Endocrinol Metab.  2020 Jun;35(2):443-455. 10.3803/EnM.2020.35.2.443.

Mechanisms of the Impact of Hashimoto Thyroiditis on Papillary Thyroid Carcinoma Progression: Relationship with the Tumor Immune Microenvironment

Affiliations
  • 1Laboratory of Pathology CSD Health Care
  • 2Ukrainian Research and Practical Center for Endocrine Surgery
  • 3Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv
  • 4Department of Endocrinology, Bogomolets National Medical University, Kyiv, Ukraine

Abstract

Background
The relationship between Hashimoto thyroiditis (HT) and papillary thyroid carcinoma (PTC) remains uncertain. We assessed the impact of HT on the tumor immune microenvironment (TIME) in PTC.
Methods
Thirty patients with PTC (group 1) and 30 patients with PTC and HT (group 2) were enrolled in this pilot study. The distribution and number of CD8+ lymphocytes, plasma cells (CD138+), regulatory T cells (forkhead box P3 [FOXP3+)], mast cell tryptase (MCT+), and M2 macrophages (CD163+) were evaluated. To test the hypothesis that HT impacts PTC development via signal transducer and activator of transcription 6 (STAT6) activation and M2 macrophage polarization, we investigated STAT6 expression in tumor and stromal cells. We also evaluated vascular endothelial growth factor (VEGF) expression by lymph node metastasis (LNM) status.
Results
TIME showed significant between-group differences. Group 1 patients demonstrated immune desert or immune-excluded immunophenotypes, while an inflamed phenotype with more CD8+ cells (P<0.001) predominated in group 2. Immune-excluded TIME was associated with the highest LNM rate. In PTC, LNM was associated with more numerous CD163+ cells. Moreover, LNM in group 1 was associated with increased numbers of mast cells peritumorally and FOXP3+ cells intratumorally and peritumorally. Group 2 demonstrated higher STAT6 but not higher VEGF expression in tumor cells. High VEGF expression was associated with LNM regardless of HT status.
Conclusion
Concomitant HT impacted PTC signaling via STAT6 and TIME by increasing the number of CD8+ cells. LNM is associated with increases in CD163+ cells and VEGF expression in PTC, whereas HT affected LNM through different mechanisms.

Keyword

Thyroid cancer, papillary; Hashimoto disease; Metastasis; T-lymphocytes; Macrophages

Figure

  • Fig. 1 Differences in the number of cytotoxic T lymphocytes in patients of group 1 (A) and group 2 (B, C). (B) Zone of infiltration of intact thyroid tissue in a patient with concomitant Hashimoto thyroiditis. (C) Intratumoral area. Immunohistochemistry using antibodies to CD8 (×200).

  • Fig. 2 An increased number of M2 macrophages was independently associated with lymph node metastasis (LNM) development in patients with concomitant Hashimoto thyroiditis. (A) A patient in group 1 without LNM demonstrated a low number of M2 macrophages (×200). (B) A patient in group 2 without LNM (×200). (C) A high number of M2 macrophages in papillary thyroid carcinoma with LNM (×100). (D) Numerous M2 macrophages around metastases of thyroid cancer in the lymph node (×100). Immunohistochemistry using antibodies to CD163 (×200).

  • Fig. 3 The number of immune cells in and around papillary thyroid carcinoma (PTC) in patients with and without concomitant Hashimoto thyroiditis (HT) by lymph node metastasis (LNM) status. (A, B) Number of M2 macrophages (CD163 positive cells) within tumor and in peritumor stroma respectively. (C, D) Count of mast cells (mast cell tryptase [MCT] positive cells) within tumor and in peritumor stroma respectively. (E, F) Number of T-cytotoxic lymphocytes (CD8 positive cells) within tumor and in peritumor stroma respectively. (G, H) Count of plasma cells (CD138 positive cells) within tumor and in peritumor stroma respectively. (I, J) Number of T-regulatory cells (forkhead box P3 [FOXP3] positive cells) within tumor and in peritumor stroma respectively. aStatistically significant difference between patients with and without LNM at P<0.05.

  • Fig. 4 Signal transducer and activator of transcription 6 (STAT6) and vascular endothelial growth factor (VEGF) expression in patients with papillary thyroid carcinoma (PTC) according to Hashimoto thyroiditis (HT) and lymph node metastasis (LNM). (A) STAT6 expression in patients of group 2 in the intact thyroid (×200). (B) STAT6 expression in follicular cells in areas of lymphocyte infiltration associated with histoarchitectural and cytological atypia in group 2 (×200). (C) STAT6 expression in group 2 patients (×100). (D) Low VEGF expression in group 2 patients without LNM. (E) High expression of VEGF in PTC tumor cells associated with LNM. Immunohistochemistry (×200). (F, G) Relationship between STAT6 and VEGF expression in the tumor cells of patients with PTC according to HT (F) and LNM (G) status. IHC, immunohistochemical. aStatistically significant difference in the total score of biomarker expression regarding HT or LNM status at P<0.05.


Reference

1. Zhu F, Shen YB, Li FQ, Fang Y, Hu L, Wu YJ. The effects of Hashimoto thyroiditis on lymph node metastases in unifocal and multifocal papillary thyroid carcinoma: a retrospective Chinese cohort study. Medicine (Baltimore). 2016; 95:e2674.
2. Tronko M, Mabuchi K, Bogdanova T, Hatch M, Likhtarev I, Bouville A, et al. Thyroid cancer in Ukraine after the Chernobyl accident (in the framework of the Ukraine-US Thyroid Project). J Radiol Prot. 2012; 32:N65–9.
Article
3. Lee KH, Seok EY, Kim EY, Yun JS, Park YL, Park CH. Different prognostic values of individual hematologic parameters in papillary thyroid cancer due to age-related changes in immunity. Ann Surg Treat Res. 2019; 96:70–7.
Article
4. Sulaieva O, Chernenko O, Chereshneva Y, Tsomartova D, Larin O. Thyroid stimulating hormone levels and BRAFV600E mutation contribute to pathophysiology of papillary thyroid carcinoma: relation to outcomes? Pathophysiology. 2019; 26:129–35.
Article
5. Resende de Paiva C, Gronhoj C, Feldt-Rasmussen U, von Buchwald C. Association between Hashimoto’s thyroiditis and thyroid cancer in 64,628 patients. Front Oncol. 2017; 7:53.
Article
6. Konturek A, Barczynski M, Wierzchowski W, Stopa M, Nowak W. Coexistence of papillary thyroid cancer with Hashimoto thyroiditis. Langenbecks Arch Surg. 2013; 398:389–94.
Article
7. Fang W, Ye L, Shen L, Cai J, Huang F, Wei Q, et al. Tumor-associated macrophages promote the metastatic potential of thyroid papillary cancer by releasing CXCL8. Carcinogenesis. 2014; 35:1780–7.
Article
8. Borowczyk M, Janicki A, Dworacki G, Szczepanek-Parulska E, Danieluk M, Barnett J, et al. Decreased staging of differentiated thyroid cancer in patients with chronic lymphocytic thyroiditis. J Endocrinol Invest. 2019; 42:45–52.
Article
9. Marotta V, Sciammarella C, Chiofalo MG, Gambardella C, Bellevicine C, Grasso M, et al. Hashimoto’s thyroiditis predicts outcome in intrathyroidal papillary thyroid cancer. Endocr Relat Cancer. 2017; 24:485–93.
Article
10. Huang BY, Hseuh C, Chao TC, Lin KJ, Lin JD. Well-differentiated thyroid carcinoma with concomitant Hashimoto’s thyroiditis present with less aggressive clinical stage and low recurrence. Endocr Pathol. 2011; 22:144–9.
Article
11. Means C, Clayburgh DR, Maloney L, Sauer D, Taylor MH, Shindo ML, et al. Tumor immune microenvironment characteristics of papillary thyroid carcinoma are associated with histopathological aggressiveness and BRAF mutation status. Head Neck. 2019; 41:2636–46.
Article
12. Binnewies M, Roberts EW, Kersten K, Chan V, Fearon DF, Merad M, et al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat Med. 2018; 24:541–50.
Article
13. Caruana I, Simula L, Locatelli F, Campello S. T lymphocytes against solid malignancies: winning ways to defeat tumours. Cell Stress. 2018; 2:200–12.
Article
14. Can NY, Ayturk S, Celik M, Sezer YA, Ozyilmaz F, Tastekin E, et al. Histological perspective on the effects of tumor-associated macrophages in the tumor microenvironment surrounding papillary thyroid carcinoma. Pol J Pathol. 2016; 67:332–44.
Article
15. Binnemars-Postma K, Bansal R, Storm G, Prakash J. Targeting the Stat6 pathway in tumor-associated macrophages reduces tumor growth and metastatic niche formation in breast cancer. FASEB J. 2018; 32:969–78.
16. Hebenstreit D, Wirnsberger G, Horejs-Hoeck J, Duschl A. Signaling mechanisms, interaction partners, and target genes of STAT6. Cytokine Growth Factor Rev. 2006; 17:173–88.
Article
17. Yu T, Gan S, Zhu Q, Dai D, Li N, Wang H, et al. Modulation of M2 macrophage polarization by the crosstalk between Stat6 and Trim24. Nat Commun. 2019; 10:4353.
Article
18. Czimmerer Z, Daniel B, Horvath A, Ruckerl D, Nagy G, Kiss M, et al. The transcription factor STAT6 mediates direct repression of inflammatory enhancers and limits activation of alternatively polarized macrophages. Immunity. 2018; 48:75–90.
Article
19. de Araujo-Filho VJ, Alves VA, de Castro IV, Lourenco SV, Cernea CR, Brandao LG, et al. Vascular endothelial growth factor expression in invasive papillary thyroid carcinoma. Thyroid. 2009; 19:1233–7.
Article
20. Chen DS, Mellman I. Oncology meets immunology: the cancer-immunity cycle. Immunity. 2013; 39:1–10.
Article
21. Willimsky G, Blankenstein T. Sporadic immunogenic tumours avoid destruction by inducing T-cell tolerance. Nature. 2005; 437:141–6.
Article
22. Gonzalez-Amaro R, Marazuela M. T regulatory (Treg) and T helper 17 (Th17) lymphocytes in thyroid autoimmunity. Endocrine. 2016; 52:30–8.
Article
23. Safdari V, Alijani E, Nemati M, Jafarzadeh A. Imbalances in T cell-related transcription factors among patients with Hashimoto’s thyroiditis. Sultan Qaboos Univ Med J. 2017; 17:e174–80.
Article
24. Antonelli A, Ferrari SM, Corrado A, Di Domenicantonio A, Fallahi P. Autoimmune thyroid disorders. Autoimmun Rev. 2015; 14:174–80.
Article
25. Galdiero MR, Varricchi G, Marone G. The immune network in thyroid cancer. Oncoimmunology. 2016; 5:e1168556.
Article
26. Horton BL, Fessenden TB, Spranger S. Tissue site and the cancer immunity cycle. Trends Cancer. 2019; 5:593–603.
Article
27. Bindea G, Mlecnik B, Tosolini M, Kirilovsky A, Waldner M, Obenauf AC, et al. Spatiotemporal dynamics of intratumoral immune cells reveal the immune landscape in human cancer. Immunity. 2013; 39:782–95.
Article
28. Xiong H, Mittman S, Rodriguez R, Pacheco-Sanchez P, Moskalenko M, Yang Y, et al. Coexpression of inhibitory receptors enriches for activated and functional CD8+ T cells in murine syngeneic tumor models. Cancer Immunol Res. 2019; 7:963–76.
29. Spranger S. Mechanisms of tumor escape in the context of the T-cell-inflamed and the non-T-cell-inflamed tumor microenvironment. Int Immunol. 2016; 28:383–91.
Article
30. Togashi Y, Shitara K, Nishikawa H. Regulatory T cells in cancer immunosuppression: implications for anticancer therapy. Nat Rev Clin Oncol. 2019; 16:356–71.
31. Liotti F, Prevete N, Vecchio G, Melillo RM. Recent advances in understanding immune phenotypes of thyroid carcinomas: prognostication and emerging therapies. F1000Res. 2019; 8:F1000 Faculty Rev. 227.
Article
32. Condeelis J, Pollard JW. Macrophages: obligate partners for tumor cell migration, invasion, and metastasis. Cell. 2006; 124:263–6.
Article
33. Ferrari SM, Fallahi P, Galdiero MR, Ruffilli I, Elia G, Ragusa F, et al. Immune and inflammatory cells in thyroid cancer microenvironment. Int J Mol Sci. 2019; 20:4413.
Article
34. Quaranta V, Schmid MC. Macrophage-mediated subversion of anti-tumour immunity. Cells. 2019; 8:747.
Article
35. Loke P, Allison JP. PD-L1 and PD-L2 are differentially regulated by Th1 and Th2 cells. Proc Natl Acad Sci U S A. 2003; 100:5336–41.
Article
36. Melillo RM, Guarino V, Avilla E, Galdiero MR, Liotti F, Prevete N, et al. Mast cells have a protumorigenic role in human thyroid cancer. Oncogene. 2010; 29:6203–15.
Article
37. Movahedi K, Laoui D, Gysemans C, Baeten M, Stange G, Van den Bossche J, et al. Different tumor microenvironments contain functionally distinct subsets of macrophages derived from Ly6C(high) monocytes. Cancer Res. 2010; 70:5728–39.
Article
38. Gordon S, Martinez FO. Alternative activation of macrophages: mechanism and functions. Immunity. 2010; 32:593–604.
Article
39. Zhang Y, Zhang Y, Gu W, Sun B. TH1/TH2 cell differentiation and molecular signals. Adv Exp Med Biol. 2014; 841:15–44.
Article
Full Text Links
  • ENM
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr