagala-Kulawik J. The role of the immune system in non-small cell lung carcinoma and potential for therapeutic intervention. Transl Lung Cancer Res. (2015) 4:177. 10.3978/j.issn.2218-6751.2015.01.11
    1. Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, et al. . Safety, activity, and immune correlates of anti–PD-1 antibody in cancer. N Engl J Med. (2012) 366:2443. 10.1056/NEJMoa1200690
    1. Brahmer JR, Drake CG, Wollner I, Powderly JD, Picus J, Sharfman WH, et al. . Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. J Clin Oncol. (2010) 28:3167. 10.1200/JCO.2009.26.7609
    1. Santarpia M, Giovannetti E, Rolfo C, Karachaliou N, González-Cao M, Altavilla G, et al. . Recent developments in the use of immunotherapy in non-small cell lung cancer. Expert Rev Respir Med. (2016) 10:781. 10.1080/17476348.2016.1182866
    1. Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, et al. . Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. (2012) 366:2455. 10.1056/NEJMoa1200694
    1. Donnem T, Kilvaer TK, Andersen S, Richardsen E, Paulsen EE, Hald SM, et al. . Strategies for clinical implementation of TNM-Immunoscore in resected nonsmall-cell lung cancer. Ann Oncol. (2016) 27:225. 10.1093/annonc/mdv560
    1. Banat GA, Tretyn A, Pullamsetti SS, Wilhelm J, Weigert A, Olesch C, et al. . Immune and inflammatory cell composition of human lung cancer stroma. PLoS ONE (2015) 10:e0139073. 10.1371/journal.pone.0139073
    1. Johnson SK, Kerr KM, Chapman AD, Kennedy MM, King G, Cockburn JS, et al. . Immune cell infiltrates and prognosis in primary carcinoma of the lung. Lung Cancer (2000) 27:27. 10.1016/S0169-5002(99)00095-1
    1. Passlick B, Pantel K, Kubuschok B, Angstwurm M, Neher A, Thetter O, et al. . Expression of MHC molecules and ICAM-1 on non-small cell lung carcinomas: association with early lymphatic spread of tumour cells. Eur J Cancer (1996) 32a:141. 10.1016/0959-8049(95)00551-X
    1. 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. 10.1158/1078-0432.CCR-08-0133
    1. 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. 10.1200/JCO.2007.15.0284
    1. Ohri CM, Shikotra A, Green RH, Waller DA, Bradding P. Macrophages within NSCLC tumour islets are predominantly of a cytotoxic M1 phenotype associated with extended survival. Eur Respir J. (2009) 33:118. 10.1183/09031936.00065708
    1. 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. 10.1111/j.1365-2559.2009.03379.x
    1. Schalper KA, Brown J, Carvajal-Hausdorf D, McLaughlin J, Velcheti V, Syrigos KN, et al. . Objective measurement and clinical significance of TILs in non–small cell lung cancer. JNCI J Natl Cancer Inst. (2015) 107:dju435. 10.1093/jnci/dju435
    1. Ganesan AP, Johansson M, Ruffell B, Yagui-Beltrán 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. 10.4049/jimmunol.1301317
    1. Lizotte PH, Ivanova EV, Awad MM, Jones RE, Keogh L, Liu H, et al. . Multiparametric profiling of non-small-cell lung cancers reveals distinct immunophenotypes. JCI Insight (2016) 1:e89014. 10.1172/jci.insight.89014
    1. Kargl J, Busch SE, Yang GH, Kim KH, Hanke ML, Metz HE, et al. . Neutrophils dominate the immune cell composition in non-small cell lung cancer. Nat Commun. (2017) 8:14381. 10.1038/ncomms14381
    1. Lardinois D, De Leyn P, Van Schil P, Porta RR, Waller D, Passlick B, et al. . ESTS guidelines for intraoperative lymph node staging in non-small cell lung cancer. Eur J Cardio Thorac Surg. (2006) 30:787. 10.1016/j.ejcts.2006.08.008
    1. Silina K, Soltermann A, Attar FM, Casanova R, Uckeley ZM, Thut H, et al. . Germinal centers determine the prognostic relevance of tertiary lymphoid structures and are impaired by corticosteroids in lung squamous cell carcinoma. Cancer Res. (2018) 78:1308. 10.1158/0008-5472.CAN-17-1987
    1. Sallusto F, Lenig D, Forster R, Lipp M, Lanzavecchia A. Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature (1999) 401:708. 10.1038/44385
    1. Geginat J, Lanzavecchia A, Sallusto F. Proliferation and differentiation potential of human CD8+ memory T-cell subsets in response to antigen or homeostatic cytokines. Blood (2003) 101:4260 10.1182/blood-2002-11-3577
    1. de Chaisemartin L, Goc J, Damotte D, Validire P, Magdeleinat P, Alifano M, et al. . Characterization of chemokines and adhesion molecules associated with T cell presence in tertiary lymphoid structures in human lung cancer. Cancer Res. (2011) 71:6391. 10.1158/0008-5472.CAN-11-0952
    1. Jackson SM, Wilson PC, James JA, Capra JD. Human B cell subsets. Adv Immunol. (2008) 98:151. 10.1016/S0065-2776(08)00405-7
    1. 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. 10.1164/rccm.201309-1611OC
    1. Przekopowitz M, Kuppers R, Weniger MA. A large fraction of human tonsillar B cells expressing CD27 are germinal center B cells. Immunol Cell Biol. (2015) 93:429. 10.1038/icb.2015.6
    1. Clavarino G, Delouche N, Vettier C, Laurin D, Pernollet M, Raskovalova T, et al. . Novel strategy for phenotypic characterization of human B lymphocytes from precursors to effector cells by flow cytometry. PLoS ONE (2016) 11:e0162209. 10.1371/journal.pone.0162209
    1. Richter L, Landsverk OJB, Atlasy N, Bujko A, Yaqub S, Horneland R, et al. . Transcriptional profiling reveals monocyte-related macrophages phenotypically resembling DC in human intestine. Mucosal Immunol. (2018) 11:1512. 10.1038/s41385-018-0060-1
    1. Huang A, Zhang B, Wang B, Zhang F, Fan KX, Guo YJ. Increased CD14(+)HLA-DR (-/low) myeloid-derived suppressor cells correlate with extrathoracic metastasis and poor response to chemotherapy in non-small cell lung cancer patients. Cancer Immunol Immunother. (2013) 62:1439. 10.1007/s00262-013-1450-6
    1. Corthay A, Skovseth DK, Lundin KU, Røsjø E, Omholt H, Hofgaard PO, et al. . Primary antitumor immune response mediated by CD4+ T cells. Immunity (2005) 22:371. 10.1016/j.immuni.2005.02.003
    1. Corthay A. CD4+ T cells cooperate with macrophages for specific elimination of MHC class II-negative cancer cells. Adv Exp Med Biol. (2007) 590:195. 10.1007/978-0-387-34814-8_14
    1. Haabeth OA, Lorvik KB, Hammarström C, Donaldson IM, Haraldsen G, Bogen B, et al. . Inflammation driven by tumour-specific Th1 cells protects against B-cell cancer. Nat Commun. (2011) 2:240. 10.1038/ncomms1239
    1. Müller E, Christopoulos PF, Halder S, Lunde A, Beraki K, Speth M, et al. . Toll-like receptor ligands and interferon-gamma synergize for induction of antitumor M1 macrophages. Front Immunol. (2017) 8:1383. 10.3389/fimmu.2017.01383
    1. Müller E, Speth M, Christopoulos PF, Lunde A, Avdagic A, Øynebråten I, et al. . Both type I and type II interferons can activate antitumor M1 macrophages when combined with TLR stimulation. Front Immunol. (2018) 9:2520. 10.3389/fimmu.2018.02520
    1. Basham TY, Merigan TC. Recombinant interferon-gamma increases HLA-DR synthesis and expression. J Immunol. (1983) 130:1492.
    1. Saikh KU, Dyas B, Kissner T, Ulrich RG. CD56+-T-cell responses to bacterial superantigens and immune recognition of attenuated vaccines. Clin Diagn Lab Immunol. (2003) 10:1065. 10.1128/CDLI.10.6.1065-1073.2003
    1. Lavin Y, Kobayashi S, Leader A, Amir ED, Elefant N, Bigenwald C, et al. . Innate immune landscape in early lung adenocarcinoma by paired single-cell analyses. Cell (2017) 169:750. 10.1016/j.cell.2017.04.014
    1. Kataki A, Scheid P, Piet M, Marie B, Martinet N, Martinet Y, et al. . Tumor infiltrating lymphocytes and macrophages have a potential dual role in lung cancer by supporting both host-defense and tumor progression. J Lab Clin Med. (2002) 140:320. 10.1067/mlc.2002.128317
    1. Zocchi MR, Ferrarini M, Rugarli C. Selective lysis of the autologous tumor by delta TCS1+ gamma/delta+ tumor-infiltrating lymphocytes from human lung carcinomas. Eur J Immunol. (1990) 20:2685. 10.1002/eji.1830201224
    1. Wakabayashi O, Yamazaki K, Oizumi S, Hommura F, Kinoshita I, Ogura S, et al. CD4+ T cells in cancer stroma, not CD8+ T cells in cancer cell nests, are associated with favorable prognosis in human non-small cell lung cancers. Cancer Sci. (2003) 94:1003 10.1111/j.1349-7006.2003.tb01392.x
    1. Hiraoka K, Miyamoto M, Cho Y, Suzuoki M, Oshikiri T, Nakakubo Y, et al. . Concurrent infiltration by CD8+ T cells and CD4+ T cells is a favourable prognostic factor in non-small-cell lung carcinoma. Br J Cancer (2006) 94:275. 10.1038/sj.bjc.6602934
    1. Joly-Battaglini A, Hammarström C, Stankovic B, Aamodt H, Stjärne J, Brustugun OT, et al. . Rituximab efficiently depletes B cells in lung tumors and normal lung tissue. F1000Res (2016) 5:38. 10.12688/f1000research.7599.1
    1. Gottlin EB, Bentley RC, Campa MJ, Pisetsky DS, Herndon JE, Patz EF. The Association of Intratumoral Germinal Centers with early-stage non-small cell lung cancer. J Thorac Oncol. (2011) 6:1687. 10.1097/JTO.0b013e3182217bec
    1. Segura E, Touzot M, Bohineust A, Cappuccio A, Chiocchia G, Hosmalin A, et al. . Human inflammatory dendritic cells induce Th17 cell differentiation. Immunity (2013) 38:336. 10.1016/j.immuni.2012.10.018
    1. Haniffa M, Shin A, Bigley V, McGovern N, Teo P, See P, et al. . Human tissues contain CD141hi cross-presenting dendritic cells with functional homology to mouse CD103+ nonlymphoid dendritic cells. Immunity (2012) 37:60. 10.1016/j.immuni.2012.04.012
    1. Granot T, Senda T, Carpenter DJ, Matsuoka N, Weiner J, Gordon CL, et al. . Dendritic cells display subset and tissue-specific maturation dynamics over human life. Immunity (2017) 46:504. 10.1016/j.immuni.2017.02.019
    1. Guilliams M, Ginhoux F, Jakubzick C, Naik SH, Onai N, Schraml BU, et al. . Dendritic cells, monocytes and macrophages: a unified nomenclature based on ontogeny. Nat Rev Immunol. (2014) 14:571. 10.1038/nri3712
    1. Rajagopal D, Paturel C, Morel Y, Uematsu S, Akira S, Diebold SS. Plasmacytoid dendritic cell-derived type I interferon is crucial for the adjuvant activity of Toll-like receptor 7 agonists. Blood (2010) 115:1949. 10.1182/blood-2009-08-238543
    1. Forde PM, Chaft JE, Smith KN, Anagnostou V, Cottrell TR, Hellmann MD, et al. . Neoadjuvant PD-1 blockade in resectable lung cancer. N Engl J Med. (2018) 378:1976. 10.1056/NEJMoa1716078
    1. Carrega P, Morandi B, Costa R, Frumento G, Forte G, Altavilla G, et al. . Natural killer cells infiltrating human nonsmall-cell lung cancer are enriched in CD56 bright CD16(-) cells and display an impaired capability to kill tumor cells. Cancer (2008) 112:863. 10.1002/cncr.23239
    1. Platonova S, Cherfils-Vicini J, Damotte D, Crozet L, Vieillard V, Validire P, et al. . Profound coordinated alterations of intratumoral NK cell phenotype and function in lung carcinoma. Cancer Res. (2011) 71:5412. 10.1158/0008-5472.CAN-10-4179
    1. Eruslanov EB, Bhojnagarwala PS, Quatromoni JG, Stephen TL, Ranganathan A, Deshpande C, et al. . Tumor-associated neutrophils stimulate T cell responses in early-stage human lung cancer. J Clin Invest. (2014) 124:5466. 10.1172/JCI77053
    1. Fridlender ZG, Sun J, Kim S, Kapoor V, Cheng G, Ling L, et al. . Polarization of tumor-associated neutrophil phenotype by TGF-beta: “N1” versus “N2” TAN. Cancer Cell (2009) 16:183. 10.1016/j.ccr.2009.06.017
    1. Ilie M, Hofman V, Ortholan C, Bonnetaud C, Coëlle C, Mouroux J, et al. . Predictive clinical outcome of the intratumoral CD66b-positive neutrophil-to-CD8-positive T-cell ratio in patients with resectable nonsmall cell lung cancer. Cancer (2012) 118:1726. 10.1002/cncr.26456
    1. Carus A, Ladekarl M, Hager H, Pilegaard H, Nielsen PS, Donskov F. Tumor-associated neutrophils and macrophages in non-small cell lung cancer: no immediate impact on patient outcome. Lung Cancer (2013) 81:130. 10.1016/j.lungcan.2013.03.003

Source: PubMed

3
Subskrybuj