Skip to main content

Advertisement

Log in

Integrated analysis of tertiary lymphoid structures in relation to tumor-infiltrating lymphocytes and patient survival in pancreatic ductal adenocarcinoma

  • Original Article―Liver, Pancreas, and Biliary Tract
  • Published:
Journal of Gastroenterology Aims and scope Submit manuscript

Abstract

Background

Tertiary lymphoid structure (TLS) reflects an intense immune response against cancer, which correlates with favorable patient survival. However, the association of TLS with tumor-infiltrating lymphocytes (TILs) and clinical outcomes has not been investigated comprehensively in pancreatic ductal adenocarcinoma (PDAC).

Methods

We utilized an integrative molecular pathological epidemiology database on 162 cases with resected PDAC, and examined TLS in relation to levels of TILs, patient survival, and treatment response. In whole-section slides, we assessed the formation of TLS and conducted immunohistochemistry for tumor-infiltrating T cells (CD4, CD8, CD45RO, and FOXP3). As confounding factors, we assessed alterations of four main driver genes (KRAS, TP53, CDKN2A [p16], and SMAD4) using next-generation sequencing and immunohistochemistry, and tumor CD274 (PD-L1) expression assessed by immunohistochemistry.

Results

TLSs were found in 112 patients with PDAC (69.1%). TLS was associated with high levels of CD4+ TILs (multivariable odds ratio [OR], 3.50; 95% confidence interval [CI] 1.65–7.80; P = 0.0002), CD8+ TILs (multivariable OR, 11.0; 95% CI 4.57–29.7, P < 0.0001) and CD45RO+ TILs (multivariable OR, 2.65; 95% CI 1.25–5.80, P = 0.01), but not with levels of FOXP3+ TILs. TLS was associated with longer pancreatic cancer-specific survival (multivariable hazard ratio, 0.37; 95% CI 0.25–0.56, P < 0.0001) and favorable outcomes of adjuvant S-1-treatment. TLS was not associated with driver gene alterations but tumor CD274 negative expression.

Conclusions

Our comprehensive data supports the surrogacy of TLS for vigorous anti-tumor immune response characterized by high levels of helper and cytotoxic T cells and their prognostic role.

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

Similar content being viewed by others

Data availability

The sequence data reported are available in the DNA Data Bank of Japan (DDBJ) Sequenced Read Archive under the accession number DRA011317.

References

  1. Mizrahi JD, Surana R, Valle JW, et al. Pancreatic cancer. Lancet. 2020;395:2008–20.

    Article  CAS  PubMed  Google Scholar 

  2. Quail DF, Joyce JA. Microenvironmental regulation of tumor progression and metastasis. Nat Med. 2013;19:1423–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Ziani L, Chouaib S, Thiery J. Alteration of the antitumor immune response by cancer-associated fibroblasts. Front Immunol. 2018;9:414.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Foucher ED, Ghigo C, Chouaib S, et al. Pancreatic ductal adenocarcinoma: A strong imbalance of Good and bad immunological cops in the tumor microenvironment. Front Immunol. 2018;9:1044.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Ino Y, Yamazaki-Itoh R, Shimada K, et al. Immune cell infiltration as an indicator of the immune microenvironment of pancreatic cancer. Br J Cancer. 2013;108:914–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Yang JJ, Hu ZG, Shi WX, et al. Prognostic significance of neutrophil to lymphocyte ratio in pancreatic cancer: a meta-analysis. World J Gastroenterol. 2015;21:2807–15.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Colbeck EJ, Ager A, Gallimore A, et al. Tertiary lymphoid structures in cancer: drivers of antitumor immunity, immunosuppression, or bystander sentinels in disease? Front Immunol. 2017;8:1830.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Dieu-Nosjean MC, Giraldo NA, Kaplon H, et al. Tertiary lymphoid structures, drivers of the anti-tumor responses in human cancers. Immunol Rev. 2016;271:260–75.

    Article  CAS  PubMed  Google Scholar 

  9. Ager A. High endothelial venules and other blood vessels: critical regulators of lymphoid organ development and function. Front Immunol. 2017;8:45.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Martinet L, Garrido I, Filleron T, et al. Human solid tumors contain high endothelial venules: association with T− and B- lymphocyte infiltration and favorable prognosis in breast cancer. Cancer Res. 2011;71:5678–87.

    Article  CAS  PubMed  Google Scholar 

  11. Lee HJ, Kim JY, Park IA, et al. Prognostic significance of tumor-infiltrating lymphocytes and the tertiary lymphoid structures in HER2-positive breast cancer treated with adjuvant trastuzumab. Am J Clin Pathol. 2015;144:278–88.

    Article  CAS  PubMed  Google Scholar 

  12. 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.

    Article  CAS  PubMed  Google Scholar 

  13. Goc J, Germain C, Vo-Bourgais TKD, et al. 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. 2014;74:705–15.

    Article  CAS  PubMed  Google Scholar 

  14. Di Caro GD, Bergomas F, Grizzi F, et al. Occurrence of tertiary lymphoid tissue is associated with T-cell infiltration and predicts better prognosis in early-stage colorectal cancers. Clin Cancer Res. 2014;20:2147–58.

    Article  PubMed  Google Scholar 

  15. Posch F, Silina K, Leibl S, et al. Maturation of tertiary lymphoid structures and recurrence of stage II and III colorectal cancer. Oncoimmunology. 2018;7: e1378844.

    Article  PubMed  Google Scholar 

  16. Hiraoka N, Ino Y, Yamazaki-Itoh R, et al. Intratumoral tertiary lymphoid organ is a favourable prognosticator in patients with pancreatic cancer. Br J Cancer. 2015;112:1782–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Kuwabara S, Tsuchikawa T, Nakamura T, et al. Prognostic relevance of tertiary lymphoid organs following neoadjuvant chemoradiotherapy in pancreatic ductal adenocarcinoma. Cancer Sci. 2019;110:1853–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Waddell N, Pajic M, Patch AM, et al. Whole genomes redefine the mutational landscape of pancreatic cancer. Nature. 2015;518:495–501.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Qian ZR, Rubinson DA, Nowak JA, et al. Association of alterations in main driver genes with outcomes of patients with resected pancreatic ductal adenocarcinoma. JAMA Oncol. 2018;4: e173420.

    Article  PubMed  Google Scholar 

  20. Chen H, Chong W, Teng C, et al. The immune response-related mutational signatures and driver genes in non-small-cell lung cancer. Cancer Sci. 2019;110:2348–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Luen S, Virassamy B, Savas P, et al. The genomic landscape of breast cancer and its interaction with host immunity. Breast. 2016;29:241–50.

    Article  PubMed  Google Scholar 

  22. Giannakis M, Mu XJ, Shukla SA, et al. Genomic correlates of immune-cell infiltrates in colorectal carcinoma. Cell Rep. 2016;15:857–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Jiang X, Wang J, Deng X, et al. Role of the tumor microenvironment in PD-L1/PD-1-mediated tumor immune escape. Mol Cancer. 2019;18:10.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Iwatate Y, Hoshino I, Yokota H, et al. Radiogenomics for predicting p53 status, PD-L1 expression, and prognosis with machine learning in pancreatic cancer. Br J Cancer. 2020;123:1253–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Petrackova A, Vasinek M, Sedlarikova L, et al. Standardization of sequencing coverage depth in NGS: recommendation for detection of clonal and subclonal mutations in Cancer Diagnostics. Front Oncol. 2019;9:851.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Cancer Genome Atlas Research Network. Electronic address: andrew_aguirre@dfci.harvard.edu, Cancer Genome Atlas Research Network. Integrated genomic characterization of pancreatic ductal adenocarcinoma. Cancer Cell. 2017;32:185–203.e13.

  27. Yang SR, Lin CY, Stehr H, et al. Comprehensive genomic profiling of malignant effusions in patients with metastatic lung adenocarcinoma. J Mol Diagn. 2018;20:184–94.

    Article  PubMed  Google Scholar 

  28. Suzuki H, Aoki K, Chiba K, et al. Mutational landscape and clonal architecture in grade II and III gliomas. Nat Genet. 2015;47:458–68.

    Article  CAS  PubMed  Google Scholar 

  29. Mazaika E, Homsy J. Digital droplet PCR: CNV analysis and other applications. Curr Protoc Hum Genet. 2014;82:7.24.1-13.

    PubMed  Google Scholar 

  30. Lassaletta A, Zapotocky M, Mistry M, et al. Therapeutic and prognostic implications of BRAF V600E in pediatric low-grade gliomas. J Clin Oncol. 2017;35:2934–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Tsujimae M, Masuda A, Ikegawa T, et al. Comprehensive analysis of molecular biologic characteristics of pancreatic ductal adenocarcinoma concomitant with intraductal papillary mucinous neoplasm. Ann Surg Oncol. 2022;29:4924–34.

    Article  PubMed  Google Scholar 

  32. Yachida S, White CM, Naito Y, et al. Clinical significance of the genetic landscape of pancreatic cancer and implications for identification of potential long-term survivors. Clin Cancer Res. 2012;18:6339–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Ohtsubo K, Watanabe H, Yamaguchi Y, et al. Abnormalities of tumor suppressor gene p16 in pancreatic carcinoma: immunohistochemical and genetic findings compared with clinicopathological parameters. J Gastroenterol. 2003;38:663–71.

    Article  CAS  PubMed  Google Scholar 

  34. Tascilar M, Skinner HG, Rosty C, et al. The SMAD4 protein and prognosis of pancreatic ductal adenocarcinoma. Clin Cancer Res. 2001;7:4115–21.

    CAS  PubMed  Google Scholar 

  35. Ying S, Humbert M, Barkans J, et al. Expression of IL-4 and IL-5 mRNA and protein product by CD4+ and CD8+ T cells, eosinophils, and mast cells in bronchial biopsies obtained from atopic and nonatopic (intrinsic) asthmatics. J Immunol. 1997;158:3539–44.

    Article  CAS  PubMed  Google Scholar 

  36. Vonderheide RH. The Immune Revolution: A Case for Priming. Not Checkpoint Cancer Cell. 2018;33:563–9.

    Article  CAS  PubMed  Google Scholar 

  37. Zitvogel L, Galluzzi L, Kepp O, et al. Type I interferons in anticancer immunity. Nat Rev Immunol. 2015;15:405–14.

    Article  CAS  PubMed  Google Scholar 

  38. Hotta K, Sho M, Fujimoto K, et al. Prognostic significance of CD45RO+ memory T cells in renal cell carcinoma. Br J Cancer. 2011;105:1191–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Tan AH, Goh SY, Wong SC, et al. T helper cell-specific regulation of inducible costimulator expression via distinct mechanisms mediated by T-bet and GATA-3. J Biol Chem. 2008;283:128–36.

    Article  CAS  PubMed  Google Scholar 

  40. Masugi Y, Abe T, Ueno A, et al. Characterization of spatial distribution of tumor-infiltrating CD8 + T cells refines their prognostic utility for pancreatic cancer survival. Mod Pathol. 2019;32:1495–507.

    Article  CAS  PubMed  Google Scholar 

  41. Takada K, Kashiwagi S, Asano Y, et al. Clinical verification of the relationship between smoking and the immune microenvironment of breast cancer. J Transl Med. 2019;17:13.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Liu C, Xu B, Li Q, et al. Smoking history influences the prognostic value of peripheral naïve CD4+ T cells in advanced non-small cell lung cancer. Cancer Cell Int. 2019;19:176.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Pineda S, Maturana EL, Yu K, et al. Tumor-Infiltrating B- and T-Cell Repertoire in Pancreatic Cancer Associated With Host and Tumor Features. Front Immunol. 2021;12: 730746.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Chen DS, Mellman I. Elements of cancer immunity and the cancer-immune set point. Nature. 2017;541:321–30.

    Article  CAS  PubMed  Google Scholar 

  45. Dong H, Strome SE, Salomao DR, et al. Tumor-associated B7–H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med. 2002;8:793–800.

    Article  CAS  PubMed  Google Scholar 

  46. Chen DS, Mellman I. Oncology meets immunology: the cancer-immunity cycle. Immunity. 2013;39:1–10.

    Article  PubMed  Google Scholar 

  47. Mundry CS, Eberle KC, Singh PK, et al. Local and systemic immunosuppression in pancreatic cancer: targeting the stalwarts in tumor’s arsenal. Biochim Biophys Acta Rev Cancer. 2020;1874: 188387.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Sautès-Fridman C, Petitprez F, Calderaro J, et al. Tertiary lymphoid structures in the era of cancer immunotherapy. Nat Rev Cancer. 2019;19:307–25.

    Article  PubMed  Google Scholar 

  49. Farren MR, Mace TA, Geyer S, et al. Systemic immune activity predicts overall survival in treatment-naïve patients with metastatic pancreatic cancer. Clin Cancer Res. 2016;22:2565–74.

    Article  CAS  PubMed  Google Scholar 

  50. Lee KH, Kim EY, Yun JS, et al. The prognostic and predictive value of tumor-infiltrating lymphocytes and hematologic parameters in patients with breast cancer. BMC Cancer. 2018;18:938.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Huang Y, Lou XY, Zhu YX, et al. Local environment in biopsy better predict the pathological response to neoadjuvant chemoradiotherapy in rectal cancer. Biosci Rep 2019; 39.

  52. Uesaka K, Boku N, Fukutomi A, et al. Adjuvant chemotherapy of S-1 versus gemcitabine for resected pancreatic cancer: a phase 3, open-label, randomised, non-inferiority trial (JASPAC 01). Lancet. 2016;388:248–57.

    Article  CAS  PubMed  Google Scholar 

  53. Zitvogel L, Apetoh L, Ghiringhelli F, et al. Immunological aspects of cancer chemotherapy. Nat Rev Immunol. 2008;8:59–73.

    Article  CAS  PubMed  Google Scholar 

  54. Adachi Y, Taketani S, Oyaizu H, et al. Apoptosis of colorectal adenocarcinoma induced by 5-FU and/or IFN-gamma through caspase 3 and caspase 8. Int J Oncol. 1999;15:1191–6.

    CAS  PubMed  Google Scholar 

  55. Dunn GP, Koebel CM, Schreiber RD. Interferons, immunity and cancer Immunoediting. Nat Rev Immunol. 2006;6:836–48.

    Article  CAS  PubMed  Google Scholar 

  56. Kursunel MA, Esendagli G. The untold story of IFN-γ in cancer biology. Cytokine Growth Factor Rev. 2016;31:73–81.

    Article  PubMed  Google Scholar 

  57. Choudhry H, Helmi N, Abdulaal WH, et al. Prospects of IL-2 in cancer immunotherapy. Biomed Res Int. 2018; p. 9056173.

  58. Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science. 2015;348:69–74.

    Article  CAS  PubMed  Google Scholar 

  59. Pu N, Chen Q, Gao S, Liu G, Zhu Y, Yin L, et al. Genetic landscape of prognostic value in pancreatic ductal adenocarcinoma microenvironment. Ann Transl Med. 2019;7:645.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Wartenberg M, Cibin S, Zlobec I, et al. Integrated genomic and immunophenotypic classification of pancreatic cancer reveals three distinct subtypes with prognostic/predictive significance. Clin Cancer Res. 2018;24:4444–54.

    Article  CAS  PubMed  Google Scholar 

  61. Cheng H, Fan K, Luo G, et al. Kras G12D mutation contributes to regulatory T cell conversion through activation of the MEK/ERK pathway in pancreatic cancer. Cancer Lett. 2019;446:103–11.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors would like to thank Takashi Omori (Clinical & Translational Research Center, Kobe University), Nobuyuki Kakiuchi (Department of Pathology and Tumor Biology, Kyoto University) and Yohei Masugi (Department of Pathology, Keio University School of Medicine) for their valuable comments. We would like to thank Editage (www.edita ge.jp) for the English language editing.

Funding

This work was supported by JSPS KAKENHI (Grants-in-Aid for Scientific Research), Grant no. 19K08444 (A.M.), and Grant no. 19H03698 (Y.K.). This work was also supported by Pancreas Research Foundation of Japan (A.M.).

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed to the study conception and design. Material preparation, data collection and analysis were performed by TT, JI, TI, ST, RN, YY, SA, MT, KY, SA, MG, SM, NI, HU, SK and KN. The first draft of the manuscript was written by TT, and AM designed the study concept, analyzed data, and wrote the manuscript. MK and TI reviewed all slides in the study and advised on pathological diagnosis as needed. TH, HT, KS, HS, AS, TK, YU, TF and YK were involved in study supervision and revised the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Atsuhiro Masuda.

Ethics declarations

Conflict of interest

The authors declare no potential conflicts of interest.

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.

535_2022_1939_MOESM1_ESM.tif

Supplementary Fig. 1 Representative immuno-histochemistry images. a Typical images of high or low levels of CD4+, CD8+, CD45RO+ and FOXP3+ TILs. b Typical images of presence or absence of CD274 (PD-L1) expression. c Typical images of intact or altered (lost or over expression) of protein expression of TP53, CDKN2A and SMAD4. Original magnification (a-c, 200×). Scale bars, 50 μm (a-c). PDL1, Programmed death ligand 1; TILs, tumor-infiltrating lymphocytes

535_2022_1939_MOESM2_ESM.tif

Supplementary Fig. 2 CSS of PDAC patients according to the intratumoral TLS and peritumoral TLS. Intratumoral TLSs were detected within and around the tumor; peritumoral TLSs were detected only around the tumor. The CSS curves were estimated using the Kaplan–Meier method, and the significant differences between the two groups were evaluated using a log-rank test. The number of patients at risk is shown in the CSS curves. CSS, cancer-specific survival; PDAC, pancreatic ductal adenocarcinoma; TLS, tertiary lymphoid structure

535_2022_1939_MOESM3_ESM.tif

Supplementary Fig. 3 CSS of PDAC patients according to the level of NLR or extent of CD8+ TILs. The CSS of PDAC patients according to high or low NLR were investigated. The differences of the CSS in adjuvant chemotherapy among the groups treated with S-1, gemcitabine, and non-treated groups according to the high or low NLR were also examined. a All patients b Patients with low NLR c Patients with high NLR. The CSS of PDAC according to the high or low levels of CD8+ TILs were investigated. The differences of the CSS in adjuvant chemotherapy among the groups treated with S-1, gemcitabine, and non-treated groups according to the high or low levels of CD8+ TILs were also examined. d All patients e Patients with high levels of CD8+ TILs f Patients with low levels of CD8+ TILs. The CSS curves were estimated using the Kaplan-Meier method, and the significant differences between the two groups were evaluated by a log-rank test. The number of patients at risk is shown in the CSS curves. CSS, cancer-specific survival; NLR, neutrophil-to lymphocyte ratio; PDAC, pancreatic ductal adenocarcinoma; TILs, tumor-infiltrating lymphocytes

535_2022_1939_MOESM4_ESM.tif

Supplementary Fig. 4 In situ hybridization for IFN-gamma and IL-2, and apoptosis assay. a Typical images of IFN-gamma and IL-2 expression in TLS- present and absent patients. IFN-gamma-positive cells and IL-2-positive cells show distinct red staining. b The expression levels of IFN-gamma or IL-2 per cm2 in TLS-present patients (n=5) and TLS-absent patients (n=5) were counted. Mean ± SD (bars). *, P < 0.05. c Double immunohistochemistry-in situ hybridization. To distinguish between IHC of CD4+ and CD8+ T cells (brown) and ISH of IFN-gamma (pink) more clearly, brown is changed to green using color replacement tools of Photoshop. IFN-gamma mRNA (pink arrowhead) was localized to CD4+ and CD8+ T cells (green). d TUNEL assays were performed 7 days after the addition of IFN-gamma (100 units/mL), IL-2 (500 units/ml), 5-FU (0.05 mg/mL), and their combination to detect the apoptotic cells. TUNEL analysis showed a significant difference in cell numbers at day 7 between the combination therapy group and other treatment groups. Mean ± SD (bars). **, P < 0.001, ***, P < 0.01. Original magnification (a, c, 400×). Scale bars, 20 μm. IFN, interferon; IL, interleukin; PDAC, pancreatic ductal adenocarcinoma; SD, standard deviation; TLS, tertiary lymphoid structure

Supplementary file5 (DOCX 59 KB)

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Tanaka, T., Masuda, A., Inoue, J. et al. Integrated analysis of tertiary lymphoid structures in relation to tumor-infiltrating lymphocytes and patient survival in pancreatic ductal adenocarcinoma. J Gastroenterol 58, 277–291 (2023). https://doi.org/10.1007/s00535-022-01939-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00535-022-01939-8

Keywords

Navigation