Clin Exp Otorhinolaryngol.  2022 May;15(2):183-193. 10.21053/ceo.2021.02215.

Transcriptomic Analysis of Papillary Thyroid Cancer: A Focus on Immune-Subtyping, Oncogenic Fusion, and Recurrence

Affiliations
  • 1Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea
  • 2Department of Bioscience, University of Science and Technology, Daejeon, Korea
  • 3Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
  • 4Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
  • 5Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University College of Medicine, Seoul, Korea
  • 6Department of Pathology, Chungnam National University College of Medicine, Daejeon, Korea
  • 7Department of Surgery, Chungnam National University College of Medicine, Daejeon, Korea
  • 8Department of Otolaryngology-Head and Neck Surgery, Chungnam National University College of Medicine, Daejeon, Korea
  • 9Korean Bioinformation Center (KOBIC), Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea

Abstract


Objectives
. Thyroid cancer is the most common endocrine tumor, with rapidly increasing incidence worldwide. However, its transcriptomic characteristics associated with immunological signatures, driver fusions, and recurrence markers remain unclear. We aimed to investigate the transcriptomic characteristics of advanced papillary thyroid cancer.
Methods
. This study included 282 papillary thyroid cancer tumor samples and 155 normal samples from Chungnam National University Hospital and Seoul National University Hospital. Transcriptomic quantification was determined by high-throughput RNA sequencing. We investigated the associations of clinical parameters and molecular signatures using RNA sequencing. We validated predictive biomarkers using the Cancer Genome Atlas database.
Results
. Through a comparison of differentially expressed genes, gene sets, and pathways in papillary thyroid cancer compared to normal tumor-adjacent tissue, we found increased immune signaling associated with cytokines or T cells and decreased thyroid hormone synthetic pathways. In addition, patients with recurrence presented increased CD8+ T-cell and Th1-cell signatures. Interestingly, we found differentially overexpressed genes related to immune-escape signaling such as CTLA4, IDO1, LAG3, and PDCD1 in advanced papillary thyroid cancer with a low thyroid differentiation score. Fusion analysis showed that the PI3K and mitogen-activated protein kinase (MAPK) signaling pathways were regulated differently according to the RET fusion partner genes (CCDC6 or NCOA4). Finally, we identified HOXD9 as a novel molecular biomarker that predicts the recurrence of thyroid cancer in addition to known risk factors (tumor size, lymph node metastasis, and extrathyroidal extension).
Conclusion
. We identified a high association with immune-escape signaling in the immune-hot group with aggressive clinical characteristics among Korean thyroid cancer patients. Moreover, RET fusion differentially regulated PI3K and MAPK signaling depending on the partner gene of RET, and HOXD9 was found to be a recurrence marker for advanced papillary thyroid cancer.

Keyword

Thyroid Cancer; Korean Thyroid Cancer; Advanced Papillary Thyroid Cancer; RNA Sequencing; Immune Subtyping; Immune-Escape Signaling; Fusion Outlier; Predictive Biomarker

Figure

  • Fig. 1. Transcriptomic overview and immuno-clinical associations in Korean papillary thyroid cancer patients. (A) Volcano plots showing differentially expressed genes across groups. Red and gray dots represent significance (Fisher exact test; P<0.01 and fold change >1.5) and nonsignificance, respectively. (B) Results of gene-set enrichment analysis using the KEGG and Hallmark database from Enrichr. Light red and light blue indicate upregulated and downregulated terms in tumors compared to normal tissues. (C) Increased immune-related signaling was exclusively observed when lymph node metastasis (LNM) and recurrence occurred. Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and tumor necrosis factor-alpha (TNF-α) were only enriched when LNM occurred and CD8+ T cells and Th1 cells were only enriched when recurrence occurred. EMT, epithelial-mesenchymal transition; UV, ultraviolet.

  • Fig. 2. Association of immune signatures with the thyroid differentiation score (TDS) in advanced papillary thyroid cancer (PTC). (A) The immune landscape of PTC using the immune signature of xCell. The immune subtype based on the immune score was represented as immunehot (n=35), immune-intermediate (n=107), and immune-cold (n=140). (B) The TDS across three immune subtypes. (C) Heatmap sorted by TDS and expression of the top 20 genes that showed high correlations with the TDS. (D) Boxplots showing the major immune-checkpoint inhibitors, including CTLA4, IDO1, LAG3, and PDCD1. (E) Hub gene discovery through network module analysis for each immune subtype. ECI, extracapsular invasion; ETE, extrathyroidal extension; C_LNM, central lymph node metastasis; L_LNM, lateral lymph node metastasis; LVI, lymphovascular invasion; EMT, epithelial-mesenchymal transition.

  • Fig. 3. Different regulation patterns of the PI3K and mitogen-activated protein kinase (MAPK) signaling pathways according to the partner genes of RET fusion. (A) Bar plot showing the fusion count across papillary thyroid cancer (PTC) tumors. (B) Outlier analysis of RET fusion. Blue and red dots indicate the expression of the RET gene in samples without and with fusions, respectively. (C) Impacts of the two partner genes of RET fusion on the MAPK and PI3K pathways. “Common” indicates overlapping genes in both pathways. Circles and squares indicate cis- and trans-acting genes, but there is no cis-interaction, and their sizes indicate the significance of the P-value. (D, E) The genes regulated by RET fusion of CCDC6 and NCOA4 may bridge the MAPK signaling pathway. (D) PTC in the Korean Thyroid Cancer (KTC) cohort. (E) The Thyroid Carcinoma cohort from the Cancer Genome Atlas (TCGA-THCA). (F) Two ways that RET fusion regulates MAPK signaling with different partner genes. WT, wildtype.

  • Fig. 4. HOXD9 is a candidate gene associated with the recurrence of papillary thyroid cancer (PTC). (A) Discovery of recurrence-related factors in PTC samples. The Fisher exact test was performed with a threshold of P<0.01. (B) Barplots showing the spectrum of samples in the PTC group for well-known risk factors such as tumor size, extrathyroidal extension (ETE), and lateral lymph node metastasis (L_LNM). (C) Workflow for selecting HOXD9. (D) Kaplan-Meier plot of thyroid cancer patients based on the expression of HODX9 (high or low based on the median value from the GEPIA database. (E) Barplots showing the recurrence rate based on the expression of HOXD9. (F) Enriched signaling pathways in the HOXD9-high group. (G) Correlation of gene expression between HOXD9 and transcription factors such as transcription factor 3 (TCF3) and enhancer of zeste 2 polycomb repressive complex 2 subunit (EZH2) that regulate HOXD9 in the Korean Thyroid Cancer (KTC) cohort and the Thyroid Carcinoma cohort from the Cancer Genome Atlas (TCGA-THCA cohort). (H) Summary of the two routes to recurrence from Korean PTC samples. ECI, extracapsular invasion; C_LNM, central lymph node metastasis; L_LVI, lateral-lymphovascular invasion; NS, not significant; RF, risk factor; DFS, disease free survival; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; TNF-α, tumor necrosis factoralpha; TF, transcription factor.


Cited by  1 articles

Mitochondrial Ribosomal Protein L14 Promotes Cell Growth and Invasion by Modulating Reactive Oxygen Species in Thyroid Cancer
Hae Jong Kim, Quoc Khanh Nguyen, Seung-Nam Jung, Mi Ae Lim, Chan Oh, Yudan Piao, YanLi Jin, Ju-Hui Kim, Young Il Kim, Yea Eun Kang, Jae Won Chang, Ho-Ryun Won, Bon Seok Koo
Clin Exp Otorhinolaryngol. 2023;16(2):184-197.    doi: 10.21053/ceo.2022.01760.


Reference

1. Ahn HS, Kim HJ, Welch HG. Korea’s thyroid-cancer “epidemic”: screening and overdiagnosis. N Engl J Med. 2014; Nov. 371(19):1765–7.
Article
2. Morris LG, Sikora AG, Tosteson TD, Davies L. The increasing incidence of thyroid cancer: the influence of access to care. Thyroid. 2013; Jul. 23(7):885–91.
Article
3. Yeon EK, Sohn YM, Seo M, Kim EJ, Eun YG, Park WS, et al. Diagnostic performance of a combination of shear wave elastography and B-mode ultrasonography in differentiating benign from malignant thyroid nodules. Clin Exp Otorhinolaryngol. 2020; May. 13(2):186–93.
Article
4. Giani F, Vella V, Tumino D, Malandrino P, Frasca F. The possible role of cancer stem cells in the resistance to kinase inhibitors of advanced thyroid cancer. Cancers (Basel). 2020; Aug. 12(8):2249.
Article
5. Haas BJ, Dobin A, Li B, Stransky N, Pochet N, Regev A. Accuracy assessment of fusion transcript detection via read-mapping and de novo fusion transcript assembly-based methods. Genome Biol. 2019; Oct. 20(1):213.
Article
6. Aran D, Hu Z, Butte AJ. xCell: digitally portraying the tissue cellular heterogeneity landscape. Genome Biol. 2017; Nov. 18(1):220.
Article
7. Dobin A, Gingeras TR. Optimizing RNA-Seq mapping with STAR. Methods Mol Biol. 2016; 1415:245–62.
Article
8. Ahn SH, Kim WS. The effect of prophylactic central neck dissection during hemithyroidectomy on locoregional recurrence in patients with papillary thyroid carcinoma: a meta-analysis. Clin Exp Otorhinolaryngol. 2020; May. 13(2):194–202.
Article
9. Cancer Genome Atlas Research Network. Integrated genomic characterization of papillary thyroid carcinoma. Cell. 2014; Oct. 159(3):676–90.
10. Yoo SK, Lee S, Kim SJ, Jee HG, Kim BA, Cho H, et al. Comprehensive analysis of the transcriptional and mutational landscape of follicular and papillary thyroid cancers. PLoS Genet. 2016; Aug. 12(8):e1006239.
Article
11. Yoo SK, Song YS, Lee EK, Hwang J, Kim HH, Jung G, et al. Integrative analysis of genomic and transcriptomic characteristics associated with progression of aggressive thyroid cancer. Nat Commun. 2019; Jun. 10(1):2764.
Article
12. Salvatore D, Santoro M, Schlumberger M. The importance of the RET gene in thyroid cancer and therapeutic implications. Nat Rev Endocrinol. 2021; May. 17(5):296–306.
Article
13. Wirth LJ, Sherman E, Robinson B, Solomon B, Kang H, Lorch J, et al. Efficacy of selpercatinib in RET-altered thyroid cancers. N Engl J Med. 2020; Aug. 383(9):825–35.
Article
14. Santoro M, Carlomagno F. Central role of RET in thyroid cancer. Cold Spring Harb Perspect Biol. 2013; Dec. 5(12):a009233.
Article
15. Kim HJ, Park HK, Byun DW, Suh K, Yoo MH, Min YK, et al. Iodine intake as a risk factor for BRAF mutations in papillary thyroid cancer patients from an iodine-replete area. Eur J Nutr. 2018; Mar. 57(2):809–15.
Article
16. Kim WB, Jeon MJ, Kim WG, Kim TY, Shong YK. Unmet clinical needs in the treatment of patients with thyroid cancer. Endocrinol Metab (Seoul). 2020; Mar. 35(1):14–25.
Article
17. Kim HJ. Correlation of BRAF mutation of papillary thyroid carcinoma and urine iodine status in Korean population. Seoul: Yonsei University Health System;2021.
18. Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013; Jan. 29(1):15–21.
Article
19. Trapnell C, Roberts A, Goff L, Pertea G, Kim D, Kelley DR, et al. Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks. Nat Protoc. 2012; Mar. 7(3):562–78.
Article
20. Liberzon A, Birger C, Thorvaldsdottir H, Ghandi M, Mesirov JP, Tamayo P. The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Syst. 2015; Dec. 1(6):417–25.
21. Hanzelmann S, Castelo R, Guinney J. GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinformatics. 2013; Jan. 14:7.
Article
22. Kuleshov MV, Jones MR, Rouillard AD, Fernandez NF, Duan Q, Wang Z, et al. Enrichr: a comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res. 2016; Jul. 44(W1):W90–7.
Article
23. Russo PS, Ferreira GR, Cardozo LE, Burger MC, Arias-Carrasco R, Maruyama SR, et al. CEMiTool: a Bioconductor package for performing comprehensive modular co-expression analyses. BMC Bioinformatics. 2018; Feb. 19(1):56.
Article
24. Wang X, Peng W, Li C, Qin R, Zhong Z, Sun C. Identification of an immune-related signature indicating the dedifferentiation of thyroid cells. Cancer Cell Int. 2021; Apr. 21(1):231.
Article
25. Cunha LL, Domingues GA, Morari EC, Soares FA, Vassallo J, Ward LS. The immune landscape of the microenvironment of thyroid cancer is closely related to differentiation status. Cancer Cell Int. 2021; Jul. 21(1):387.
Article
26. de Groot JW, Links TP, Plukker JT, Lips CJ, Hofstra RM. RET as a diagnostic and therapeutic target in sporadic and hereditary endocrine tumors. Endocr Rev. 2006; Aug. 27(5):535–60.
27. Li AY, McCusker MG, Russo A, Scilla KA, Gittens A, Arensmeyer K, et al. RET fusions in solid tumors. Cancer Treat Rev. 2019; Dec. 81:101911.
Article
28. Paratala BS, Chung JH, Williams CB, Yilmazel B, Petrosky W, Williams K, et al. RET rearrangements are actionable alterations in breast cancer. Nat Commun. 2018; Nov. 9(1):4821.
Article
29. Santoro M, Moccia M, Federico G, Carlomagno F. RET gene fusions in malignancies of the thyroid and other tissues. Genes (Basel). 2020; Apr. 11(4):424.
Article
30. Park JL, Kim SK, Jeon S, Jung CK, Kim YS. MicroRNA profile for diagnostic and prognostic biomarkers in thyroid cancer. Cancers (Basel). 2021; Feb. 13(4):632.
Article
31. Jo K, Lim DJ. Clinical implications of anti-thyroglobulin antibody measurement before surgery in thyroid cancer. Korean J Intern Med. 2018; Nov. 33(6):1050–7.
Article
32. Randolph GW, Duh QY, Heller KS, LiVolsi VA, Mandel SJ, Steward DL, et al. The prognostic significance of nodal metastases from papillary thyroid carcinoma can be stratified based on the size and number of metastatic lymph nodes, as well as the presence of extranodal extension. Thyroid. 2012; Nov. 22(11):1144–52.
Article
33. Wang O, Zheng Z, Wang Q, Jin Y, Jin W, Wang Y, et al. ZCCHC12, a novel oncogene in papillary thyroid cancer. J Cancer Res Clin Oncol. 2017; Sep. 143(9):1679–86.
Article
34. Ho WJ, Croessmann S, Lin J, Phyo ZH, Charmsaz S, Danilova L, et al. Systemic inhibition of PTPN22 augments anticancer immunity. J Clin Invest. 2021; Jul. 131(17):e146950.
Article
35. Cubas R, Khan Z, Gong Q, Moskalenko M, Xiong H, Ou Q, et al. Autoimmunity linked protein phosphatase PTPN22 as a target for cancer immunotherapy. J Immunother Cancer. 2020; Oct. 8(2):e001439.
Article
36. Ross MD, Bruggeman LA, Hanss B, Sunamoto M, Marras D, Klotman ME, et al. Podocan, a novel small leucine-rich repeat protein expressed in the sclerotic glomerular lesion of experimental HIV-associated nephropathy. J Biol Chem. 2003; Aug. 278(35):33248–55.
Article
37. Duval S, Abu-Thuraia A, Elkholi IE, Chen R, Seebun D, Mayne J, et al. Shedding of cancer susceptibility candidate 4 by the convertases PC7/furin unravels a novel secretory protein implicated in cancer progression. Cell Death Dis. 2020; Aug. 11(8):665.
Article
38. Rotte A. Combination of CTLA-4 and PD-1 blockers for treatment of cancer. J Exp Clin Cancer Res. 2019; Jun. 38(1):255.
Article
39. Nguyen LT, Ohashi PS. Clinical blockade of PD1 and LAG3: potential mechanisms of action. Nat Rev Immunol. 2015; Jan. 15(1):45–56.
Article
40. Prendergast GC, Mondal A, Dey S, Laury-Kleintop LD, Muller AJ. Inflammatory reprogramming with IDO1 inhibitors: turning immunologically unresponsive ‘cold’ tumors ‘hot’. Trends Cancer. 2018; Jan. 4(1):38–58.
Article
41. Lin J, Hou Y, Huang S, Wang Z, Sun C, Wang Z, et al. Exportin-T promotes tumor proliferation and invasion in hepatocellular carcinoma. Mol Carcinog. 2019; Feb. 58(2):293–304.
Article
42. Hojilla CV, Mohammed FF, Khokha R. Matrix metalloproteinases and their tissue inhibitors direct cell fate during cancer development. Br J Cancer. 2003; Nov. 89(10):1817–21.
Article
43. Bublik DR, Bursac S, Sheffer M, Orsolic I, Shalit T, Tarcic O, et al. Regulatory module involving FGF13, miR-504, and p53 regulates ribosomal biogenesis and supports cancer cell survival. Proc Natl Acad Sci U S A. 2017; Jan. 114(4):E496–505.
Article
44. Somanath PR, Razorenova OV, Chen J, Byzova TV. Akt1 in endothelial cell and angiogenesis. Cell Cycle. 2006; Mar. 5(5):512–8.
Article
45. Ding ZY, Li R, Zhang QJ, Wang Y, Jiang Y, Meng QY, et al. Prognostic role of cyclin D2/D3 in multiple human malignant neoplasms: a systematic review and meta-analysis. Cancer Med. 2019; Jun. 8(6):2717–29.
Article
46. Lv X, Li L, Lv L, Qu X, Jin S, Li K, et al. HOXD9 promotes epithelial-mesenchymal transition and cancer metastasis by ZEB1 regulation in hepatocellular carcinoma. J Exp Clin Cancer Res. 2015; Oct. 34:133.
Article
47. Zhu H, Dai W, Li J, Xiang L, Wu X, Tang W, et al. HOXD9 promotes the growth, invasion and metastasis of gastric cancer cells by transcriptional activation of RUFY3. J Exp Clin Cancer Res. 2019; Sep. 38(1):412.
Article
48. Verzella D, Pescatore A, Capece D, Vecchiotti D, Ursini MV, Franzoso G, et al. Life, death, and autophagy in cancer: NF-κB turns up everywhere. Cell Death Dis. 2020; Mar. 11(3):210.
Article
49. Dhawan P, Richmond A. A novel NF-kappa B-inducing kinase-MAPK signaling pathway up-regulates NF-kappa B activity in melanoma cells. J Biol Chem. 2002; Mar. 277(10):7920–8.
Full Text Links
  • CEO
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