Skip to main content

Advertisement

Log in

The clinical significance of tertiary lymphoid structure and its relationship with peripheral blood characteristics in patients with surgically resected non-small cell lung cancer: a single-center, retrospective study

  • Original Article
  • Published:
Cancer Immunology, Immunotherapy Aims and scope Submit manuscript

Abstract

Introduction

The presence of tertiary lymphoid structure (TLS) in tumor tissues has been reported to be a factor associated with a good prognosis in several types of cancers. However, the relationship between TLS formation and peripheral blood findings remains unclear. The purposes of the study were to evaluate the effect of the presence of TLS on survival and determine the peripheral blood characteristics associated with TLS formation in non-small cell lung cancer (NSCLC) patients.

Methods

A total of 147 consecutive NSCLC patients who underwent lung resection at Fukushima Medical University Hospital between 2013 and 2017 were enrolled. TLS expression was evaluated, and the relationships between clinical parameters and outcomes were analyzed. Peripheral blood mononuclear cells (PBMCs) were further analyzed by mass cytometry to characterize the TLS-positive microenvironment.

Results

Forty-six patients had high TLS expression, and the remaining 101 patients had low TLS expression. In stage II to IV patients (n = 35), disease-free survival was longer in the high TLS expression group (p = 0.027). A low neutrophil to lymphocyte ratio (NLR) < 2.75 in the peripheral blood was associated with high TLS expression (p = 0.003). Citrus analysis after mass cytometry assay showed that the number of cells expressing HLA-DR and CD9 in PBMCs was lower in the high TLS expression group.

Conclusion

High TLS expression is associated with a good prognosis after surgery in stage II and III NSCLC patients. In the peripheral blood, a low NLR and few antigen-presenting cells indicate the presence of TLS in the tumor microenvironment.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4

Similar content being viewed by others

Abbreviations

HEV:

High endothelial venule

ICI:

Immune checkpoint inhibitor

NLR:

Neutrophil to lymphocyte ratio

NSCLC:

Non-small cell lung cancer

PD-L1:

Programmed cell death-ligand 1

PNAd:

Peripheral node addressin

TIL:

Tumor-infiltrating lymphocyte

TLS:

Tertiary lymphoid structure

References

  1. Sung H, Ferlay J, Siegel RL et al (2021) Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: A Cancer J Clin. https://doi.org/10.3322/caac.21660

    Article  Google Scholar 

  2. Shimizu H, Okada M, Tangoku A et al (2020) Thoracic and cardiovascular surgeries in Japan during 2017. Gen Thorac Cardiovasc Surg 68:414–449. https://doi.org/10.1007/s11748-020-01298-2

    Article  PubMed  Google Scholar 

  3. Okami J, Shintani Y, Okumura M et al (2019) Demographics, safety and quality, and prognostic information in both the seventh and eighth editions of the TNM classification in 18,973 surgical cases of the Japanese joint committee of lung cancer registry database in 2010. J Thorac Oncol 14:212–222. https://doi.org/10.1016/j.jtho.2018.10.002

    Article  PubMed  Google Scholar 

  4. Taube JM, Klein A, Brahmer JR et al (2014) Association of PD-1, PD-1 ligands, and other features of the tumor immune microenvironment with response to anti-PD-1 therapy. Clin Cancer Res 20:5064–5074. https://doi.org/10.1158/1078-0432.Ccr-13-3271

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Cottrell TR, Taube JM (2018) PD-L1 and emerging biomarkers in immune checkpoint blockade therapy. Cancer J 24:41–46. https://doi.org/10.1097/ppo.0000000000000301

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Yarchoan M, Hopkins A, Jaffee EM (2017) Tumor mutational burden and response rate to PD-1 inhibition. N Engl J Med 377:2500–2501. https://doi.org/10.1056/NEJMc1713444

    Article  PubMed  PubMed Central  Google Scholar 

  7. Ozaki Y, Muto S, Takagi H et al (2020) Tumor mutation burden and immunological, genomic, and clinicopathological factors as biomarkers for checkpoint inhibitor treatment of patients with non-small-cell lung cancer. Cancer Immunol Immunother 69:127–134. https://doi.org/10.1007/s00262-019-02446-1

    Article  CAS  PubMed  Google Scholar 

  8. Subudhi SK, Aparicio A, Gao J et al (2016) Clonal expansion of CD8 T cells in the systemic circulation precedes development of ipilimumab-induced toxicities. Proc Natl Acad Sci U S A 113:11919–11924. https://doi.org/10.1073/pnas.1611421113

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Jacquelot N, Roberti MP, Enot DP et al (2017) Predictors of responses to immune checkpoint blockade in advanced melanoma. Nat Commun 8:592. https://doi.org/10.1038/s41467-017-00608-2

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Gopalakrishnan V, Spencer CN, Nezi L et al (2018) Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science 359:97–103. https://doi.org/10.1126/science.aan4236

    Article  CAS  PubMed  Google Scholar 

  11. Matson V, Fessler J, Bao R et al (2018) The commensal microbiome is associated with anti–PD-1 efficacy in metastatic melanoma patients. Science 359:104. https://doi.org/10.1126/science.aao3290

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Routy B, Le Chatelier E, Derosa L et al (2018) Gut microbiome influences efficacy of PD-1–based immunotherapy against epithelial tumors. Science 359:91. https://doi.org/10.1126/science.aan3706

    Article  CAS  PubMed  Google Scholar 

  13. Colbeck EJ, Ager A, Gallimore A et al (2017) Tertiary lymphoid structures in cancer: drivers of antitumor immunity, immunosuppression, or bystander sentinels in disease? Front Immunol 8:1830. https://doi.org/10.3389/fimmu.2017.01830

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Dieu-Nosjean MC, Giraldo NA, Kaplon H et al (2016) Tertiary lymphoid structures, drivers of the anti-tumor responses in human cancers. Immunol Rev 271:260–275. https://doi.org/10.1111/imr.12405

    Article  CAS  PubMed  Google Scholar 

  15. Tsou P, Katayama H, Ostrin EJ et al (2016) The emerging role of B cells in tumor immunity. Cancer Res 76:5597–5601. https://doi.org/10.1158/0008-5472.Can-16-0431

    Article  CAS  PubMed  Google Scholar 

  16. Fridman WH, Zitvogel L, Sautès-Fridman C et al (2017) The immune contexture in cancer prognosis and treatment. Nat Rev Clin Oncol 14:717–734. https://doi.org/10.1038/nrclinonc.2017.101

    Article  CAS  PubMed  Google Scholar 

  17. Dieu-Nosjean MC, Goc J, Giraldo NA et al (2014) Tertiary lymphoid structures in cancer and beyond. Trends Immunol 35:571–580. https://doi.org/10.1016/j.it.2014.09.006

    Article  CAS  PubMed  Google Scholar 

  18. Ager A (2017) High endothelial venules and other blood vessels: critical regulators of lymphoid organ development and function. Front Immunol 8:45. https://doi.org/10.3389/fimmu.2017.00045

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Rakaee M, Kilvaer TK, Jamaly S et al (2021) Tertiary lymphoid structure score: a promising approach to refine the TNM staging in resected non-small cell lung cancer. Br J Cancer. https://doi.org/10.1038/s41416-021-01307-y

    Article  PubMed  PubMed Central  Google Scholar 

  20. Cottrell TR, Thompson ED, Forde PM et al (2018) Pathologic features of response to neoadjuvant anti-PD-1 in resected non-small-cell lung carcinoma: a proposal for quantitative immune-related pathologic response criteria (irPRC). Ann Oncol 29:1853–1860. https://doi.org/10.1093/annonc/mdy218

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Zhu W, Germain C, Liu Z et al (2015) A high density of tertiary lymphoid structure B cells in lung tumors is associated with increased CD4(+) T cell receptor repertoire clonality. Oncoimmunology 4:e1051922. https://doi.org/10.1080/2162402x.2015.1051922

    Article  PubMed  PubMed Central  Google Scholar 

  22. E-aD Amir, Davis KL, Tadmor MD et al (2013) viSNE enables visualization of high dimensional single-cell data and reveals phenotypic heterogeneity of leukemia. Nat Biotechnol 31:545–552. https://doi.org/10.1038/nbt.2594

    Article  CAS  Google Scholar 

  23. Bruggner RV, Bodenmiller B, Dill DL et al (2014) Automated identification of stratifying signatures in cellular subpopulations. Proc Natl Acad Sci 111:E2770–E2777. https://doi.org/10.1073/pnas.1408792111

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Martinet L, Garrido I, Filleron T et al (2011) Human solid tumors contain high endothelial venules: association with T- and B-lymphocyte infiltration and favorable prognosis in breast cancer. Cancer Res 71:5678–5687. https://doi.org/10.1158/0008-5472.Can-11-0431

    Article  CAS  PubMed  Google Scholar 

  25. Remark R, Alifano M, Cremer I et al (2013) Characteristics and clinical impacts of the immune environments in colorectal and renal cell carcinoma lung metastases: influence of tumor origin. Clin Cancer Res 19:4079–4091. https://doi.org/10.1158/1078-0432.Ccr-12-3847

    Article  CAS  PubMed  Google Scholar 

  26. Dieu-Nosjean MC, Antoine M, Danel C et al (2008) Long-term survival for patients with non-small-cell lung cancer with intratumoral lymphoid structures. J Clin Oncol 26:4410–4417. https://doi.org/10.1200/jco.2007.15.0284

    Article  CAS  PubMed  Google Scholar 

  27. Ladányi A, Kiss J, Somlai B et al (2007) Density of DC-LAMP(+) mature dendritic cells in combination with activated T lymphocytes infiltrating primary cutaneous melanoma is a strong independent prognostic factor. Cancer Immunol Immunother 56:1459–1469. https://doi.org/10.1007/s00262-007-0286-3

    Article  PubMed  Google Scholar 

  28. Cabrita R, Lauss M, Sanna A et al (2020) Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature 577:561–565. https://doi.org/10.1038/s41586-019-1914-8

    Article  CAS  PubMed  Google Scholar 

  29. Calderaro J, Petitprez F, Becht E et al (2019) Intra-tumoral tertiary lymphoid structures are associated with a low risk of early recurrence of hepatocellular carcinoma. J Hepatol 70:58–65. https://doi.org/10.1016/j.jhep.2018.09.003

    Article  PubMed  Google Scholar 

  30. Mandaliya H, Jones M, Oldmeadow C et al (2019) Prognostic biomarkers in stage IV non-small cell lung cancer (NSCLC): neutrophil to lymphocyte ratio (NLR), lymphocyte to monocyte ratio (LMR), platelet to lymphocyte ratio (PLR) and advanced lung cancer inflammation index (ALI). Transl Lung Cancer Res 8:886–894. https://doi.org/10.21037/tlcr.2019.11.16

    Article  PubMed  PubMed Central  Google Scholar 

  31. Sarraf KM, Belcher E, Raevsky E et al (2009) Neutrophil/lymphocyte ratio and its association with survival after complete resection in non-small cell lung cancer. J Thorac Cardiovasc Surg 137:425–428. https://doi.org/10.1016/j.jtcvs.2008.05.046

    Article  PubMed  Google Scholar 

  32. Kersey JH, LeBien TW, Abramson CS et al (1981) P-24: a human leukemia-associated and lymphohemopoietic progenitor cell surface structure identified with monoclonal antibody. J Exp Med 153:726–731. https://doi.org/10.1084/jem.153.3.726

    Article  CAS  PubMed  Google Scholar 

  33. Fernvik EVA, HalldÉEn G, Hed JAN et al (1995) Intracellular and surface distribution of CD9 in human eosinophils. APMIS 103:699–706. https://doi.org/10.1111/j.1699-0463.1995.tb01426.x

    Article  CAS  PubMed  Google Scholar 

  34. Horváth G, Serru V, Clay D et al (1998) CD19 Is Linked to the integrin-associated tetraspans CD9, CD81, and CD82*. J Biol Chem 273:30537–30543. https://doi.org/10.1074/jbc.273.46.30537

    Article  PubMed  Google Scholar 

  35. Tai XG, Yashiro Y, Abe R et al (1996) A role for CD9 molecules in T cell activation. J Exp Med 184:753–758. https://doi.org/10.1084/jem.184.2.753

    Article  CAS  PubMed  Google Scholar 

  36. Sautès-Fridman C, Petitprez F, Calderaro J et al (2019) Tertiary lymphoid structures in the era of cancer immunotherapy. Nat Rev Cancer 19:307–325. https://doi.org/10.1038/s41568-019-0144-6

    Article  CAS  PubMed  Google Scholar 

  37. Allen E, Jabouille A, Rivera LB et al (2017) Combined antiangiogenic and anti-PD-L1 therapy stimulates tumor immunity through HEV formation. Sci Transl Med 9(385):eaak9679. https://doi.org/10.1126/scitranslmed.aak9679

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Yamane Y, Ishii G, Goto K et al (2010) A novel histopathological evaluation method predicting the outcome of non-small cell lung cancer treated by neoadjuvant therapy: the prognostic importance of the area of residual tumor. J Thorac Oncol 5:49–55. https://doi.org/10.1097/JTO.0b013e3181c0a1f8

    Article  PubMed  Google Scholar 

  39. Goc J, Germain C, Vo-Bourgais TK et al (2014) Dendritic cells in tumor-associated tertiary lymphoid structures signal a Th1 cytotoxic immune contexture and license the positive prognostic value of infiltrating CD8+ T cells. Cancer Res 74:705–715. https://doi.org/10.1158/0008-5472.Can-13-1342

    Article  CAS  PubMed  Google Scholar 

  40. Germain C, Gnjatic S, Tamzalit F et al (2014) 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 189:832–844. https://doi.org/10.1164/rccm.201309-1611OC

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors would like to thank Ms. Eiko Ohtomo, Ms. Mie Ohtsuki, and Ms. Yukiko Kikuta (Department of Chest Surgery, Fukushima Medical University, Japan) for their technical assistance.

Funding

This research did not receive any specific grant from funding agencies in the public, commercial, or non-for-profit sectors.

Author information

Authors and Affiliations

Authors

Contributions

MF, SM and HS contributed to the study conception and design. Material preparation and data collection were performed by MF, SM, SI, HY, HM, HT, YO, MW, TI, TY, NO, YM, TH, JO, MH, MH and YS. Data analysis was performed by MF and SM. The first draft of the manuscript was written by MF. Review and editing were performed by SM and HS. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Satoshi Muto.

Ethics declarations

Conflict of interest

The authors declare that they have no competing interests.

Consent to participate

Written, informed consent was obtained from all participants.

Ethical approval

This study was approved by the Human Ethics Committee at Fukushima Medical University (approval no. 30161).

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Below is the link to the electronic supplementary material.

Supplementary file1 (PDF 55 KB)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fukuhara, M., Muto, S., Inomata, S. et al. The clinical significance of tertiary lymphoid structure and its relationship with peripheral blood characteristics in patients with surgically resected non-small cell lung cancer: a single-center, retrospective study. Cancer Immunol Immunother 71, 1129–1137 (2022). https://doi.org/10.1007/s00262-021-03067-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00262-021-03067-3

Keywords

Navigation