A narrative overview of the prognostic significance of the immune cellular milieu in tumor draining lymph nodes in non-small cell lung cancer
Review Article: Oncology: Lung Cancer

A narrative overview of the prognostic significance of the immune cellular milieu in tumor draining lymph nodes in non-small cell lung cancer

Praveen Sridhar1, Anne Sailer2

1Division of Thoracic Surgery, Department of Surgery, Boston University School of Medicine, Boston Medical Center; Boston, MA, USA; 2Department of Radiology, Yale University School of Medicine, New Haven, CT, USA

Contributions: (I) Conception and design: Both authors; (II) Administrative support: Both authors; (III) Provision of study materials or patients: None; (IV) Collection and assembly of data: Both authors; (V) Data analysis and interpretation: None; (VI) Manuscript writing: Both authors; (VII) Final approval of manuscript: Both authors.

Correspondence to: Praveen Sridhar, MD. Boston University School of Medicine, Surgical Education Office, 88 E. Newton Street, Collamore Building Suite C-515, Boston, MA 02118, USA. Email: praveen.sridhar@bmc.org.

Abstract: Lung cancer is both the most common malignancy in the United States and the most fatal. Non-small cell lung cancer accounts for most lung malignancies and involves a complex immune response that has become the recent target of systemic therapy. Late-stage disease is now effectively treated with immunotherapy. As the role of immunotherapy expands, it is becoming increasingly important to understand the biology of the immune response in patients with non-small cell lung cancer and the association of this response with response to therapy and outcomes. In this narrative overview, we review the role of immune cells comprising tumor draining lymph nodes, the structure of tumor draining lymph nodes, and their role in the tumor microenvironment. To identify relevant immune cell interactions within the tumor draining lymph nodes, we reviewed published papers focusing on tumor draining lymph nodes and delineated the significance of the immune cells to the tumor microenvironment in both animal models and humans. This is the first comprehensive review of tumor-draining lymph nodes and their role in the tumor microenvironment and provides a foundation for further investigating the tumor microenvironment and the role of humoral and innate immune response mechanisms in non-small cell lung cancer.

Keywords: Tumor draining lymph node; non-small cell lung cancer (NSCLC); immune response


Received: 04 October 2020; Accepted: 26 January 2021; Published: 25 December 2021.

doi: 10.21037/amj-20-171


Introduction

There will be an estimated 230,000 new cases of lung cancer by the end of the year 2020, and it remains the leading cause of cancer death among men and women in the United States (1). Non-small cell lung cancer (NSCLC) has a high mutational burden that contributes to host anti-tumor T cell responses through tumor-specific neoantigens. As our understanding of the immunosuppressive mechanisms that allow tumor cells to evade adaptive immunity improve, immune targets for therapy such as the program death protein and ligand (PD-1/PD-L1) and the cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) have emerged (2,3). Therapeutic targets within the immune system are limited, however, as approximately 80% of patients treated with current immune checkpoint inhibitors (ICIs) will not respond (2). Central to the tumor immune response is the activation of T cells within lymphoid structures and regional lymph nodes.

Tumor-draining lymph nodes (TDLNs) are lymphoid structures that are largely the first organs of metastases in many cancers, where the presentation of neoantigens leads to the activation of lymphocytes (4). The characteristics of tumor cells and neoantigens in lymph nodes provides one of the first insights into the likely efficacy of ICI. Additionally, TDLNs are important prognosticators in several malignancies, as approximately 30–40% of patients with lung, colorectal, and breast cancer are diagnosed at the time of nodal metastasis (5). It is critical to understand the structure and function of the cellular components of TLDNs which may provide information regarding prognosis and utility of ICI therapy.

Several populations of immune cells in the primary tumor, tumor microenvironment, and regional lymph nodes contribute to the complete immune response to NSCLC. Several studies have attempted to associate patterns of immune cellular clonality, proliferation and protein expression with disease progression and overall survival (2,6-13). Furthermore, these immune responses have been shown to be unique in the TLDN relative to the non-TLDN in several malignancies (14,15). Here we review the immune cellular response in NSCLC within TLDNs and its impact on both prognosis and therapy. We present the following article in accordance with the Narrative Review reporting checklist (available at https://amj.amegroups.com/article/view/10.21037/amj-20-171/rc).


Methodology

A literature search was performed using the MEDLINE database using a combination of keywords including “Non small cell lung cancer”, “tumor draining lymph node”, “immune response”, “T-cell”, “regulatory T-cell”, “cytotoxic T-cell”, “dendritic cell”, “natural killer cell”, “tumor associated macrophages”, “antigen presenting cell”, “B-cell”, “humoral immunity”, “innate immunity”, “immune cytoarchitecture”, and “prognosis”. Full text articles written in the English language and published between the years 2000 and 2020 were subjective assessed by the authors for content and relevance.


Clinical significance of the tumor draining lymph node

Patterns of lymph node drainage in NSCLC

Current prognostication in NSCLC is predicated upon the American Joint Committee on Cancer (AJCC) Tumor, Lymph Node, and Metastasis (TNM) staging system (4). The pattern of lymphatic spread in NSCLC has been described, though increasing scrutiny suggests that our initial understanding of this spread is limited (16,17). The theorized route of lymphatic spread occurs via intraparenchymal lymphatic drainage to the hilum and mediastinum (17,18). Variations to this traditionally described drainage have been observed, including both skip metastases and micrometastases (17,19). The impact of variable lymphatic drainage on prognosis is not well understood and raises questions regarding our fundamental knowledge of the interaction between primary tumor cells and reactive immune cells, and the biological and clinical significance of this interaction. The role of tumor evasion of secondary lymphoid functions beginning with the TDLN, such as immune cell homing, priming, and anti-tumor reactivity is not fully elucidated (20). NSCLC is one of a few cancers in which immune system evasion has been, to a degree, successfully targeted with ICI. The mechanism by which ICIs exert their influence to evade immune reactivity within the TDLN is the subject of ongoing research.

Microenvironment, cellular composition, and immune markers of the tumor draining lymph node

Lymph node microenvironment as well as the cellular composition may provide insight into the immune response and thereby the prognosis of NSCLC as previous reports have shown immunologic parameters, such as the density of tumor infiltrating lymphocytes, better predict clinical outcomes than TMN staging (13,21-25). Lymphatic flow begins with afferent lymphatic vessels draining into subcapsular sinuses on the convexity of the node and into cortical sinuses. These sinuses course between lymphatic nodules with B-cell germinal centers and progress from the outer cortex to the deeper cortex and finally the medulla of the lymph node before exiting the efferent lymphatics (26). Simply, antigen presenting cells (APCs) induce and stimulate both CD8+ (cytotoxic) and CD4+ (helper) T cells in the cortical spaces as well as humoral immunity from B-cell stimulation within the cortex and germinal centers.

Paramount to the antitumor response in TDLN in NSCLC are mature dendritic cell (DC) stimulation of CD8+ T cells (27). The relative stimulation of cytotoxic T cells to helper T cells has been studied by several investigators as a critical event in the antitumor response (9,27-29). Ito et al. demonstrated that the cytokine transforming growth factor beta-1 (TGF-β1) released by tumor cells results in DC apoptosis in TDLNs (27). Additional data in both NSCLC and other cancers suggests that the suppression of CD8+ cytotoxic T cells occurs via multiple mechanisms in NSCLC tumor microenvironment (28,29). Immunosuppressive tumor derived cytokines prevent DC maturation and lead to a tumor supportive milieu via lymphangiogenesis, blood vessel remodeling, and altered immune cell composition in cervical cancer (30). The importance of mature DCs to the anti-tumor response is not limited to the primary tumor microenvironment and TDLN. Tertiary lymphoid structures (TLS) in non-tumor bronchial associated lymphoid tissue with high mature DC density have also demonstrated favorable prognoses in NSCLC (31).

Tumor associated macrophages, or CD169+ macrophages have been examined in several animal models and some human solid tumors where they are associated with increase antitumor activity (32-34). Furthermore, CD169+ macrophage density correlated with CD8+ T cell density, and was in turn associated with fewer cases of lymph node metastases (32). While mature DC and cytotoxic T cell activation are associated with a more robust anti-tumor response and even improved prognoses, several authors have shown that their suppression occurs in conjunction with an upregulation of CD4+/CD25+ T cells (10,15). This immunosuppressive phenotype results in anergy and immune system evasion.

Cytotoxic T-lymphocyte associated protein-4 (CTLA-4) and programmed death protein-1 and its ligand (PD-1/PD-L1) have both been implicated in the immune evasion of tumors in TDLNs (9,12,15,30,35-37). The expression of PD-L1 on tumor promoting macrophages as well as the increased presence of PD-1 in TDLNs compared to peripheral blood and non-TDLNs has been shown by multiple investigators (9,15,30,36). Consequently, these proteins have increasingly been targeted for therapeutic intervention with ICI in patients with NSCLC. ICI has now improved survival stage IV NSCLC; however, ICI is not indicated in 70–80% of these late-stage patients based on their expression of PD-L1. We will next review the impact of specific immune cell populations within TDLNs in relation to prognosis and therapy.


Key cells involved in the immune response in tumor draining lymph nodes

T cell overview

In patients with early-stage lung cancer, T cells comprise nearly half of all CD45+ cells (24). CD4+ T cells made 26% of the T cell population while cytotoxic CD8+ T cells were responsible for 22% (24). Naive T cells are exposed to tumor neoantigens in lymphoid structures, thereby establishing an initial immune response. T cell migration in the TDLN occurs via high endothelial venules (HEV), which allow naïve T cells migrate into the paracortex for their initial encounter with antigen presenting DCs. This pattern of migration is necessary to activate the antitumor response (38). T cell activation and function within the TDLN is not only vital to the initial antitumor response but also to immune exhaustion and evasion through peripheral tolerance (6,7,10,12,15,39-44). T cell presence in tumors has been positively correlated with their presence in TDLNs and with survival in NSCLC, though the mechanism of this is not clear (8,11,45-54). We will provide a narrative review of the role of several T cell subtypes within the TDLN and the prognostic significance that these cellular subtypes may have in NSCLC.

CD4+ T cells

CD4+ T cells participate anti-tumor activity by priming CD8+ T cell, recruiting innate immune cells in the tumor bed, and directly promoting tumor cell death. CD4+ T cells recognize neoantigens presented on major histocompatibility (MHC) class II molecules expressed by APCs. CD4+ T cells may subsequently engage in antitumor CD8+ T cell priming or in pro-tumor regulatory T cell (Treg) generation. In the TDLN, neoantigen presentation to naive CD4+ T cells results in the maturation of T-helper (Th) cells (10). Th cells that mature into T helper type 2 cells are then involved in generating the humoral immune response via B-cell activation. Priming of the humoral immune response has been shown to facilitate carcinogenesis and tumor progression in cancer models (55). The Th1 lineage of CD4+ T cells, however, is thought to be involved in the antitumor cellular response, as the number of Th1 cells has been shown to correlate inversely with primary tumor size in patients with resectable NSCLC (2,26,55). There may be a prognostic significance of a predominant Th1 presence in the TDLN after treatment with monoclonal antibodies (anti PD-1, anti-CTLA-4, anti-CD137, against TNF alpha receptor 9, anti-CD19), as in vitro and murine models of lung carcinoma and melanoma show long-term tumor free remission. Failure to maintain the Th1 response after mAb administration coincides with recurrence with a Th2 predominant response (55).

In a lung adenocarcinoma mouse model, tumor antigen specific CD4+ T cells that are activated and proliferate within the TDLN cannot be efficiently primed, rather they become anergic peripheral Treg precursor cells (10). Data suggests that the same anergic genetic expression signature additionally arises from chronically stimulated CD4+ T cells in patients with untreated metastatic melanoma and NSCLC relative to healthy patients. These studies may signify the importance of CD4+ T cells in the mechanism of cancer immune evasion and tolerance (10). Specific CD4+ T cell subsets are present in different numbers in peripheral blood, lung, and tumor draining lymph nodes (56). In thoracic lymph nodes sampled via endobronchial ultrasound and needle aspirate in human NSCLC patients, the relative depletion of effector CD4+ in TDLNs compared to non-draining nodes has been associated with higher tumor PD-L1 expression (12). This may have implications on the selection of patients for PD-1/PD-L1 blockade. CD4+ T cells additionally play an important role in the initiation of the antitumor response not only through proliferation of the Th1 lineage but via priming of CD8+ T cells.

CD8+ T cells

Many studies have characterized the importance of tumor infiltrating CD8+ T cells and the initial response to neoantigens (3,8,45-54,57-59). To summarize, neoantigens are presented directly to T cells and are presented via APCs (45). T cell clonal expansion is associated with favorable outcomes in multiple cancers (3). Prior studies have demonstrated an association between the density of CD8+ cytotoxic effector T cells at the site of the primary tumor and survival of patients across multiple cancer models (48,57). Despite their association with improved survival and infiltration of tumor nests, antitumor cytotoxic effector T cells are not clearly functionally active within these sites. Functional studies measuring IFN-γ expression as a surrogate for CD8+ T cell activity localized CD8+ T cell activity to the peritumoral, rather than intratumoral, region. This suggests that while cytotoxic effector T cells infiltrate tumor nests, intratumoral CD8+ T cell do not mount a robust antitumor response (13). It is hypothesized that the chronic inflammatory stimulation from tumor cells may also contribute to T cell anergy (46).

The presence of a CD8+ T cell response within TDLNs in NSCLC murine models may occur prior to any clinical or radiographic manifestation of disease spread (8). In murine melanoma models, CD8+ T cell stimulation within TDLN from lymph nodes by APCs result in a more robust anti-tumoral cytotoxic response than direct stimulation from tumor cells, though the cytotoxic response even from TDLNs was muted in tumors that had been present for longer periods of time (50). Tumors that have been established for relatively long periods of time may still have high cytotoxic T cell stimulation within TDLNs, however these T cells do not complete maturation to become effective antitumor effector T cells (50). The same data did suggest that re-priming of the T cell response in TDLNs with APCs can re-invigorate T cell stimulation and maturation (50). Subsequent work has revealed the importance of CD103+CD8+ cells, or resident memory cells to the process of re-priming (49). Analysis of The Cancer Genome Atlas (TCGA) lung cancer dataset revealed that a relatively high expression of CD103+ on CD8+ cells (T resident memory) conferred a survival benefit (49).

Lastly, high PD-L1 expression in NSCLC provides immune evasion by circumventing T cell activation. Patients with NSCLC with a high level of tumor infiltrating CD8+ cells and similar expression patterns of PD-L1 in tumors and their corresponding draining lymph nodes may experience a better overall survival than patients who display variability in PD-L1 expression between tumors and their corresponding metastatic lymph nodes (Table 1) (59).

Table 1

Summary of studies with notable findings pertaining to the prognostic significance of tumor draining lymph nodes in NSCLC

Author, journal Stage N Cell type Summary of findings
Schneider et al., Journal of Thoracic Oncology 2011 I–III 20 Treg Tregs enhanced in TDLN for adenocarcinoma but not squamous cell carcinoma
Hanagiri et al., Lung Cancer 2013 I–III 158 Treg Patients with high proportion of Treg in TDLN had 63.5% 5-year survival compared to 84.4% for low Treg TDLN tumors (P=0.0056)
Resected Stage I NSCLC had 91.4% 5-year survival in Treg-low group compared to 72.1% in Treg-high group (P=0.0147)
Hanagiri et al., Anticancer Research 2014 I 131 Treg Evaluation of TDLNs for FOXP3 expression
5-year survival for low FOXP3 group 90.3% compared to 79.3% in high FOXP3 expression group (P=0.0419)
Reineke et al., EJR Open Research 2017 II–IV 11 Treg Evaluate T cell composition and PD-1 expression in TDLN
High Treg frequency in TDLN of 1 patient with rapid mortality suggesting immune evasion
Yang et al., Cancer Medicine 2018 I–IV 58 CD8+ T cell Inconsistent PD-L1 expression between primary tumors and TDLNs in nearly 40% of patients
For 60% of patients with consistent expression of PD-L1 in tumor and TDLN, CD8+ TIL correlated with longer overall survival
Murthy et al., Lung Cancer 2019 I–IV 20 Treg 12 TDLNs and non-draining lymph node aspirates via endobronchial ultrasound for evaluation of immunophenotype
Lower density of CD4+ T cells in TDLN compared to nondraining nodes
Higher Tregs in TDLN suggesting immunosuppressive phenotype in TDLNs

NSCLC, non-small cell lung cancer; TDLN, tumor-draining lymph node.

Treg

Tregs are CD25+CD4+ that express the forkhead/winged helix transcription factor (FOXP3) have been found to suppress the host immune response and present a pathway to immune evasion by inhibiting the activation and maturation of T cells (6-8,10,12,15,39,40,42,43).

Peripheral tolerance of tumor activity occurs via Treg induced anergy. This has been characterized the maturation and differentiation of precursor Treg cells from the CD4+ population. Alonso et al. demonstrated in murine models that anergic peripheral Treg are present in the TDLN cells. In contrast to studies that demonstrate associations between survival and tumor infiltrating CD8+ and CD4+ cells, there are a few studies that have demonstrated a concordant prognostic significance of Treg cells in the tumor draining lymph nodes (6-8,10,12,15,39,40,42,43).

In patients with stage I, II, and III NSCLC, a higher proportion of Tregs found within the TDLN was associated with decreased 5-year survival (6). Importantly, the 5-year survival was significantly lower in patients with stage I disease as well (6). Patients with high proportion of Treg in TDLN had 63.5% 5-year survival compared to 84.4% for low Treg TDLN tumors (P=0.0056). Resected stage I NSCLC had 91.4% 5-year survival in Treg-low group compared to 72.1% in Treg-high group (P=0.0147) (6). The relative expression of FOXP3 in TDLNs in stage I patients appears to influence survival. Hanagiri et al. demonstrated that a high relative FOXP3 expression in TDLNs was associated with a significantly worse 5-year survival (7). Specifically, 5-year survival for low FOXP3 group 90.3% compared to 79.3% in high FOXP3 expression group (P=0.0419) (7). Prior studies have shown conflicting associations between survival and Treg cell volume both generally and in tumor stroma, specifically (39,41,43,44,47). This raises the possibility that Treg expression specifically within the TDLN may carry meaningful potential as a tool for prognostication compared to tumor or systemic Treg expression.

B cells

Stankovic et al. found that B cells were the second most prevalent immune cell type in NSCLC tumors, with CD19+ B cells comprising 16% of all CD45+ immune cells (24). Kargl et al. found a 7-fold increase in B cells within the tumor region when compared to normal lung tissue in NSCLC patients (2). The percentage of naïve and plasma B cells has been shown to be similar between tumor and normal peripheral lung tissue; however memory B cells may be associated with tumor tissue in both adeno and squamous cell carcinoma of the lung (24). B cells are activated in secondary lymphoid organs in the primary follicle following antigen binding, and proliferate and expand to form secondary follicles that progress to germinal centers after stimulation from a CD4+ T effector cells (57).

There is a paucity of literature associating the B cell response within tumor draining lymph nodes and survival, however pooled data suggests that there may be an association between the presence of intra and peritumoral B cell presence and improved survival (43). There is currently no data associating the density or function of B cells within the TDLN and survival. Several studies have associated the presence of B cells within TLS in the region of the tumor with improved survival for NSCLC patients; however, these findings have not been expanded to the TDLN (29,31,43,60).

APCs

APCs are a vital initiator of the antitumor immune response, as they can activate both CD4+ and CD8+ effector cells, thereby kickstarting the immune response to cancer (52). APCs consist of tumor-associated macrophages, DCs, and natural killer (NK) cells. While there is data correlating APC intra- and peritumoral density and subtype with prognostic information, these findings have largely not been established in TDLNs (16,27,32,52,61-68). While there is no data to specifically associate survival and APC function or density within the TDLN, there are several studies that provide some insight into the mechanism of immune evasion at the level of antigen presentation and effector cell priming within the TDLN (27,41,65).

DCs

DCs migrate from tumor tissue to regional TDLNs where they stimulate naïve T cells (63). There is no survival association with DC function in TDLN; however, important data regarding immune evasion by tumor cells has been elucidated in TDLN tissue in multiple studies owing to upregulation of PDL family co-inhibitory molecules and inhibitory cytokine release from TILs (63,65). Dieu-Nosjean et al. demonstrated an association between mature DC in TLS and overall and disease-free survival in early-stage NSCLC (31).

Tumor-associated macrophages (TAMs)

Macrophages were found to comprise between 5% and 15% of the immune infiltrate in NSCLC (2,24). TAMs differentiate into either an antitumor M1 phenotype or a protumor, immunosuppressive, M2 phenotype. Macrophages have been shown to have both a favorable and an unfavorable prognostic significance in NSCLC in several studies reviewed by Suzuki et al. (13). These studies have focused primarily on peri- and intratumoral TAM density and function, rather than within the TDLN.

NK cells

NK cells are part of the innate immune system, play a key role in the host antimicrobial and antiviral response, and can initiate an anti-tumor response without the requisite tumor associated antigen (26). In both murine models and human lungs, NK cells are more differentiated relative to circulating NK cells with a lesser degree of effector function. Pooled data suggests that a relatively high intratumoral and peritumoral NK cell activity correlates to improved overall and disease-free survival (43). Similar to other APCs, there is no data to suggest that TDLN NK density or function has a similar association.


Conclusions and future directions

The importance of the immune cell milieu within the tumor and peritumor microenvironment has been studied extensively. Similar research regarding immune function and cytoarchitecture with respect to the TDLN is underway. Results from these studies have provided new insight regarding the prognostic significance of the immune cell composition and function within TDLNs as well as the potential for ICIs. While the focus of this research has been effector T cells, the lack of data with respect to the prognostic impact of APCs and B cells within TDLNs provides a new potential for future investigation.


Acknowledgments

Funding: Internal departmental funding.


Footnote

Provenance and Peer Review: This article was commissioned by the Guest Editor (Kei Suzuki) for the series “Immune Response in Lung Cancer” published in AME Medical Journal. The article has undergone external peer review.

Reporting Checklist: The authors have completed the Narrative Review reporting checklist. Available at https://amj.amegroups.com/article/view/10.21037/amj-20-171/rc

Conflicts of Interest: Both authors have completed the ICMJE uniform disclosure form (available at https://amj.amegroups.com/article/view/10.21037/amj-20-171/coif). The series “Immune Response in Lung Cancer” was commissioned by the editorial office without any funding or sponsorship. The authors have no other conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

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


References

  1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA Cancer J Clin 2020;70:7-30. [Crossref] [PubMed]
  2. Kargl J, Busch SE, Yang GH, et al. Neutrophils dominate the immune cell composition in non-small cell lung cancer. Nat Commun 2017;8:14381. [Crossref] [PubMed]
  3. Reuben A, Zhang J, Chiou SH, et al. Comprehensive T cell repertoire characterization of non-small cell lung cancer. Nat Commun. 2020;11:603. [Crossref] [PubMed]
  4. Leong SPL, Zuber M, Ferris RL, et al. Impact of nodal status and tumor burden in sentinel lymph nodes on the clinical outcomes of cancer patients. J Surg Oncol 2011;103:518-30. [Crossref] [PubMed]
  5. Steeg PS. Tumor metastasis: Mechanistic insights and clinical challenges. Nat Med 2006;12:895-904. [Crossref] [PubMed]
  6. Hanagiri T, Shigematsu Y, Shinohara S, et al. Clinical significance of the frequency of regulatory T cells in regional lymph node lymphocytes as a prognostic factor for non-small-cell lung cancer. Lung Cancer 2013;81:475-9. [Crossref] [PubMed]
  7. Hanagiri T, Fukumoto M, Koyanagi Y, et al. Regulatory T-Cells and micrometastasis in lymph nodes of stage I NSCLC. Anticancer Res 2014;34:7185-90. [PubMed]
  8. Fear VS, Forbes CA, Chee J, et al. Neo-antigen specific T cell responses indicate the presence of metastases before imaging. Sci Rep 2019;9:14640. [Crossref] [PubMed]
  9. Catacchio I, Scattone A, Silvestris N, et al. Immune Prophets of Lung Cancer: The Prognostic and Predictive Landscape of Cellular and Molecular Immune Markers. Transl Oncol 2018;11:825-35. [Crossref] [PubMed]
  10. Alonso R, Flament H, Lemoine S, et al. Induction of anergic or regulatory tumor-specific CD4+ T cells in the tumor-draining lymph node. Nat Commun 2018;9:2113. [Crossref] [PubMed]
  11. Shuang ZY, Mao YZ, Liu YC, et al. The tumor-draining lymph nodes are immunosuppressed in patients with hepatocellular carcinoma. Transl Cancer Res 2017;6:1188-96. [Crossref]
  12. Murthy V, Katzman DP, Tsay JCJ, et al. Tumor-draining lymph nodes demonstrate a suppressive immunophenotype in patients with non-small cell lung cancer assessed by endobronchial ultrasound-guided transbronchial needle aspiration: A pilot study. Lung Cancer 2019;137:94-9. [Crossref] [PubMed]
  13. Suzuki K, Kachala SS, Kadota K, et al. Prognostic immune markers in non-small cell lung cancer. Clin Cancer Res 2011;17:5247-56. [Crossref] [PubMed]
  14. Cochran AJ, Huang RR, Lee J, et al. Tumour-induced immune modulation of sentinel lymph nodes. Nat Rev Immunol 2006;6:659-70. [Crossref] [PubMed]
  15. van de Ven R, Niemeijer AN, Stam AGM, et al. High PD-1 expression on regulatory and effector T-cells in lung cancer draining lymph nodes. ERJ Open Res 2017;3:00110-2016. [Crossref] [PubMed]
  16. Kim R, Emi M, Tanabe K, et al. Immunobiology of the sentinel lymph node and its potential role for antitumour immunity. Lancet Oncol 2006;7:1006-16. [Crossref] [PubMed]
  17. Kim AW. Lymph Node Drainage Patterns and Micrometastasis in Lung Cancer. Semin Thorac Cardiovasc Surg 2009;21:298-308. [Crossref] [PubMed]
  18. Sharma A, Fidias P, Hayman LA, et al. Patterns of lymphadenopathy in thoracic malignancies. Radiographics 2004;24:419-34. [Crossref] [PubMed]
  19. Takizawa H, Sakiyama S, Tsuboi M, et al. Demonstration of the skip metastasis pathway for N2 non-small cell lung cancer. J Thorac Cardiovasc Surg 2014;147:e50-2. [Crossref] [PubMed]
  20. Carrière V, Colisson R, Jiguet-Jiglaire C, et al. Cancer cells regulate lymphocyte recruitment and leukocyte-endothelium interactions in the tumor-draining lymph node. Cancer Res 2005;65:11639-48. [Crossref] [PubMed]
  21. Kilic A, Landreneau RJ, Luketich JD, et al. 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. [Crossref] [PubMed]
  22. Horne ZD, Jack R, Gray ZT, 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;171:1-5. [Crossref] [PubMed]
  23. Li H, Wang R, Zhang D, et al. Lymph node metastasis outside of a tumor-bearing lobe in primary lung cancer and the status of interlobar fissures: The necessity for removing lymph nodes from an adjacent lobe. Medicine (Baltimore) 2019;98:e14800. [Crossref] [PubMed]
  24. Stankovic B, Bjørhovde HAK, Skarshaug R, et al. Immune Cell Composition in Human Non-small Cell Lung Cancer. Front Immunol 2019;9:3101. [Crossref] [PubMed]
  25. Liu J, Duan Y, Cheng X, et al. IL-17 is associated with poor prognosis and promotes angiogenesis via stimulating VEGF production of cancer cells in colorectal carcinoma. Biochem Biophys Res Commun 2011;407:348-54. [Crossref] [PubMed]
  26. Chandrasekaran S, King MR. Microenvironment of tumor-draining lymph nodes: opportunities for liposome-based targeted therapy. Int J Mol Sci 2014;15:20209-39. [Crossref] [PubMed]
  27. Ito M, Minamiya Y, Kawai H, et al. Tumor-Derived TGFβ-1 Induces Dendritic Cell Apoptosis in the Sentinel Lymph Node. J Immunol 2006;176:5637-43. [Crossref] [PubMed]
  28. Rotman JA, Plodkowski AJ, Hayes SA, et al. Postoperative complications after thoracic surgery for lung cancer. Clin Imaging 2015;39:735-49. [Crossref] [PubMed]
  29. Remark R, Becker C, Gomez JE, et al. The non-small cell lung cancer immune contexture: A major determinant of tumor characteristics and patient outcome. Am J Respir Crit Care Med 2015;191:377-90. [Crossref] [PubMed]
  30. Rotman J, Koster BD, Jordanova ES, et al. Unlocking the therapeutic potential of primary tumor-draining lymph nodes. Cancer Immunol Immunother 2019;68:1681-8. [Crossref] [PubMed]
  31. Dieu-Nosjean MC, Antoine M, Danel C, et al. Long-term survival for patients with non-small-cell lung cancer with intratumoral lymphoid structures. J Clin Oncol 2008;26:4410-7. [Crossref] [PubMed]
  32. Hu J, Xu J, Li M, et al. Targeting Lymph Node Sinus Macrophages to Inhibit Lymph Node Metastasis. Mol Ther Nucleic Acids 2019;16:650-62. [Crossref] [PubMed]
  33. Komohara Y, Ohnishi K, Takeya M. Possible functions of CD169-positive sinus macrophages in lymph nodes in anti-tumor immune responses. Cancer Sci 2017;108:290-5. [Crossref] [PubMed]
  34. Shiota T, Miyasato Y, Ohnishi K, et al. The Clinical Significance of CD169-Positive Lymph Node Macrophage in Patients with Breast Cancer. PLoS One 2016;11:e0166680. [Crossref] [PubMed]
  35. Buchbinder EI, Desai A. CTLA-4 and PD-1 Pathways: Similarities, Differences, and Implications of Their Inhibition. Am J Clin Oncol 2016;39:98-106. [Crossref] [PubMed]
  36. Wei S, Shreiner AB, Takeshita N, et al. Tumor-induced immune suppression of in vivo effector T-cell priming is mediated by the B7-H1/PD-1 axis and transforming growth factor β. Cancer Res 2008;68:5432-8. [Crossref] [PubMed]
  37. Fransen MF, Schoonderwoerd M, Knopf P, et al. Tumor-draining lymph nodes are pivotal in PD-1/PD-L1 checkpoint therapy. JCI Insight 2018;3:e124507. [Crossref] [PubMed]
  38. Lavin Y, Kobayashi S, Leader A, et al. Innate Immune Landscape in Early Lung Adenocarcinoma by Paired Single-Cell Analyses. Cell 2017;169:750-765.e17. [Crossref] [PubMed]
  39. Jackute J, Zemaitis M, Pranys D, et al. The prognostic influence of tumor infiltrating Foxp3(+)CD4(+), CD4(+) and CD8(+) T cells in resected non-small cell lung cancer. J Inflamm (Lond) 2015;12:63. [Crossref] [PubMed]
  40. Pircher A, Gamerith G, Amann A, et al. Lung Cancer Neoadjuvant chemo-immunotherapy modifies CD4 + CD25 + regulatory T cells (Treg) in non-small cell lung cancer (NSCLC) patients. Lung Cancer 2014;85:81-7. [Crossref] [PubMed]
  41. Schneider T, Kimpfler S, Warth A, et al. Foxp3+ regulatory T cells and natural killer cells distinctly infiltrate primary tumors and draining lymph nodes in pulmonary adenocarcinoma. J Thorac Oncol 2011;6:432-8. [Crossref] [PubMed]
  42. Shigematsu Y, Hanagiri T, Shiota H, et al. Immunosuppressive effect of regulatory T lymphocytes in lung cancer, with special reference to their effects on the induction of autologous tumor-specific cytotoxic T lymphocytes. Oncol Lett 2012;4:625-30. [Crossref] [PubMed]
  43. Soo RA, Chen Z, Yan Teng RS, et al. Prognostic significance of immune cells in non-small cell lung cancer: meta-analysis. Oncotarget 2018;9:24801-20. [Crossref] [PubMed]
  44. Zhou X, Zhao S, He Y, et al. Precise Spatiotemporal Interruption of Regulatory T-cell-Mediated CD8 þ T-cell Suppression Leads to Tumor Immunity. Cancer Res 2019;79:585-97. [Crossref] [PubMed]
  45. Bai XF, Gao JX, Liu J, et al. On the site and mode of antigen presentation for the initiation of clonal expansion of CD8 T cells specific for a natural tumor antigen. Cancer Res 2001;61:6860-7. [PubMed]
  46. Broderick L, Brooks SP, Takita H, et al. IL-12 reverses anergy to T cell receptor triggering in human lung tumor-associated memory T cells. Clin Immunol 2006;118:159-69. [Crossref] [PubMed]
  47. Barua S, Fang P, Sharma A, et al. Spatial interaction of tumor cells and regulatory T cells correlates with survival in non-small cell lung cancer. Lung Cancer 2018;117:73-9. [Crossref] [PubMed]
  48. Galon J, Costes A, Sanchez-Cabo F, et al. Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science 2006;313:1960-4. [Crossref] [PubMed]
  49. Ganesan AP, Clarke J, Wood O, et al. Tissue-resident memory features are linked to the magnitude of cytotoxic T cell responses in human lung cancer. Nat Immunol 2017;18:940-50. [Crossref] [PubMed]
  50. Hargadon KM, Brinkman CC, Sheasley-O’Neill SL, et al. Incomplete Differentiation of Antigen-Specific CD8 T Cells in Tumor-Draining Lymph Nodes. J Immunol 2006;177:6081-90. [Crossref] [PubMed]
  51. Heim L, Friedrich J, Engelhardt M, et al. NFATc1 promotes antitumoral effector functions and memory CD8+ t-cell differentiation during non-small cell lung cancer development. Cancer Res 2018;78:3619-33. [PubMed]
  52. Huang B, Liu R, Wang P, et al. CD8 + CD57 + T cells exhibit distinct features in human non-small cell lung cancer. J Immunother Cancer 2020;8:e000639. [Crossref] [PubMed]
  53. E J. CD8+CXCR5+ T cells in tumor-draining lymph nodes are highly activated and predict better prognosis in colorectal cancer. Hum Immunol 2018;79:446-52. [Crossref] [PubMed]
  54. Menares E, Gálvez-Cancino F, Cáceres-Morgado P, et al. Tissue-resident memory CD8+ T cells amplify anti-tumor immunity by triggering antigen spreading through dendritic cells. Nat Commun 2019;10:4401. [Crossref] [PubMed]
  55. Dai M, Hellstrom I, Yip YY, et al. Tumor Regression and Cure Depends on Sustained Th1 Responses. J Immunother 2018;41:369-78. [Crossref] [PubMed]
  56. Sheng SY, Gu Y, Lu CG, et al. The distribution and function of human memory T cell subsets in lung cancer. Immunol Res 2017;65:639-50. [Crossref] [PubMed]
  57. Fridman WH, Zitvogel L, Sautès-Fridman C, et al. The immune contexture in cancer prognosis and treatment. Nat Rev Clin Oncol 2017;14:717-34. [Crossref] [PubMed]
  58. Xu H, Chen X, Tao M, et al. B7-H3 and B7-H4 are independent predictors of a poor prognosis in patients with pancreatic cancer. Oncol Lett 2016;11:1841-6. [Crossref] [PubMed]
  59. Yang H, Shi J, Lin D, et al. Prognostic value of PD-L1 expression in combination with CD8+ TILs density in patients with surgically resected non-small cell lung cancer. Cancer Med 2018;7:32-45. [Crossref] [PubMed]
  60. Germain C, Gnjatic S, Tamzalit F, 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. [Crossref] [PubMed]
  61. Carus A, Ladekarl M, Hager H, et al. Lung Cancer Tumor-associated neutrophils and macrophages in non-small cell lung cancer: No immediate impact on patient outcome. Lung Cancer 2013;81:130-7. [Crossref] [PubMed]
  62. Cochran AJ, Morton DL, Stern S, et al. Sentinel lymph nodes show profound downregulation of antigen-presenting cells of the paracortex: Implications for tumor biology and treatment. Mod Pathol 2001;14:604-8. [Crossref] [PubMed]
  63. Kimura H, Dobrenkov K, Iida T, et al. Tumor-draining lymph nodes of primary lung cancer patients: A potent source of tumor-specific killer cells and dendritic cells. Anticancer Res 2005;25:85-94. [PubMed]
  64. Morisaki T, Matsumoto K, Onishi H, et al. Dendritic cell-based combined immunotherapy with autologous tumor-pulsed dendritic cell vaccine and activated T cells for cancer patients: rationale, current progress, and perspectives. Hum Cell 2003;16:175-82. [Crossref] [PubMed]
  65. Schneider T, Hoffmann H, Dienemann H, et al. Non-small cell lung cancer induces an immunosuppressive phenotype of dendritic cells in tumor microenvironment by upregulating B7-H3. J Thorac Oncol 2011;6:1162-8. [Crossref] [PubMed]
  66. Shan B, Li Q, He M, et al. Ex vivo stimulation of tumor-draining lymph node cells from lung cancer patients: A potential resource for adoptive immunotherapy. Cell Mol Immunol 2008;5:307-13. [Crossref] [PubMed]
  67. Takenoyama M, Baurain JF, Yasuda M, et al. A point mutation in the NFYC gene generates an antigenic peptide recognized by autologous cytolytic T lymphocytes on a human squamous cell lung carcinoma. Int J Cancer 2006;118:1992-7. [Crossref] [PubMed]
  68. Thomas SN, Vokali E, Lund AW, et al. Targeting the tumor-draining lymph node with adjuvanted nanoparticles reshapes the anti-tumor immune response. Biomaterials 2014;35:814-24. [Crossref] [PubMed]
doi: 10.21037/amj-20-171
Cite this article as: Sridhar P, Sailer A. A narrative overview of the prognostic significance of the immune cellular milieu in tumor draining lymph nodes in non-small cell lung cancer. AME Med J 2021;6:40.

Download Citation