Welcome to Francis Academic Press

Frontiers in Medical Science Research, 2022, 4(12); doi: 10.25236/FMSR.2022.041203.

Pathogenesis and Targeted Biologic Therapies for Thyroid-Associated Orbitopathy: A Review

Author(s)

Rui Ma1, Fuguang He2, Yueyue Chai1, Yuting Shao1, Fuqiang Liu1, Hong Lai1

Corresponding Author:
Hong Lai
Affiliation(s)

1Qilu Hospital of Shandong University, Jinan City, Shandong Province, China

2Pingyuan Hospital of Traditional Chinese Medicine, Dezhou City, Shandong Province, China

Abstract

Purpose: To review the pathogenesis and current targeted biologic therapies of Thyroid-associated orbitopathy (TAO). Methods: Pubmed, Cochrane Library databases, Wanfang database and recent relevant journal articles were searched. Results: TAO, a potentially vision-threatening disease, is the most common extrathyroidal manifestation of Graves’ Disease (GD). The complex pathogenesis of TAO involves in multiple factors, which has puzzled scientists for many years. With emerging advances in pathogenesis of TAO, novel therapeutic targets have been discovered. For example, rituximab (that acts against CD20), tocilizumab (that acts against the IL-6 receptor), teprotumumab (that blocks IGF-1R) and TNF-αinhibitors have proven to be useful and safe therapeutic options in TAO treatment. Conclusions: Targeted biological agents can provide many benefits for TAO treatments. And there is still a long way to go before the potential molecular pathogenesis and novel treatment options of GO are revealed.

Keywords

Thyroid-Associated Orbitopathy, Pathogenesis, Monoclonal Antibodies

Cite This Paper

Rui Ma, Fuguang He, Yueyue Chai, Yuting Shao, Fuqiang Liu, Hong Lai. Pathogenesis and Targeted Biologic Therapies for Thyroid-Associated Orbitopathy: A Review. Frontiers in Medical Science Research (2022) Vol. 4, Issue 12: 11-19. https://doi.org/10.25236/FMSR.2022.041203.

References

[1] Hodgson, N.M. and F. Rajaii, Current Understanding of the Progression and Management of Thyroid Associated Orbitopathy: A Systematic Review. Ophthalmol Ther, 2020. 9(1): p. 21-33.

[2] Smith, T.J. and L. Hegedüs, Graves' Disease. N Engl J Med, 2016. 375(16): p. 1552-1565.

[3] Cao, J.M., et al., Epigenetics effect on pathogenesis of thyroid-associated ophthalmopathy. Int J Ophthalmol, 2021. 14(9): p. 1441-1448.

[4] Men, C.J., A.L. Kossler, and S.T. Wester, Updates on the understanding and management of thyroid eye disease. Ther Adv Ophthalmol, 2021. 13: p. 25158414211027760.

[5] Rundle, F.F. and C.W. Wilson, Development and course of exophthalmos and ophthalmoplegia in Graves' disease with special reference to the effect of thyroidectomy. Clin Sci, 1945. 5(3-4): p. 177-94.

[6] Mishra, S., et al., Clinical Management and Therapeutic Strategies for the Thyroid-Associated Ophthalmopathy: Current and Future Perspectives. Curr Eye Res, 2020. 45(11): p. 1325-1341.

[7] Antonelli, A., et al., Autoimmune thyroid disorders. Autoimmun Rev, 2015. 14(2): p. 174-80.

[8] Neag, E.J. and T.J. Smith, 2021 update on thyroid-associated ophthalmopathy. J Endocrinol Invest, 2022. 45(2): p. 235-259.

[9] Huang, Y., et al., Progress in the pathogenesis of thyroid-associated ophthalmopathy and new drug development. Taiwan J Ophthalmol, 2020. 10(3): p. 174-180.

[10] Feliciello, A., et al., Expression of thyrotropin-receptor mRNA in healthy and Graves' disease retro-orbital tissue. Lancet, 1993. 342(8867): p. 337-8.

[11] Heufelder, A.E., et al., Detection of TSH receptor RNA in cultured fibroblasts from patients with Graves' ophthalmopathy and pretibial dermopathy. Thyroid, 1993. 3(4): p. 297-300.

[12] Starkey, K.J., et al., Adipose thyrotrophin receptor expression is elevated in Graves' and thyroid eye diseases ex vivo and indicates adipogenesis in progress in vivo. J Mol Endocrinol, 2003. 30(3): p. 369-80.

[13] Khoo, D.H., et al., Graves' ophthalmopathy in the absence of elevated free thyroxine and triiodothyronine levels: prevalence, natural history, and thyrotropin receptor antibody levels. Thyroid, 2000. 10(12): p. 1093-100.

[14] Khoo, D.H., et al., The combination of absent thyroid peroxidase antibodies and high thyroid-stimulating immunoglobulin levels in Graves' disease identifies a group at markedly increased risk of ophthalmopathy. Thyroid, 1999. 9(12): p. 1175-80.

[15] Iyer, S. and R. Bahn, Immunopathogenesis of Graves' ophthalmopathy: the role of the TSH receptor. Best Pract Res Clin Endocrinol Metab, 2012. 26(3): p. 281-9.

[16] Pritchard, J., et al., Immunoglobulin activation of T cell chemoattractant expression in fibroblasts from patients with Graves' disease is mediated through the insulin-like growth factor I receptor pathway. J Immunol, 2003. 170(12): p. 6348-54.

[17] Tsui, S., et al., Evidence for an association between thyroid-stimulating hormone and insulin-like growth factor 1 receptors: a tale of two antigens implicated in Graves' disease. J Immunol, 2008. 181(6): p. 4397-405.

[18] Minich, W.B., et al., Autoantibodies to the IGF1 receptor in Graves' orbitopathy. J Clin Endocrinol Metab, 2013. 98(2): p. 752-60.

[19] Smith, T.J., L. Hegedüs, and R.S. Douglas, Role of insulin-like growth factor-1 (IGF-1) pathway in the pathogenesis of Graves' orbitopathy. Best Pract Res Clin Endocrinol Metab, 2012. 26(3): p. 291-302.

[20] Armitage, R.J., et al., Molecular and biological characterization of a murine ligand for CD40. Nature, 1992. 357(6373): p. 80-2.

[21] Smith, M.J., et al., Activation of thyroid antigen-reactive B cells in recent onset autoimmune thyroid disease patients. J Autoimmun, 2018. 89: p. 82-89.

[22] Yanaba, K., et al., B-lymphocyte contributions to human autoimmune disease. Immunol Rev, 2008. 223: p. 284-99.

[23] Prabhakar, B.S., R.S. Bahn, and T.J. Smith, Current perspective on the pathogenesis of Graves' disease and ophthalmopathy. Endocr Rev, 2003. 24(6): p. 802-35.

[24] Howland, K.C., et al., The roles of CD28 and CD40 ligand in T cell activation and tolerance. J Immunol, 2000. 164(9): p. 4465-70.

[25] Huang, Y., et al., The involvement of T cell pathogenesis in thyroid-associated ophthalmopathy. Eye (Lond), 2019. 33(2): p. 176-182.

[26] Otto, E.A., et al., Orbital tissue-derived T lymphocytes from patients with Graves' ophthalmopathy recognize autologous orbital antigens. J Clin Endocrinol Metab, 1996. 81(8): p. 3045-50.

[27] Bahn, R.S., Graves' ophthalmopathy. N Engl J Med, 2010. 362(8): p. 726-38.

[28] Feldon, S.E., et al., Autologous T-lymphocytes stimulate proliferation of orbital fibroblasts derived from patients with Graves' ophthalmopathy. Invest Ophthalmol Vis Sci, 2005. 46(11): p. 3913-21.

[29] Fang, S., et al., IL-17A Exacerbates Fibrosis by Promoting the Proinflammatory and Profibrotic Function of Orbital Fibroblasts in TAO. J Clin Endocrinol Metab, 2016. 101(8): p. 2955-65.

[30] Dik, W.A., S. Virakul, and L. van Steensel, Current perspectives on the role of orbital fibroblasts in the pathogenesis of Graves' ophthalmopathy. Exp Eye Res, 2016. 142: p. 83-91.

[31] Khoo, T.K., et al., Evidence for enhanced Thy-1 (CD90) expression in orbital fibroblasts of patients with Graves' ophthalmopathy. Thyroid, 2008. 18(12): p. 1291-6.

[32] Koumas, L., et al., Thy-1 expression in human fibroblast subsets defines myofibroblastic or lipofibroblastic phenotypes. Am J Pathol, 2003. 163(4): p. 1291-300.

[33] Smith, T.J., Potential Roles of CD34+ Fibrocytes Masquerading as Orbital Fibroblasts in Thyroid-Associated Ophthalmopathy. J Clin Endocrinol Metab, 2019. 104(2): p. 581-594.

[34] Douglas, R.S., et al., Increased generation of fibrocytes in thyroid-associated ophthalmopathy. J Clin Endocrinol Metab, 2010. 95(1): p. 430-8.

[35] Wu, T., et al., Thyrotropin and CD40L Stimulate Interleukin-12 Expression in Fibrocytes: Implications for Pathogenesis of Thyroid-Associated Ophthalmopathy. Thyroid, 2016. 26(12): p. 1768-1777.

[36] Shen, S., et al., B-cell targeted therapy with rituximab for thyroid eye disease: closer to the clinic. Surv Ophthalmol, 2013. 58(3): p. 252-65.

[37] Reff, M.E., et al., Depletion of B cells in vivo by a chimeric mouse human monoclonal antibody to CD20. Blood, 1994. 83(2): p. 435-45.

[38] Smith, M.R., Rituximab (monoclonal anti-CD20 antibody): mechanisms of action and resistance. Oncogene, 2003. 22(47): p. 7359-68.

[39] El Fassi, D., et al., The rationale for B lymphocyte depletion in Graves' disease. Monoclonal anti-CD20 antibody therapy as a novel treatment option. Eur J Endocrinol, 2006. 154(5): p. 623-32.

[40] Edwards, J.C., et al., Efficacy of B-cell-targeted therapy with rituximab in patients with rheumatoid arthritis. N Engl J Med, 2004. 350(25): p. 2572-81.

[41] Stan, M.N., et al., Randomized controlled trial of rituximab in patients with Graves' orbitopathy. J Clin Endocrinol Metab, 2015. 100(2): p. 432-41.

[42] Savino, G., et al., Intraorbital injection of rituximab versus high dose of systemic glucocorticoids in the treatment of thyroid-associated orbitopathy. Endocrine, 2015. 48(1): p. 241-7.

[43] Salvi, M., et al., Efficacy of B-cell targeted therapy with rituximab in patients with active moderate to severe Graves' orbitopathy: a randomized controlled study. J Clin Endocrinol Metab, 2015. 100(2): p. 422-31.

[44] Chen, J., G. Chen, and H. Sun, Intravenous rituximab therapy for active Graves' ophthalmopathy: a meta-analysis. Hormones (Athens), 2021. 20(2): p. 279-286.

[45] Li, J., et al., The Efficacy of Rituximab Combined with 131I for Ophthalmic Outcomes of Graves' Ophthalmopathy Patients. Pharmacology, 2017. 99(3-4): p. 144-152.

[46] Erdei, A., et al., Rapid response to and long-term effectiveness of anti-CD20 antibody in conventional therapy resistant Graves' orbitopathy: A five-year follow-up study. Autoimmunity, 2014. 47(8): p. 548-55.

[47] Mitchell, A.L., et al., The effect of B cell depletion therapy on anti-TSH receptor antibodies and clinical outcome in glucocorticoid-refractory Graves' orbitopathy. Clin Endocrinol (Oxf), 2013. 79(3): p. 437-42.

[48] Insull, E.A., et al., Early low-dose rituximab for active thyroid eye disease: An effective and well-tolerated treatment. Clin Endocrinol (Oxf), 2019. 91(1): p. 179-186.

[49] Du Pasquier-Fediaevsky, L., et al., Low-Dose Rituximab for Active Moderate to Severe Graves' Orbitopathy Resistant to Conventional Treatment. Ocul Immunol Inflamm, 2019. 27(5): p. 844-850.

[50] Eid, L., et al., The effects of Rituximab on Graves'orbitopathy: A retrospective study of 14 patients. Eur J Ophthalmol, 2020. 30(5): p. 1008-1013.

[51] Deltour, J.B., et al., Efficacy of rituximab in patients with Graves' orbitopathy: a retrospective multicenter nationwide study. Graefes Arch Clin Exp Ophthalmol, 2020. 258(9): p. 2013-2021.

[52] Silkiss, R.Z., et al., Rituximab for thyroid eye disease. Ophthalmic Plast Reconstr Surg, 2010. 26(5): p. 310-4.

[53] Salvi, M., et al., Treatment of Graves' disease and associated ophthalmopathy with the anti-CD20 monoclonal antibody rituximab: an open study. Eur J Endocrinol, 2007. 156(1): p. 33-40.

[54] El Fassi, D., et al., B lymphocyte depletion with the monoclonal antibody rituximab in Graves' disease: a controlled pilot study. J Clin Endocrinol Metab, 2007. 92(5): p. 1769-72.

[55] Copperman, T., et al., Subcutaneous Tocilizumab for Thyroid Eye Disease: Simplified Dosing and Delivery. Ophthalmic Plast Reconstr Surg, 2019. 35(3): p. e64-e66.

[56] Jyonouchi, S.C., et al., Interleukin-6 stimulates thyrotropin receptor expression in human orbital preadipocyte fibroblasts from patients with Graves' ophthalmopathy. Thyroid, 2001. 11(10): p. 929-34.

[57] Leszczynska, A., et al., Cytokine production in thyroid eye disease: in vitro effects of dexamethasone and IL-6 blockade with tocilizumab. Graefes Arch Clin Exp Ophthalmol, 2019. 257(10): p. 2307-2314.

[58] Smolen, J.S. and G. Steiner, Therapeutic strategies for rheumatoid arthritis. Nat Rev Drug Discov, 2003. 2(6): p. 473-88.

[59] Bartalena, L., et al., The 2021 European Group on Graves' orbitopathy (EUGOGO) clinical practice guidelines for the medical management of Graves' orbitopathy. Eur J Endocrinol, 2021. 185(4): p. G43-g67.

[60] Perez-Moreiras, J.V., et al., Efficacy of Tocilizumab in Patients With Moderate-to-Severe Corticosteroid-Resistant Graves Orbitopathy: A Randomized Clinical Trial. Am J Ophthalmol, 2018. 195: p. 181-190.

[61] Sánchez-Bilbao, L., et al., Anti-IL-6 Receptor Tocilizumab in Refractory Graves' Orbitopathy: National Multicenter Observational Study of 48 Patients. J Clin Med, 2020. 9(9).

[62] Pérez-Moreiras, J.V., et al., Steroid-Resistant Graves' Orbitopathy Treated with Tocilizumab in Real-World Clinical Practice: A 9-Year Single-Center Experience. J Clin Med, 2021. 10(4).

[63] Bennedjaï, A., et al., Tocilizumab versus Rituximab in Patients with Moderate to Severe Steroid-resistant Graves' Orbitopathy. Ocul Immunol Inflamm, 2020: p. 1-6.

[64] Burmester, G.R., et al., A randomised, double-blind, parallel-group study of the safety and efficacy of subcutaneous tocilizumab versus intravenous tocilizumab in combination with traditional disease-modifying antirheumatic drugs in patients with moderate to severe rheumatoid arthritis (SUMMACTA study). Ann Rheum Dis, 2014. 73(1): p. 69-74.

[65] Stevens, S.M., N. Pirakitikulr, and B.W. Lee, Subcutaneous tocilizumab for active thyroid eye disease refractory to orbital radiation and systemic steroids in tobacco smokers. Taiwan J Ophthalmol, 2022. 12(1): p. 39-43.

[66] Silkiss, R.Z., et al., Treatment of corticosteroid-resistant thyroid eye disease with subcutaneous tocilizumab. Can J Ophthalmol, 2021. 56(1): p. 66-70.

[67] Chen, H., et al., Teprotumumab, an IGF-1R blocking monoclonal antibody inhibits TSH and IGF-1 action in fibrocytes. J Clin Endocrinol Metab, 2014. 99(9): p. E1635-40.

[68] Chen, H., et al., TSH-Mediated TNFα Production in Human Fibrocytes Is Inhibited by Teprotumumab, an IGF-1R Antagonist. PLoS One, 2015. 10(6): p. e0130322.

[69] Markham, A., Teprotumumab: First Approval. Drugs, 2020. 80(5): p. 509-512.

[70] Ju, Y. and J. Yang, Teprotumumab for the treatment of thyroid eye disease. Expert Rev Clin Immunol, 2020. 16(8): p. 739-743.

[71] Ding, Y., S. Yang, and H. Gao, Teprotumumab: The Dawn of Therapies in Moderate-to-Severe Thyroid-Associated Ophthalmopathy. Horm Metab Res, 2021. 53(4): p. 211-218.

[72] Smith, T.J., et al., Teprotumumab for Thyroid-Associated Ophthalmopathy. N Engl J Med, 2017. 376(18): p. 1748-1761.

[73] Douglas, R.S., et al., Teprotumumab for the Treatment of Active Thyroid Eye Disease. N Engl J Med, 2020. 382(4): p. 341-352.

[74] Kapadia, M.K. and P.A. Rubin, The emerging use of TNF-alpha inhibitors in orbital inflammatory disease. Int Ophthalmol Clin, 2006. 46(2): p. 165-81.

[75] Baughman, R.P., D.A. Bradley, and E.E. Lower, Infliximab in chronic ocular inflammation. Int J Clin Pharmacol Ther, 2005. 43(1): p. 7-11.

[76] Prendiville, C., et al., The use of infliximab in ocular inflammation. Br J Ophthalmol, 2008. 92(6): p. 823-5.

[77] Garrity, J.A., et al., Treatment of recalcitrant idiopathic orbital inflammation (chronic orbital myositis) with infliximab. Am J Ophthalmol, 2004. 138(6): p. 925-30.

[78] Sobrin, L., et al., Infliximab therapy for the treatment of refractory ocular inflammatory disease. Arch Ophthalmol, 2007. 125(7): p. 895-900.

[79] van Steensel, L., et al., Whole orbital tissue culture identifies imatinib mesylate and adalimumab as potential therapeutics for Graves' ophthalmopathy. Br J Ophthalmol, 2011. 95(5): p. 735-8.

[80] Ayabe, R., et al., Adalimumab as steroid-sparing treatment of inflammatory-stage thyroid eye disease. Ophthalmic Plast Reconstr Surg, 2014. 30(5): p. 415-9.

[81] Paridaens, D., et al., The effect of etanercept on Graves' ophthalmopathy: a pilot study. Eye (Lond), 2005. 19(12): p. 1286-9.

[82] Boskovic, O., et al., Etanercept in the treatment of Graves' ophthalmopathy with primary hypothyroidism and rheumatoid arthritis. Cent Eur J Immunol, 2019. 44(4): p. 463-465.

[83] Bartalena, L., et al., Epidemiology, Natural History, Risk Factors, and Prevention of Graves' Orbitopathy. Front Endocrinol (Lausanne), 2020. 11: p. 615993.