Skip to main content

Advertisement

Log in

Diagnostic and prognostic value of tumor-infiltrating B cells in lymph node metastases of papillary thyroid carcinoma

  • Original Article
  • Published:
Virchows Archiv Aims and scope Submit manuscript

Abstract

Lymph node metastases are strongly associated with unfavorable prognosis in papillary thyroid carcinoma (PTC) patients. However, there are few sensitive or specific indicators that can diagnose or predict lymph node metastases in PTC. The objective of our study was to identify reliable indicators for the diagnosis and prediction of lymph node metastases of PTC. The PTC data set was obtained from The Cancer Genome Atlas (TCGA) cohort. Information on tumor-infiltrating immune cells in PTC was acquired using single-sample gene set enrichment analysis (ssGSEA). Then, the progression-free survival (PFS) rates of PTC patients were evaluated by Kaplan–Meier curves. A tissue microarray including 58 normal thyroid tissues and 57 PTC tissues was processed for CD19 immunohistochemistry staining. Finally, evaluation of phenotype permutations was performed using gene set enrichment analysis (GSEA). There was an appreciable association between immune infiltration and lymph node metastases in PTC. Among those immune cells, B cells and cytotoxic cells showed significant predictive accuracy for lymph node metastases in PTC. Tumor-infiltrating B cells and NK cells were associated with favorable prognosis, while tumor-associated NK CD56bright cells were correlated with poor prognosis in PTC patients. IHC analyses of PTC further confirmed a notably negative correlation between B cell infiltration and lymph node metastases in PTC. Additionally, mutations in BRAF, a dominant cause of tumor mutation burden (TMB), were positively correlated with reduced B cell infiltration and lymph node metastases in PTC. GSEA revealed that epithelial-mesenchymal transition, IL-6/JAK/STAT3 signaling, the inflammatory response, and TNF-α signaling via the NFκB pathway were remarkably suppressed pathways in patients with BRAF mutations. Tumor-associated lymphocytic infiltration, especially B cell infiltration, provides diagnostic and prognostic value for lymph node metastases in PTC.

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
Fig. 5

Similar content being viewed by others

Data availability

All data generated or analyzed during this study are included in the article and its supplementary files.

Abbreviations

AUC:

Area under the ROC curve

GSEA:

Gene set enrichment analysis

HR:

Hazard ratio

IHC:

Immunohistochemistry

NK:

Natural killer

ROC:

Receiver operating characteristic

ssGSEA:

Single-sample gene set enrichment analysis

TCGA:

The Cancer Genome Atlas

PTC:

Papillary thyroid carcinoma

TMB:

Tumor mutation burden

95% CI:

95% Confidence interval

References

  1. Varricchi G et al (2019) The immune landscape of thyroid cancer in the context of immune checkpoint inhibition. Int J Mol Sci 20(16):3934

    Article  CAS  Google Scholar 

  2. Li N et al (2013) Impact of enhanced detection on the increase in thyroid cancer incidence in the United States: review of incidence trends by socioeconomic status within the Surveillance, Epidemiology, and End Results registry, 1980–2008. Thyroid 23(1):103–110

    Article  Google Scholar 

  3. Park JL et al (2020) Comprehensive DNA methylation profiling identifies novel diagnostic biomarkers for thyroid cancer.Thyroid 30(2):192–203

  4. Cabrera RN et al (2016) The role of SPECT/CT lymphoscintigraphy and radioguided sentinel lymph node biopsy in managing papillary thyroid cancer. JAMA Otolaryngol Head Neck Surg 142(9):834–841

    Article  Google Scholar 

  5. Liu X et al (2019) Regulatory T cells and M2 macrophages present diverse prognostic value in gastric cancer patients with different clinicopathologic characteristics and chemotherapy strategies. J Transl Med 17(1):192

    Article  Google Scholar 

  6. Holland BC et al (2019) Age and sex have no impact on expression levels of markers of immune cell infiltration and immune checkpoint pathways in patients with muscle-invasive urothelial carcinoma of the bladder treated with radical cystectomy. Cancer Immunol Immunother 68(6):991–997

    Article  CAS  Google Scholar 

  7. Zhou J et al (2018) Cancer-associated fibroblasts correlate with tumor-associated macrophages infiltration and lymphatic metastasis in triple negative breast cancer patients. J Cancer 9(24):4635–4641

    Article  CAS  Google Scholar 

  8. Lee HE et al (2011) High FOXP3+ regulatory T-cell density in the sentinel lymph node is associated with downstream non-sentinel lymph-node metastasis in gastric cancer. Br J Cancer 105(3):413–419

    Article  CAS  Google Scholar 

  9. Liao H et al (2018) Expression of V-domain immunoglobulin suppressor of T cell activation is associated with the advanced stage and presence of lymph node metastasis in ovarian cancer. Oncol Lett 16(3):3465–3472

    PubMed  PubMed Central  Google Scholar 

  10. Song J et al (2019) Patterns of immune infiltration in HNC and their clinical implications: a gene expression-based study. Front Oncol 9:1285

    Article  Google Scholar 

  11. Lee-Chang C et al (2019) Myeloid-derived suppressive cells promote B cell-mediated immunosuppression via transfer of PD-L1 in glioblastoma. Cancer Immunol Res 7(12):1928–1943

    Article  CAS  Google Scholar 

  12. Pinto R et al (2019) KRAS-driven lung adenocarcinoma and B cell infiltration: novel insights for immunotherapy. Cancers (Basel) 11(8):1145

  13. Bindea G et al (2013) Spatiotemporal dynamics of intratumoral immune cells reveal the immune landscape in human cancer. Immunity 39(4):782–795

    Article  CAS  Google Scholar 

  14. Chalmers ZR et al (2017) Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med 9(1):34

    Article  Google Scholar 

  15. Xing M et al (2015) Association between BRAF V600E mutation and recurrence of papillary thyroid cancer. J Clin Oncol 33(1):42–50

    Article  Google Scholar 

  16. Kim TH et al (2012) The association of the BRAF(V600E) mutation with prognostic factors and poor clinical outcome in papillary thyroid cancer: a meta-analysis. Cancer 118(7):1764–1773

    Article  CAS  Google Scholar 

  17. Ma S, Song W, Xu Y, Si X, Zhang D, Lv S, Yang C, Ma L, Tang Z, Chen X (2020) Neutralizing tumor-promoting inflammation with polypeptide-dexamethasone conjugate for microenvironment modulation and colorectal cancer therapy. Biomaterials 232:119676. https://doi.org/10.1016/j.biomaterials.2019.119676

  18. Yuan Y et al (2016) Role of the tumor microenvironment in tumor progression and the clinical applications (review). Oncol Rep 35(5):2499–2515

    Article  CAS  Google Scholar 

  19. Anfray C et al (2019) Current strategies to target tumor-associated-macrophages to improve anti-tumor immune responses. Cells 9(1):46

  20. Ferrari SM et al (2019) Immune and inflammatory cells in thyroid cancer microenvironment. Int J Mol Sci 20(18):4413

  21. Liotti F, Visciano C, Melillo RM (2012) Inflammation in thyroid oncogenesis. Am J Cancer Res 2(3):286–297

    PubMed  PubMed Central  Google Scholar 

  22. Antonelli A, Ferrari SM, Fallahi P (2018) Current and future immunotherapies for thyroid cancer. Expert Rev Anticancer Ther 18(2):149–159

    Article  CAS  Google Scholar 

  23. Muppa P et al (2019) Immune cell infiltration may be a key determinant of long-term survival in small cell lung cancer. J Thorac Oncol 14(7):1286–1295

    Article  CAS  Google Scholar 

  24. Pages F et al (2018) International validation of the consensus Immunoscore for the classification of colon cancer: a prognostic and accuracy study. Lancet 391(10135):2128–2139

    Article  Google Scholar 

  25. Yin H et al (2020) Immune microenvironment of thyroid cancer. J Cancer 11(16):4884–4896

    Article  CAS  Google Scholar 

  26. Na KJ, Choi H (2018) Immune landscape of papillary thyroid cancer and immunotherapeutic implications. Endocr Relat Cancer 25(5):523–531

    Article  CAS  Google Scholar 

  27. Schreiber RD, Old LJ, Smyth MJ (2011) Cancer immunoediting: integrating immunitys roles in cancer suppression and promotion. Science 331(6024):1565–1570

    Article  CAS  Google Scholar 

  28. Qian G et al (2020) Thyroid cancer metastasis is associated with an overabundance of defective follicular helper T cells. APMIS 128(8):487–496

    Article  CAS  Google Scholar 

  29. Harrell MI, Iritani BM, Ruddell A (2007) Tumor-induced sentinel lymph node lymphangiogenesis and increased lymph flow precede melanoma metastasis. Am J Pathol 170(2):774–786

    Article  Google Scholar 

  30. Miligy I et al (2017) Prognostic significance of tumour infiltrating B lymphocytes in breast ductal carcinoma in situ. Histopathology 71(2):258–268

    Article  Google Scholar 

  31. Schmidt M et al (2008) The humoral immune system has a key prognostic impact in node-negative breast cancer. Cancer Res 68(13):5405–5413

    Article  CAS  Google Scholar 

  32. West NR et al (2011) Tumor-infiltrating lymphocytes predict response to anthracycline-based chemotherapy in estrogen receptor-negative breast cancer. Breast Cancer Res 13(6):R126

    Article  CAS  Google Scholar 

  33. Rody A et al (2009) T-cell metagene predicts a favorable prognosis in estrogen receptor-negative and HER2-positive breast cancers. Breast Cancer Res 11(2):R15

    Article  Google Scholar 

  34. Eiro N et al (2012) Impact of CD68/(CD3+CD20) ratio at the invasive front of primary tumors on distant metastasis development in breast cancer. PLoS One 7(12):e52796

    Article  CAS  Google Scholar 

  35. Gu Y et al (2019) Tumor-educated B cells selectively promote breast cancer lymph node metastasis by HSPA4-targeting IgG. Nat Med 25(2):312–322

    Article  CAS  Google Scholar 

  36. Zhang Z et al (2017) Yin-yang effect of tumor infiltrating B cells in breast cancer: from mechanism to immunotherapy. Cancer Lett 393:1–7

    Article  CAS  Google Scholar 

  37. Wang SS et al (2019) Tumor-infiltrating B cells: their role and application in anti-tumor immunity in lung cancer. Cell Mol Immunol 16(1):6–18

    Article  CAS  Google Scholar 

  38. Lee-Chang C et al (2013) Inhibition of breast cancer metastasis by resveratrol-mediated inactivation of tumor-evoked regulatory B cells. J Immunol 191(8):4141–4151

    Article  CAS  Google Scholar 

  39. Aghajani MJ et al (2018) Predictive relevance of programmed cell death protein 1 and tumor-infiltrating lymphocyte expression in papillary thyroid cancer. Surgery 163(1):130–136

    Article  Google Scholar 

  40. Li Y et al (2015) Transforming growth factor beta1 could influence thyroid nodule elasticity and also improve cervical lymph node metastasis in papillary thyroid carcinoma. Ultrasound Med Biol 41(11):2866–2872

    Article  Google Scholar 

  41. Wang N et al (2014) Expression of TGF-beta1, SNAI1 and MMP-9 is associated with lymph node metastasis in papillary thyroid carcinoma. J Mol Histol 45(4):391–399

    Article  CAS  Google Scholar 

  42. Howell GM et al (2013) BRAF V600E mutation independently predicts central compartment lymph node metastasis in patients with papillary thyroid cancer. Ann Surg Oncol 20(1):47–52

    Article  Google Scholar 

  43. Ylli D et al (2019) Microfluidic droplet digital PCR is a powerful tool for detection of BRAF and TERT mutations in papillary thyroid carcinomas. Cancers (Basel) 11(12):1916

  44. Angell TE et al (2014) BRAF V600E in papillary thyroid carcinoma is associated with increased programmed death ligand 1 expression and suppressive immune cell infiltration. Thyroid 24(9):1385–1393

    Article  CAS  Google Scholar 

  45. Huang M, Xin W (2018) Matrine inhibiting pancreatic cells epithelial-mesenchymal transition and invasion through ROS/NF-kappaB/MMPs pathway. Life Sci 192:55–61

    Article  CAS  Google Scholar 

  46. Yoo SK et al (2019) Integrative analysis of genomic and transcriptomic characteristics associated with progression of aggressive thyroid cancer. Nat Commun 10(1):2764

    Article  Google Scholar 

  47. Wang J et al (2019) Natural naphthohydroquinone dimer rubioncolin C exerts anti-tumor activity by inducing apoptotic and autophagic cell death and inhibiting the NF-kappaB and Akt/mTOR/P70S6K pathway in human cancer cells. Cells 8(12):1593

Download references

Acknowledgements

Special thanks to Shanghai Outdo Biotech Co., Ltd., for helping us create the tissue microarray. Thank you to Dr. Ya Xiao for guidance in data analysis.

Funding

This work was supported by grants from the National Natural Science Foundation of China (grant no. 81602730) and Medical Technology Innovation Fund of Chongqing General Hospital (Y2020ZDXM06).

Author information

Authors and Affiliations

Authors

Contributions

Designed the research: ZY, FZ, and WY; acquisition of data: ZY; analysis and interpretation of data: ZY and WY; collection of human PTC and normal thyroid tissue microarray: LY, YZ, YL, HC, and SY; draft of the manuscript: WY and ZY; critical revision of the manuscript: FZ and WY; funding acquisition: LY and ZY. All authors read, revised, and approved the final manuscript.

Corresponding authors

Correspondence to Fan Zhang or Wang Yang.

Ethics declarations

Ethics approval and consent to participate

Ethics approval for this study was granted by the Institute Research Medical Ethics Committee of the Shanghai Outdo Biotech Company. Written informed consent was obtained from all patients.

Consent for publication

Informed consent for publication was obtained from all authors.

Competing interests

The authors declare no competing interests.

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.

Additional File 1

Supplementary Figure 1. Coexpression patterns among diverse immune cells. (PDF 48 kb)

Additional File 2

Supplementary Figure 2. Coexpression patterns between B cell infiltration and immunosuppressive markers. (PDF 8 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yang, Z., Yin, L., Zeng, Y. et al. Diagnostic and prognostic value of tumor-infiltrating B cells in lymph node metastases of papillary thyroid carcinoma. Virchows Arch 479, 947–959 (2021). https://doi.org/10.1007/s00428-021-03137-y

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00428-021-03137-y

Keywords

Navigation