open access

Vol 70, No 5 (2019)
Review paper
Submitted: 2019-02-22
Accepted: 2019-04-04
Published online: 2019-10-25
Get Citation

Clinical implications of GWAS variants associated with differentiated thyroid cancer

Jarosław P. Jendrzejewski1, Krzysztof Sworczak1, Daniel F. Comiskey2, Albert de la Chapelle2
·
Pubmed: 31681970
·
Endokrynol Pol 2019;70(5):423-429.
Affiliations
  1. Department of Endocrinology and Internal Medicine, Medical University of Gdansk, Gdansk, Poland
  2. Human Cancer Genetics Program, Comprehensive Cancer Centre, The Ohio State University, Columbus, Ohio, United States

open access

Vol 70, No 5 (2019)
Review Article
Submitted: 2019-02-22
Accepted: 2019-04-04
Published online: 2019-10-25

Abstract

The genetic risk of differentiated thyroid cancer (DTC) probably consists of multiple low-penetrance, single-nucleotide polymorphisms (SNP). Such markers are difficult to uncover by linkage analysis but can be revealed by association studies. Genome-wide association studies (GWASs) have uncovered 31 SNPs associated with DTC. These markers carry a low to moderate risk for DTC, but their cumulative effect increases with each successive risk allele. These data support the important contribution of low penetrance variants in the pathogenesis of DTC. Contrary to somatic mutations such as BRAFV600E, germline variants can be ascertained prior to surgical treatment. Therefore, we hypothesise that GWAS SNPs might impact the clinical course of DTC and we can benefit from this knowledge in choosing a treatment strategy. Several associations between clinical factors and GWAS markers have been reported so far. The most important are associations between rs966423 and mortality (HR = 1.60, p = 0.038), extrathyroidal extension (ETE) (OR = 1.57, p = 0.019); rs965513 and tumour diameter (slope of regression 0.14, p = 0.025), lymph node metastasis (OR = 1.59, p = 0.030) and ETE (OR = 1.29, p = 0.045); rs944289 and distant metastasis (OR = 0.58, p = 0.042); and rs116909374 and lymph node metastasis (OR = 0.61, p = 0.016). These findings show that GWAS SNPs are not only the ignition factors (together with environmental factors) for malignant transformation of thyrocytes but might also impact the clinical course of DTC. Surprisingly, it is not always the risk allele for DTC that is associated with worse clinical outcome. The second interesting observation is that GWAS SNPs show different associations with DTC clinical features depending on their histological subtypes. These point to the complexity of DTC with putatively different roles of genes at different stages of DTC development. (Endokrynol Pol 2019; 70 (5): 423–429)

Abstract

The genetic risk of differentiated thyroid cancer (DTC) probably consists of multiple low-penetrance, single-nucleotide polymorphisms (SNP). Such markers are difficult to uncover by linkage analysis but can be revealed by association studies. Genome-wide association studies (GWASs) have uncovered 31 SNPs associated with DTC. These markers carry a low to moderate risk for DTC, but their cumulative effect increases with each successive risk allele. These data support the important contribution of low penetrance variants in the pathogenesis of DTC. Contrary to somatic mutations such as BRAFV600E, germline variants can be ascertained prior to surgical treatment. Therefore, we hypothesise that GWAS SNPs might impact the clinical course of DTC and we can benefit from this knowledge in choosing a treatment strategy. Several associations between clinical factors and GWAS markers have been reported so far. The most important are associations between rs966423 and mortality (HR = 1.60, p = 0.038), extrathyroidal extension (ETE) (OR = 1.57, p = 0.019); rs965513 and tumour diameter (slope of regression 0.14, p = 0.025), lymph node metastasis (OR = 1.59, p = 0.030) and ETE (OR = 1.29, p = 0.045); rs944289 and distant metastasis (OR = 0.58, p = 0.042); and rs116909374 and lymph node metastasis (OR = 0.61, p = 0.016). These findings show that GWAS SNPs are not only the ignition factors (together with environmental factors) for malignant transformation of thyrocytes but might also impact the clinical course of DTC. Surprisingly, it is not always the risk allele for DTC that is associated with worse clinical outcome. The second interesting observation is that GWAS SNPs show different associations with DTC clinical features depending on their histological subtypes. These point to the complexity of DTC with putatively different roles of genes at different stages of DTC development. (Endokrynol Pol 2019; 70 (5): 423–429)

Get Citation

Keywords

genome-wide association studies; GWAS; papillary thyroid carcinoma; PTC; genetic predisposition

About this article
Title

Clinical implications of GWAS variants associated with differentiated thyroid cancer

Journal

Endokrynologia Polska

Issue

Vol 70, No 5 (2019)

Article type

Review paper

Pages

423-429

Published online

2019-10-25

Page views

1673

Article views/downloads

1290

DOI

10.5603/EP.a2019.0027

Pubmed

31681970

Bibliographic record

Endokrynol Pol 2019;70(5):423-429.

Keywords

genome-wide association studies
GWAS
papillary thyroid carcinoma
PTC
genetic predisposition

Authors

Jarosław P. Jendrzejewski
Krzysztof Sworczak
Daniel F. Comiskey
Albert de la Chapelle

References (57)
  1. Goldgar DE, Easton DF, Cannon-Albright LA, et al. Systematic population-based assessment of cancer risk in first-degree relatives of cancer probands. J Natl Cancer Inst. 1994; 86(21): 1600–1608.
  2. Dong C, Hemminki K. Modification of cancer risks in offspring by sibling and parental cancers from 2,112,616 nuclear families. Int J Cancer. 2001; 92(1): 144–150.
  3. Risch N. The genetic epidemiology of cancer: interpreting family and twin studies and their implications for molecular genetic approaches. Cancer Epidemiol Biomarkers Prev. 2001; 10(7): 733–741.
  4. Czene K, Lichtenstein P, Hemminki K. Environmental and heritable causes of cancer among 9.6 million individuals in the Swedish Family-Cancer Database. Int J Cancer. 2002; 99(2): 260–266.
  5. Eng C, Clayton D, Schuffenecker I, et al. The relationship between specific RET proto-oncogene mutations and disease phenotype in multiple endocrine neoplasia type 2. International RET mutation consortium analysis. JAMA. 1996; 276(19): 1575–1579.
  6. Donis-Keller H, Dou S, Chi D, et al. Mutations in the RET proto-oncogene are associated with MEN 2A and FMTC. Hum Mol Genet. 1993; 2(7): 851–856.
  7. Marsh D, Learoyd D, Andrew S, et al. Somatic mutations in theRETproto-oncogene in sporadic medullary thyroid carcinoma. Clini Endocrinol (Oxf). 1996; 44(3): 249–257.
  8. Jarząb B, Dedecjus M, Handkiewicz-Junak D, et al. Diagnostics and Treatment of Thyroid Carcinoma. Endokrynol Pol. 2016; 67(1): 74–107.
  9. Hemminki K, Dong C. Familial relationships in thyroid cancer by histo-pathological type. Int J Cancer. 2000; 85(2): 201–205.
  10. Hrafnkelsson J, Tulinius H, Jónasson JG, et al. Familial non-medullary thyroid cancer in Iceland. J Med Genet. 2001; 38(3): 189–191.
  11. Frich L, Glattre E, Akslen, A. Familial occurrence of nonmedullary thyroid cancer: a population-based study of 5673 first-degree relatives of thyroid cancer patients from Norway. Cancer Epidemiol Biomarkers Prev. 2001; 10(2): 113–117.
  12. Peiling Yang S, Ngeow J. Familial non-medullary thyroid cancer: unraveling the genetic maze. Endocr Relat Cancer. 2016; 23(12): R577–R595.
  13. International HapMap Consortium. A haplotype map of the human genome. Nature. 2005; 437(7063): 1299–1320.
  14. Wang DG. Large-Scale Identification, Mapping, and Genotyping of Single-Nucleotide Polymorphisms in the Human Genome. Science. 1998; 280(5366): 1077–1082.
  15. LaFramboise T. Single nucleotide polymorphism arrays: a decade of biological, computational and technological advances. Nucleic Acids Res. 2009; 37(13): 4181–4193.
  16. Klein RJ, Zeiss C, Chew EY, et al. Complement factor H polymorphism in age-related macular degeneration. Science. 2005; 308(5720): 385–389.
  17. Thakkinstian A, Han P, McEvoy M, et al. Systematic review and meta-analysis of the association between complement factor H Y402H polymorphisms and age-related macular degeneration. Hum Mol Genet. 2006; 15(18): 2784–2790.
  18. Despriet DDG, Klaver CCW, Witteman JCM, et al. Complement factor H polymorphism, complement activators, and risk of age-related macular degeneration. JAMA. 2006; 296(3): 301–309.
  19. Shaw PX, Zhang Li, Zhang M, et al. Complement factor H genotypes impact risk of age-related macular degeneration by interaction with oxidized phospholipids. Proc Natl Acad Sci USA. 2012; 109(34): 13757–13762.
  20. Gudmundsson J, Thorleifsson G, Sigurdsson JK, et al. A genome-wide association study yields five novel thyroid cancer risk loci. Nat Commun. 2017; 8: 14517.
  21. Gudmundsson J, Sulem P, Gudbjartsson DF, et al. Common variants on 9q22.33 and 14q13.3 predispose to thyroid cancer in European populations. Nat Genet. 2009; 41(4): 460–464.
  22. Gudmundsson J, Sulem P, Gudbjartsson DF, et al. Discovery of common variants associated with low TSH levels and thyroid cancer risk. Nat Genet. 2012; 44(3): 319–322.
  23. Takahashi M, Saenko VA, Rogounovitch TI, et al. The FOXE1 locus is a major genetic determinant for radiation-related thyroid carcinoma in Chernobyl. Hum Mol Genet. 2010; 19(12): 2516–2523.
  24. Mancikova V, Cruz R, Inglada-Pérez L, et al. Thyroid cancer GWAS identifies 10q26.12 and 6q14.1 as novel susceptibility loci and reveals genetic heterogeneity among populations. Int J Cancer. 2015; 137(8): 1870–1878.
  25. Son HY, Hwangbo Y, Yoo SK, et al. Genome-wide association and expression quantitative trait loci studies identify multiple susceptibility loci for thyroid cancer. Nat Commun. 2017; 8: 15966.
  26. Köhler A, Chen B, Gemignani F, et al. Genome-wide association study on differentiated thyroid cancer. J Clin Endocrinol Metab. 2013; 98(10): E1674–E1681.
  27. Figlioli G, Köhler A, Chen B, et al. Novel genome-wide association study-based candidate loci for differentiated thyroid cancer risk. J Clin Endocrinol Metab. 2014; 99(10): E2084–E2092.
  28. Kimura S. Thyroid-specific transcription factors and their roles in thyroid cancer. J Thyroid Res. 2011; 2011: 710213.
  29. Visscher PM, Wray NR, Zhang Q, et al. 10 Years of GWAS Discovery: Biology, Function, and Translation. Am J Hum Genet. 2017; 101(1): 5–22.
  30. Iyengar SK, Elston RC. The genetic basis of complex traits: rare variants or "common gene, common disease"? Methods Mol Biol. 2007; 376: 71–84.
  31. Liyanarachchi S, Wojcicka A, Li W, et al. Cumulative risk impact of five genetic variants associated with papillary thyroid carcinoma. Thyroid. 2013; 23(12): 1532–1540.
  32. Figlioli G, Chen B, Elisei R, et al. Novel genetic variants in differentiated thyroid cancer and assessment of the cumulative risk. Sci Rep. 2015; 5: 8922.
  33. Wei WJ, Lu ZW, Wang Yu, et al. Clinical significance of papillary thyroid cancer risk loci identified by genome-wide association studies. Cancer Genet. 2015; 208(3): 68–75.
  34. Haugen BR. 2015 American Thyroid Association Management Guidelines for Adult Patients with Thyroid Nodules and Differentiated Thyroid Cancer: What is new and what has changed? Cancer. 2017; 123(3): 372–381.
  35. Mankarios D, Baade P, Youl P, et al. Validation of the QTNM staging system for cancer-specific survival in patients with differentiated thyroid cancer. Endocrine. 2014; 46(2): 300–308.
  36. Yang L, Shen W, Sakamoto N. Population-based study evaluating and predicting the probability of death resulting from thyroid cancer and other causes among patients with thyroid cancer. J Clin Oncol. 2013; 31(4): 468–474.
  37. He H, Li W, Liyanarachchi S, et al. Genetic predisposition to papillary thyroid carcinoma: involvement of FOXE1, TSHR, and a novel lincRNA gene, PTCSC2. J Clin Endocrinol Metab. 2015; 100(1): E164–E172.
  38. He H, Li W, Liyanarachchi S, et al. Multiple functional variants in long-range enhancer elements contribute to the risk of SNP rs965513 in thyroid cancer. Proc Natl Acad Sci USA. 2015; 112(19): 6128–6133.
  39. Wang Y, He H, Li W, et al. MYH9 binds to lncRNA gene PTCSC2 and regulates FOXE1 in the 9q22 thyroid cancer risk locus. Proc Natl Acad Sci USA. 2017; 114(3): 474–479.
  40. Saenko VA, Rogounovitch TI. Genetic Polymorphism Predisposing to Differentiated Thyroid Cancer: A Review of Major Findings of the Genome-Wide Association Studies. Endocrinol Metab (Seoul). 2018; 33(2): 164–174.
  41. Jendrzejewski J, Liyanarachchi S, Nagy R, et al. Papillary Thyroid Carcinoma: Association Between Germline DNA Variant Markers and Clinical Parameters. Thyroid. 2016; 26(9): 1276–1284.
  42. Jendrzejewski J, Liyanarachchi S, Eiterman A, et al. Fine mapping of 14q13 reveals novel variants associated with different histological subtypes of papillary thyroid carcinoma. Int J Cancer. 2019; 144(3): 503–512.
  43. Randolph GW, Duh QY, Heller KS, et al. American Thyroid Association Surgical Affairs Committee’s Taskforce on Thyroid Cancer Nodal Surgery. 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; 22(11): 1144–1152.
  44. Podnos YD, Smith D, Wagman LD, et al. The implication of lymph node metastasis on survival in patients with well-differentiated thyroid cancer. Am Surg. 2005; 71(9): 731–734.
  45. Adam MA, Pura J, Goffredo P, et al. Presence and Number of Lymph Node Metastases Are Associated With Compromised Survival for Patients Younger Than Age 45 Years With Papillary Thyroid Cancer. J Clin Oncol. 2015; 33(21): 2370–2375.
  46. Yip, L, Nikiforova MN, Yoo JY, et al. Tumor genotype determines phenotype and disease-related outcomes in thyroid cancer: a study of 1510 patients. Ann Surg. 2015 ; 262(3 ): 519–525; discussion 524–525.
  47. Hwangbo Y, Lee E, Son HY, et al. Genome-Wide Association Study Reveals Distinct Genetic Susceptibility of Thyroid Nodules From Thyroid Cancer. J Clin Endocrinol Metab. 2018; 103(12): 4384–4394.
  48. Wang YL, Feng SH, Guo SC, et al. Confirmation of papillary thyroid cancer susceptibility loci identified by genome-wide association studies of chromosomes 14q13, 9q22, 2q35 and 8p12 in a Chinese population. J Med Genet. 2013; 50(10): 689–695.
  49. Rogounovitch TI, Bychkov A, Takahashi M, et al. The common genetic variant rs944289 on chromosome 14q13.3 associates with risk of both malignant and benign thyroid tumors in the Japanese population. Thyroid. 2015; 25(3): 333–340.
  50. Świerniak M, Wójcicka A, Czetwertyńska M, et al. Association between GWAS-Derived rs966423 Genetic Variant and Overall Mortality in Patients with Differentiated Thyroid Cancer. Clin Cancer Res. 2016; 22(5): 1111–1119.
  51. Zhao L, Zhang Q, Luan X, et al. STAT3 and STAT5b polymorphism contributes to breast cancer risk and clinical outcomes. Int J Clin Exp Pathol. 2015; 8(2): 2033–2038.
  52. Jendrzejewski J, He H, Radomska HS, et al. The polymorphism rs944289 predisposes to papillary thyroid carcinoma through a large intergenic noncoding RNA gene of tumor suppressor type. Proc Natl Acad Sci USA. 2012; 109(22): 8646–8651.
  53. Liyanarachchi S, Li W, Yan P, et al. Genome-Wide Expression Screening Discloses Long Noncoding RNAs Involved in Thyroid Carcinogenesis. J Clin Endocrinol Metab. 2016; 101(11): 4005–4013.
  54. Penna-Martinez M, Epp F, Kahles H, et al. FOXE1 association with differentiated thyroid cancer and its progression. Thyroid. 2014; 24(5): 845–851.
  55. Cunha LL, Morari EC, Guihen AC, et al. Infiltration of a mixture of different immune cells may be related to molecular profile of differentiated thyroid cancer. Endocr Relat Cancer. 2012; 19(3): L31–L36.
  56. Al Afif A, Williams BA, Rigby MH, et al. Multifocal Papillary Thyroid Cancer Increases the Risk of Central Lymph Node Metastasis. Thyroid. 2015; 25(9): 1008–1012.
  57. Bodmer D, Schepens M, Eleveld MJ, et al. Disruption of a novel gene, DIRC3, and expression of DIRC3-HSPBAP1 fusion transcripts in a case of familial renal cell cancer and t(2;3)(q35;q21). Genes Chromosomes Cancer. 2003; 38(2): 107–116.

Regulations

Important: This website uses cookies. More >>

The cookies allow us to identify your computer and find out details about your last visit. They remembering whether you've visited the site before, so that you remain logged in - or to help us work out how many new website visitors we get each month. Most internet browsers accept cookies automatically, but you can change the settings of your browser to erase cookies or prevent automatic acceptance if you prefer.

Via MedicaWydawcą jest  VM Media Group sp. z o.o., Grupa Via Medica, ul. Świętokrzyska 73, 80–180 Gdańsk

tel.:+48 58 320 94 94, faks:+48 58 320 94 60, e-mail:  viamedica@viamedica.pl