Skip to main content

EDITORIAL article

Front. Endocrinol., 29 April 2022
Sec. Thyroid Endocrinology
This article is part of the Research Topic Mechanisms and Novel Therapies in Graves’ Orbitopathy: Current Update View all 21 articles

Editorial: Mechanisms and Novel Therapies in Graves’ Orbitopathy: Current Update

  • 1Department of Ophthalmology, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
  • 2Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
  • 3Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
  • 4Department of Endocrinology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
  • 5Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
  • 6Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China

Graves’ orbitopathy (GO) is the main extra-thyroidal manifestation in patients with Graves’ disease (GD), with a prevalence of up to 50%, although in most cases GO is mild (1). GO has a variety of clinical manifestations including eyelid retraction, exophthalmos, restrictive strabismus, exposure keratitis, and dysthyroid optic neuropathy. It is the commonest orbital disease causing blindness, orbital deformity and visual disability and exerts a profound negative impact on patients’ quality of life (2).

This Research Topic provides a timely update on different aspects of GO, in 20 excellent contributions, ranging from epidemiology, pathogenesis, disease evaluation, to comments on existing therapies and novel treatment strategies. Bartalena et al. reviewed GO incidence and prevalence, then highlighted risk factors such as age and smoking, and how they might be regulated or avoided to optimize clinical management. Clinically, hyperthyroidism and orbitopathy often develop simultaneously or within a few months of each other (2). Both thyroid epithelia and orbital fibroblasts (OF) express the thyrotropin receptor (TSHR), suggesting that these two conditions may evolve from common underlying systemic processes (1). Uncontrolled hyperthyroidism, which is caused by autoantibodies (TRAB) to the TSHR in GD, is a major GO risk factor with high TRAB levels long known to correlate with GO incidence and severity (3). In this context, George et al. provided a comprehensive overview introducing thyroid-stimulating antibody (TSAb) as a biomarker for GO whilst Kelada et al. investigated polyautoimmunity as a risk factor of GO activity and severity from their retrospective cohort of 267 GO patients.

The pathogenesis of GO is complex and not fully understood although some consensus has emerged in recent years (4). Most would regard the OFs as target cells contributing to the tissue remodeling which leads to expansion of the orbital contents. Draman et al. summarized OF signaling cascades and how they affect two of these processes, adipogenesis and hyaluronan production. To further our understanding of OFs, Virakul et al. compared the proteome of DNA methylation of OFs from GO patients and healthy controls. They found over-expression of genes implicated in inflammation and proliferation, together with subtle differences between active and inactive GO profiles. A similar microarray approach was applied to the GO lacrimal gland by Tu et al.

GO is an autoimmune disease and orbital inflammation is initiated by the loss of self-tolerance to the putative, shared antigens between thyroid glands and orbits, notably the TSHR (5). Antigen-presenting cells (APC) recognize and present TSHR to T helper (Th) cells for unknown reasons, and the existence of a soluble TSHR may be implicated (6). Upon antigen activation, T cells interact physically with APCs and facilitate B cell proliferation and differentiation, ultimately leading to autoantibody production. It is widely accepted that both cellular and humoral immunities contribute to the GO pathogenesis (3, 7). Some studies have suggested the importance of Th1 cells in the early active phase and Th2 cells in the chronic fibrotic phase of GO (7). An increased number of Th17 cells have been detected at the site of orbital connective tissues (8, 9), leading to the concept of an imbalance in effector and regulatory T cells in GO autoimmunity. Fang et al. comprehensively summarized the current knowledge on T cell immunity in GO: Th1 (cytotoxic leaning), Th2 (antibody leaning) and an emerging role of Th17 (fibrotic leaning) cell subsets, in the context of key pathogenic processes such as adipogenesis and fibrosis. Hypothesizing that auto-immune responses can be locally triggered and aggravated, Lu et al. characterized the T cell repertoire infiltrating paranasal sinus mucosae in GO patients and reported increased pro-inflammatory effector T cells but reduced regulatory T cells.

Despite advances in understanding, the current treatment for GO is not satisfactory and long-term deformities and disabilities often persist. The problem is exacerbated by the fact that GO presentation can be heterogenous, as highlighted by an overview of asymmetric GO by Panagiotou and Perros, focusing on its clinical relevance and possible mechanisms. The Amsterdam declaration recommends that GO patients should be managed in joint endocrinology-ophthalmology clinics (10). An interesting study from Farag et al. described a ‘real world’ snapshot of the multidisciplinary management of a multi-ethnic GO population prior to the introduction of established GO standards.

Several existing therapies are currently used in the management of GO. In light of the inflammatory and immunological processes in operation, the first-line treatment is the administration of intravenous glucocorticoids (11). Naselli et al. reported that GO patients with high levels of low-density lipoproteins cholesterol were likely to respond poorly to glucocorticoids. In mild GO, selenium was shown to be beneficial, via its anti-oxidant and immunomodulatory effects, as reviewed by Lanzolla et al. The effects of anti-B cell therapies are somewhat controversial but indicate an effective role of rituximab for early active moderate-to-severe GO (12, 13). Campi et al. performed a post-hoc analysis on the efficacy of rituximab in GO patients, which can be helpful in the decision-making process. In addition, Zhang et al. described the salvaging benefits of rituximab in 2 patients refractory to glucocorticoids who subsequently underwent orbital decompression.

In order to develop novel effective target therapies for GO, it is essential to clarify the key pathological mechanisms, both immune and non-immune related, occurring within the orbit. The concept of circulating bone marrow derived CD34+ fibrocytes in the pathogenesis of GO is original and impactful. These TSHR-expressing progenitor cells migrate from the peripheral blood into orbital connective tissues and transit into CD34+ OFs that upon stimulation, undergo adipocytic and myofibroblastic differentiation (14). TSHR signaling in fibrocytes and OFs are partially dependent on the insulin-like growth factor 1 receptor (IGF-1R), another putative autoantigen that has received increasing attention in the past few years (15). Years of in vitro research were recently translated into a phase 2 and then a phase 3 randomized multicenter trials which consistently demonstrated that GO patients treated with teprotumumab, a fully human monoclonal IGF-1R inhibitor, were significantly more likely to experience a meaningful improvement in proptosis compared with patients treated with placebo (16, 17). Smith, one of the main researchers involved, reviewed the encouraging findings of teprotumumab as the first targeted therapy for GO. The cross-talk between TSHR and IGF-1R mediated by PKA/PI3K-FOXO signaling highlights a feasible therapeutic strategy to attenuate signaling initiated at either receptor, thereby relieving GO processes (4, 18).

Traditional immunosuppressive agents such as mycophenolate, azathioprine, and cyclosporin mainly inhibit the activation and proliferation of T cells (4). To date, no therapy targeting a particular T cell subset has been reported. Fortunately, blocking T cell related cytokines (e.g. IL-6) such as tocilizumab shows promising results in GO (19). Fallahi et al. from Antonelli’s group shared their thoughts on cytokine-based therapy in GD and GO.

Previous studies reported changes in T cell subsets during the transition from hyperthyroidism to euthyroidism and from active to inactive GO (20, 21). Thus, whilst GD and GO shared similar antibody-mediated immune attack, as highlighted by the improvement of these conditions in a patient with thyroid cancer treated with a TSHR-blocking monoclonal antibody (22), cell-mediated immunity is believed to play a central role in GO pathogenesis. A phase 1 multicenter trial revealed that ATX-GD-59, a mixture of two TSHR-derived peptides binding with HLA-DR on dendritic cells, improved free thyroid hormone levels in 70% (7/10 responders) of previously untreated mild to moderate Graves’ hyperthyroidism (23).

In vitro studies have facilitated identification of other novel treatments: Lanzolla et al. reviewed the use of antioxidant agents in mild GO; Wei et al. proposed simvastatin and ROCK inhibitors for orbital fibrosis whilst Lee et al. demonstrated potential for proprotein convertase as therapeutic target and biomarker.

The field of GO research has benefitted from the development of a robust TSHR-induced in vivo model of GO (24, 25). Comparison of the gut microbiota composition between mice housed in 2 centers revealed significant differences which may account for heterogeneity in the induced immune responses. When diseased and control mice were compared, disease-associated taxonomies were identified e.g. Firmicutes positively correlated with orbital adipogenesis (26). The same researchers modified the gut microbiota using antibiotic, probiotic and human fecal material transfer (hFMT). Experimental GO was exacerbated by hFMT and ameliorated by antibiotic treatment confirming a role for the gut microbiome in GD/GO (27). Several groups have investigated whether the same applies in human GD/GO as reviewed by Wang et al., who also provide a comprehensive summary of epigenetic and other factors implicated in the GO disease process.

In conclusion, this Research Topic provides encouraging updates made in understanding the complex interactions between genetic background and environmental factors such as the gut microbiome. It reviews the efficacies and short-comings of emerging therapies (e.g. teprotumumab and potential hearing problems) which have evolved from years of translational research to date. It illustrates an unlimited potential of non-surgical treatments, many of which target fundamental disease processes such as ligand-receptor binding and T cell subset imbalance underpinning GO development, and will ultimately benefit patient care in the near future.

Author Contributions

SF and HZ wrote the paper. ML, IM, and KC revised the paper. ML was the senior author of the paper. All authors contributed to the article and approved the submitted version.

Funding

This work was supported by the National Natural Science Foundation of China (81930024, 82071003, 82000879) and the Research Grant of the Shanghai Science and Technology Committee (20DZ2270800).

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s Note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Smith TJ, Hegedüs L. Graves’ Disease. N Engl J Med (2016) 375(16):1552–65. doi: 10.1056/NEJMra1510030

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Bahn RS. Graves’ Ophthalmopathy. N Engl J Med (2010) 362(8):726–38. doi: 10.1056/NEJMra0905750

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Davies TF, Andersen S, Latif R, Nagayama Y, Barbesino G, Brito M, et al. Graves’ Disease. Nat Rev Dis Primers (2020) 6(1):52. doi: 10.1038/s41572-020-0184-y

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Taylor PN, Zhang L, Lee RWJ, Muller I, Ezra DG, Dayan CM, et al. New Insights Into the Pathogenesis and Nonsurgical Management of Graves Orbitopathy. Nat Rev Endocrinol (2020) 16(2):104–16. doi: 10.1038/s41574-019-0305-4

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Feliciello A, Porcellini A, Ciullo I, Bonavolontà G, Avvedimento EV, Fenzi G. Expression of Thyrotropin-Receptor mRNA in Healthy and Graves’ Disease Retro-Orbital Tissue. Lancet (1993) 342(8867):337–8. doi: 10.1016/0140-6736(93)91475-2

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Draman MS, Grennan-Jones F, Taylor P, Muller I, Evans S, Haridas A, et al. Expression of Endogenous Putative TSH Binding Protein in Orbit. Curr Issues Mol Biol (2021) 43(3):1794–804. doi: 10.3390/cimb43030126

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Huang Y, Fang S, Li D, Zhou H, Li B, Fan X. The Involvement of T Cell Pathogenesis in Thyroid-Associated Ophthalmopathy. Eye (Lond) (2019) 33(2):176–82. doi: 10.1038/s41433-018-0279-9

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Fang S, Huang Y, Wang N, Zhang S, Zhong S, Li Y, et al. Insights Into Local Orbital Immunity: Evidence for the Involvement of the Th17 Cell Pathway in Thyroid-Associated Ophthalmopathy. J Clin Endocrinol Metab (2019) 104(5):1697–711. doi: 10.1210/jc.2018-01626

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Fang S, Zhang S, Huang Y, Wu Y, Lu Y, Zhong S, et al. Evidence for Associations Between Th1/Th17 “Hybrid” Phenotype and Altered Lipometabolism in Very Severe Graves Orbitopathy. J Clin Endocrinol Metab (2020) 105(6):1851–67. doi: 10.1210/clinem/dgaa124

CrossRef Full Text | Google Scholar

10. Perros P, Wiersinga WM. The Amsterdam Declaration on Graves’ Orbitopathy. Thyroid (2010) 20(3):245–6. doi: 10.1089/thy.2010.1618

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Bartalena L, Kahaly GJ, Baldeschi L, Dayan CM, Eckstein A, Marcocci C, et al. EUGOGO †. The 2021 European Group on Graves’ Orbitopathy (EUGOGO) Clinical Practice Guidelines for the Medical Management of Graves’ Orbitopathy. Eur J Endocrinol (2021) 185(4):G43–67. doi: 10.1530/EJE-21-0479

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Stan MN, Salvi M. MANAGEMENT OF ENDOCRINE DISEASE: Rituximab Therapy for Graves’ Orbitopathy - Lessons From Randomized Control Trials. Eur J Endocrinol (2017) 176(2):R101–9. doi: 10.1530/EJE-16-0552

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Salvi M, Covelli D. B Cells in Graves’ Orbitopathy: More Than Just a Source of Antibodies? Eye (Lond) (2019) 33(2):230–4. doi: 10.1038/s41433-018-0285-y

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Smith TJ. TSH-Receptor-Expressing Fibrocytes and Thyroid-Associated Ophthalmopathy. Nat Rev Endocrinol (2015) 11(3):171–81. doi: 10.1038/nrendo.2014.226

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Smith TJ, Janssen JAMJL. Insulin-Like Growth Factor-I Receptor and Thyroid-Associated Ophthalmopathy. Endocr Rev (2019) 40(1):236–67. doi: 10.1210/er.2018-00066

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Smith TJ, Kahaly GJ, Ezra DG, Fleming JC, Dailey RA, Tang RA, et al. Teprotumumab for Thyroid-Associated Ophthalmopathy. N Engl J Med (2017) 376(18):1748–61. doi: 10.1056/NEJMoa1614949

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Douglas RS, Kahaly GJ, Patel A, Sile S, Thompson EHZ, Perdok R, et al. Teprotumumab for the Treatment of Active Thyroid Eye Disease. N Engl J Med (2020) 382(4):341–52. doi: 10.1056/NEJMoa1910434

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Krieger CC, Neumann S, Gershengorn MC. TSH/IGF1 Receptor Crosstalk: Mechanism and Clinical Implications. Pharmacol Ther (2020) 209:107502. doi: 10.1016/j.pharmthera.2020.107502

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Perez-Moreiras JV, Gomez-Reino JJ, Maneiro JR, Perez-Pampin E, Lopez AR, Alvarez FMR, et al. Efficacy of Tocilizumab in Patients With Moderate-To-Severe Corticosteroid-Resistant Graves Orbitopathy: A Randomized Clinical Trial. Am J Ophthalmol (2018) 195:181–90. doi: 10.1016/j.ajo.2018.07.038

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Fallahi P, Ferrari SM, Ragusa F, Ruffilli I, Elia G, Paparo SR, et al. Th1 Chemokines in Autoimmune Endocrine Disorders. J Clin Endocrinol Metab (2020) 105(4):dgz289. doi: 10.1210/clinem/dgz289

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Hai YP, Lee ACH, Frommer L, Diana T, Kahaly GJ. Immunohistochemical Analysis of Human Orbital Tissue in Graves’ Orbitopathy. J Endocrinol Invest (2020) 43(2):123–37. doi: 10.1007/s40618-019-01116-4

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Ryder M, Wentworth M, Algeciras-Schimnich A, Morris JC, Garrity J, Sanders J, et al. Blocking the Thyrotropin Receptor With K1-70 in a Patient With Follicular Thyroid Cancer, Graves’ Disease, and Graves’ Ophthalmopathy. Thyroid (2021) 31(10):1597–602. doi: 10.1089/thy.2021.0053

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Pearce SHS, Dayan C, Wraith DC, Barrell K, Olive N, Jansson L, et al. Antigen-Specific Immunotherapy With Thyrotropin Receptor Peptides in Graves’ Hyperthyroidism: A Phase I Study. Thyroid (2019) 29(7):1003–11. doi: 10.1089/thy.2019.0036

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Banga JP, Moshkelgosha S, Berchner-Pfannschmidt U, Eckstein A. Modeling Graves’ Orbitopathy in Experimental Graves’ Disease. Horm Metab Res (2015) 47(10):797–803. doi: 10.1055/s-0035-1555956

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Berchner-Pfannschmidt U, Moshkelgosha S, Diaz-Cano S, Edelmann B, Görtz GE, Horstmann M, et al. Comparative Assessment of Female Mouse Model of Graves’ Orbitopathy Under Different Environments, Accompanied by Proinflammatory Cytokine and T-Cell Responses to Thyrotropin Hormone Receptor Antigen. Endocrinology (2016) 157(4):1673–82. doi: 10.1210/en.2015-1829

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Masetti G, Moshkelgosha S, Köhling HL, Covelli D, Banga JP, Berchner-Pfannschmidt U, et al. INDIGO Consortium. Gut Microbiota in Experimental Murine Model of Graves’ Orbitopathy Established in Different Environments may Modulate Clinical Presentation of Disease. Microbiome (2018) 6(1):97. doi: 10.1186/s40168-018-0478-4

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Moshkelgosha S, Verhasselt HL, Masetti G, Covelli D, Biscarini F, Horstmann M, et al. INDIGO Consortium. Modulating Gut Microbiota in a Mouse Model of Graves’ Orbitopathy and Its Impact on Induced Disease. Microbiome (2021) 9(1):45. doi: 10.1186/s40168-020-00952-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: graves orbitopathy, epidemiology, pathogenesis, evaluation, novel therapeutic approach

Citation: Zhou H, Muller I, Chong KK, Ludgate M and Fang S (2022) Editorial: Mechanisms and Novel Therapies in Graves’ Orbitopathy: Current Update. Front. Endocrinol. 13:902591. doi: 10.3389/fendo.2022.902591

Received: 23 March 2022; Accepted: 23 March 2022;
Published: 29 April 2022.

Edited and reviewed by:

Terry Francis Davies, Icahn School of Medicine at Mount Sinai, United States

Copyright © 2022 Zhou, Muller, Chong, Ludgate and Fang. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Huifang Zhou, fangzzfang@163.com; Sijie Fang, fangsijie89@hotmail.com

†This author share senior authorship

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.