415
Views
0
CrossRef citations to date
0
Altmetric
Editorial

Novel ways to attack inflammation in thyroid eye disease

&
Pages 277-281 | Published online: 09 Jan 2014

Graves’ disease (GD) is a systemic autoimmune disease affecting 1–2% of the US adult population Citation[1,2]. Over 90% of patients with GD present with goiter and clinical signs of hyperthyroidism while 20–50% develop thyroid eye disease (TED) Citation[3]. Nearly half a century ago, studies showed that the sera from patients with GD contained a thyroid-stimulating antibody directed against the thyrotropin receptor (TSHR) Citation[4–7]. These antibodies mimic the activity of thyroid-stimulating hormone (TSH), but have longer acting effects and are not subject to the physiologic hypothalamic–pituitary feedback loop. Despite recent advances in the genetic and molecular understanding of GD, the mechanisms behind inflammation patterns in seemingly unrelated anatomic regions as well as the self-limited nature of inflammation remain unclear.

Clinical course of TED

The orbital and periocular manifestations of GD are collectively termed TED. Most patients with TED experience mild self-limited ocular surface irritation, but 3–5% of patients have severe orbital congestion with pain, double vision or loss of vision Citation[8]. Rundle was the first to divide the course of TED into active (dynamic) and inactive (static) disease phases Citation[9]. The active phase of TED is characterized by progressive proptosis and diplopia, eyelid edema, conjunctival chemosis and injection, exposure keratitis, and even vision loss from compressive optic neuropathy. Usually within 18–24 months of the onset of active TED, inflammation resolves spontaneously and patients are left with stable proptosis, eyelid retraction and restrictive strabismus.

Current treatment of TED

Once the diagnosis of TED is made, patients who are hyperthyroid should be restored to euthyroid status and smokers are strongly encouraged to pursue smoking cessation. Both of these measures have been shown to increase response to treatment and hasten the resolution of TED Citation[10–12]. For most patients with mild TED, supportive measures such as ocular surface lubrication are sufficient Citation[13]. For more severe active TED, systemic glucocorticoids are considered, with the goal of decreasing orbital inflammation and minimizing inflammatory sequelae. Surgical intervention, in the form of staged orbital decompression, strabismus surgery and eyelid correction, are typically used for functional and cosmetic rehabilitation during inactive TED Citation[14–16]. When compressive optic neuropathy occurs, however, urgent orbital decompression may prevent vision loss. As we garner more knowledge about the molecular pathways involved in TED, we seek to devise targeted therapies directed against specific steps in the TED inflammatory pathway.

Immunology of TED

The role of T cells, B cells & cytokines

Immunohistochemical studies performed on orbital specimens from patients with active TED show intense infiltration of T lymphocytes, mast cells and occasional B cells between extraocular muscle fibers and within orbital fat, suggesting that connective tissues are the primary autoimmune targets Citation[17,18]. In the early stages of active TED, Th1 cells predominate and produce cytokines, IL-2, IFN-γ and TNF-α Citation[19,20]. A more robust Th2-dominated immune response appears in the later stages of inflammation, with the production of IL-4, IL-5 and IL-10 Citation[21]. B lymphocytes, which serve dual roles as immunoglobulin-secreting cells and antigen-presenting cells, produce IL-6, lymphotoxin, TNF-α and IL-10 Citation[22]. Macrophages, fibroblasts and adipocytes produce other inflammatory mediators, including IL-1α, IL-6 and TGF-β within the orbit Citation[23,24]. The interplay of these infiltrating immune cells with resident orbital fibroblasts is complex but is likely the lynchpin to understanding the site-specific disease process.

T-cell-targeted therapy

T-cell depletion using anti-CD3 monoclonal antibodies has been studied in vitro and in animal models of Type 1 diabetes Citation[25,26]. The use of anti-CD3 antibodies in the absence of costimulatory signals leads to CD4+ T-cell anergy and tolerance Citation[27,28]. Another T-cell-mediated pathway of interest in tumorigenesis and autoimmunity involves highly specialized T lymphocytes called regulatory T cells (Tregs). Tregs exhibit a CD4+CD25+Foxp3+ phenotype and when these cells are deficient, immunopathology can occur Citation[29]. Conversely, when Tregs are replete, autoimmune diseases are tempered Citation[30–32]. Anti-CD3 antibody depletion of pathogenic T cells increases the appearance of Tregs, thus it holds promise as a future treatment of TED by directly diminishing pathogenic T cells while indirectly enhancing beneficial regulatory T cells.

B-cell-targeted therapy

B cells are essential to the initiation of autoimmune thyroid disease Citation[33], and autoantibody generation is also dependent on the complex interplay between B and T cells. Thus, B-cell-depleting therapies and those that interrupt interactions between cognate molecules on B-cell surfaces offer great promise in the treatment of autoimmune disease. B-cell-targeted therapies used in autoimmune disease include: rituximab (RTK; anti-CD20), eprantuzumab (anti-CD22), DT2219 (anti-CD19 and CD22) and abatacept (anti-CTLA4), among others Citation[34]. Of these therapies, RTX, has recently garnered much attention in the TED literature. RTX is a humanized monoclonal antibody that binds the B-cell surface antigen CD20, attenuating CD20-dependent B-cell maturation. It was first used in 1997 to treat non-Hodgkin B-cell lymphomas Citation[35], and was subsequently used in IgM antibody-related polyneuropathies and rheumatoid arthritis Citation[36,37]. In 2006, two case reports showed significant reduction in the clinical activity in patients with TED unresponsive to steroids Citation[38,39]. A prospective, controlled study demonstrated sustained remission of hyperthyroidism in GD patients treated with RTX even though the drug failed to influence autoantibody levels Citation[40]. In another open, nonrandomized study of patients with TED, RTX was compared with intravenous glucocorticoid therapy Citation[41]. Patients receiving RTX demonstrated greater improvement of the clinical activity score with fewer side effects than those treated with glucocorticoids. Our group has demonstrated rapid and sustained resolution of orbital inflammation and optic neuropathy in patients with the most severe form of TED, who are resistant to corticosteroid treatment Citation[42]. Well-controlled, prospective and adequately powered studies remain essential to fully evaluate the role of RTX in TED.

Cytokine-targeted therapy

Disruption of the cytokine pathways in general holds substantial promise but a complete discussion is beyond the scope of this article. In particular, the TNF-α pathway has become a highly successful approach to treat rheumatoid arthritis and Crohn’s disease. In the synovium, TNF-α induces the production of proinflammatory cytokines, chemokines and cell adhesion molecules. Together, they mediate leukocyte chemoattraction, propagating inflammation. The presence of TNF-α in TED orbital tissues has previously been demonstrated Citation[43]. Three biological anti-TNF-α agents are currently in wide clinical use, including the monoclonal antibodies infliximab and adalimumab, and etanercept, a human recombinant soluble TNF-α receptor fusion protein. Two separate reports of infliximab use in patients with TED suggest reduced inflammation and improved visual function Citation[44,45]. Since TNF-α is present in the orbital tissue of TED patients and TNF-α inhibitors have been widely used in other autoimmune diseases, their use may merit consideration for clinical trials in TED.

Role of autoantigens

Research in recent decades suggests that multiple autoantigens may exist in TED, analogous to other autoimmune disorders. Most autoimmune diseases have several cycles of stimulation with different antigens before clinical disease becomes apparent. The role of autoantibodies to the TSHR is firmly established in the pathogenesis of hyperthyroidism in GD, but the evidence to explain the role of TSHR in TED is unclear. Several interesting correlations between TSHR antibody levels and disease activity have been reported, and TSHR mRNA has been detected in orbital tissues and orbital fibroblasts, but studies have been performed on limited sample sizes and do not explain pathogenesis Citation[46,47].

The IGF-1 receptor (IGF-1R)/IGF-1 pathway may be another autoantigen in TED. Orbital fibroblasts in patients with TED express elevated levels of IGF-1R Citation[48]. Treatment of these fibroblasts with either IGF-1 or pooled serum IgG from patients with GD stimulates the synthesis of powerful T-cell chemoattractants as well as the generation of hyaluronan Citation[49]. Importantly, neither IGF-1 nor GD-IgG elicited these responses in fibroblasts from individuals without autoimmune disease. These findings suggest that increased levels of IGF-1R may play a role in the pathogenesis of GD. Evidence that TSHR and IGF-1R might be functionally linked was strengthened recently when these proteins were found to colocalize Citation[50]. Several strategies for disrupting IGF-1R signaling have been developed recently and are currently being evaluated as therapy for cancer Citation[51].

Role of fibroblasts & fibrocytes

Several studies have demonstrated that orbital fibroblasts, especially those from patients with GD, are uniquely susceptible to several proinflammatory cytokines. Orbital fibroblasts fail to generate adequate levels of soluble IL-1 receptor antagonist, allowing for poorly opposed IL-1β signaling, which in turn causes increased expression of the inflammatory cyclooxygenase prostaglandin-endoperoxide H synthase (PGHS)-2 Citation[52]. Orbital fibroblasts also exhibit enhanced production of extracellular matrix components such as hyaluronan in response to these cytokines. Thus, orbital fibroblasts in GD produce proinflammatory molecules and components of connective tissue that lend themselves to the site-specific tissue remodeling occurring in TED.

Recently, our group described bone marrow-derived fibroblast-like cells called fibrocytes that are elevated in the circulation of GD patients. Fibrocytes have been shown to migrate to sites of injury and mediate site-specific inflammation and fibrosis in diseases including rheumatoid arthritis, pulmonary fibrosis and renal fibrosis Citation[53–55]. We found that these CD34+ fibrocytes selectively infiltrate orbital tissues of TED patients, suggesting a migratory role for these cells in the orbital process. Most surprisingly, we found a high expression of TSHR and IGF-1R on fibrocytes from patients with GD Citation[56]. The addition of TSH or antibodies to the TSHR to fibrocytes in culture demonstrated the production of proinflammatory cytokines including IL-6 and IL-8. Therapeutic targeting of fibrocytes or inhibiting the molecular signals of fibrocyte infiltration, such as CXCL12, may be critical for the prevention of fibrosis.

CD40 expression by T cells and fibroblasts may also play an important role in site-specific immune activation Citation[57]. CD40 binds CD40 ligand (also known as CD154) displayed on the surface of T lymphocytes, and fibroblasts provide T-cell costimulation that results in clonal expansion of naive T lymphocytes and enhances proinflammatory cytokine production, including that of IL-1, IL-6 and IL-8. Thus, disruption of the CD40–CD40 ligand interaction may represent an important therapeutic target in TED. Administration of therapeutic blocking antibodies against CD40 ligand has already proven effective in preclinical mouse models of diabetes and inflammatory bowel disease Citation[58,59].

Five-year view

Despite intensive study, the identity of the proximate antigenic target initiating TED, and the relationship between the orbital disease and the other components of GD, remain uncertain. Important insights concerning the pathogenesis of allied autoimmune diseases and increasing knowledge about their successful treatment should shed new light on the fundamental factors underlying TED and facilitate the development of therapies for this particularly vexing process. Unfortunately, studies of treatment modalities in TED are plagued by the inherently heterogeneous severity and natural history of TED. Meaningful clinical studies of treatment modalities will require a coordinated, multicenter effort to define disease severity and activity, similar to those proposed by the International Thyroid Eye Disease Society Citation[101].

Financial & competing interests disclosure

The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

No writing assistance was utilized in the production of this manuscript.

References

  • Weetman AP. Graves’ disease. N. Engl. J. Med.343, 1236–1248 (2000).
  • Brent GA. Clinical practice: Graves’ disease. N. Engl. J. Med.358, 2594–2605 (2008).
  • Bartley GB, Fatourechi V, Kadrmas EF et al. Long-term follow-up of Graves ophthalmopathy in an incidence cohort. Ophthalmology103, 958–962 (1996).
  • Adams DD, Purves HD. Abnormal responses in the assay of thyrotropins. Proc. Univ. Otago Med. Sch.34, 11–12 (1956).
  • Meek JC, Jones AE, Lewis UJ, Vanderlaan WP. Characterization of the long-acting thyroid stimulator of Graves’ disease. Proc. Natl Acad. Sci. USA52, 342–349 (1964).
  • Kriss JP, Pleshakov V, Chien JR. Isolation and identification of the long-acting thyroid stimulator and its relation to hyperthyroidism and circumscribed pretibial myxedema. J. Clin. Endocrinol. Metab.24, 1005–1028 (1964).
  • Pastan I, Roth J, Macchia V. Binding of hormone to tissue: the first step in polypeptide hormone action. Proc. Natl Acad. Sci. USA56, 1802–1809 (1966).
  • Wiersinga WM, Bartalena L. Epidemiology and prevention of Graves’ ophthalmopathy. Thyroid12, 855–860 (2002).
  • Rundle FF, Wilson CW. Development and course of exophthalmos and ophthalmoplegia in Graves’ disease with special reference to the effect of thyroidectomy. Clin. Sci.5, 177–194 (1945).
  • Thornton J, Kelly SP, Harrison RA, Edwards R. Cigarette smoking and thyroid eye disease: a systematic review. Eye21, 1135–1145 (2007).
  • Prummel MF, Wiersinga WM, Mourits MP, Koornneef L, Berghout A, Van Der Gaag R. Effect of abnormal thyroid function on the severity of Graves’ ophthalmopathy. Arch. Intern. Med.150, 1098–1101 (1990).
  • Tallstedt L, Lundell G, Blomgren H, Bring J. Does early administration of thyroxine reduce the development of Graves’ ophthalmopathy after radioiodine treatment? Eur. J. Endocrinol.130, 494–497 (1994).
  • Dickinson J, Perros P. Thyroid-associated orbitopathy: who and how to treat. Endocrinol. Metab. Clin. North Am.38, 373–388 (2009).
  • Schotthoefer EO, Wallace DK. Strabismus associated with thyroid eye disease. Curr. Opin. Ophthalmol.18, 361–365 (2007).
  • Ben Simon GJ, Mansury AM, Schwarcz RM, Lee S, McCann JD, Goldberg RA. Simultaneous orbital decompression and correction of upper eyelid retraction versus staged procedures in thyroid-related orbitopathy. Ophthalmology112, 923–932 (2005).
  • Kikkawa DO, Pornpanich K, Cruz RC, Levi L, Granet DB. Graded orbital decompression based on severity of proptosis. Ophthalmology109, 1219–1224 (2002).
  • Pappa A, Calder V, Aijan R. Analysis of extraocular muscle-infiltrating T cells in thyroid-associated ophthalmopathy (TAO). Clin. Exp. Immunol.109, 362–369 (1997).
  • Eckstein AK, Quadbeck B, Tews S et al. Thyroid associated ophthalmopathy: evidence for CD4(+) γδ T cells, de novo differentiation of RFD7(+) macrophages, but not of RFD1(+) dendritic cells, and loss of γδ and αβ T cell receptor expression. Br. J. Ophthalmol.88, 803–808 (2004).
  • Wakelkamp IM, Gerding MN, Van Der Meer JW, Prummel MF, Wiersinga WM. Both Th1- and Th2-derived cytokines in serum are elevated in Graves’ ophthalmopathy. Clin. Exp. Immunol.121, 453–457 (2000).
  • Yang D, Hiromatsu Y, Hoshino T, Inoue Y, Itoh K, Nonaka K. Dominant infiltration of T(h)1-type CD4+ T cells at the retrobulbar space of patients with thyroid-associated ophthalmopathy. Thyroid9, 305–310 (1999).
  • McLachlan SM, Prummel MF, Rapoport B. Cell-mediated or humoral immunity in Graves’ ophthalmopathy? Profiles of T-cell cytokines amplified by polymerase chain reaction from orbital tissue. J. Clin. Endocrinol. Metab.78, 1070–1074 (1994).
  • Paul W. Fundamental Immunology(Fifth Edition). Lippincott Williams and Wilkins, PA, USA (2003).
  • Kumar S, Bahn RS. Relative overexpression of macrophage-derived cytokines in orbital adipose tissue from patients with Graves’ ophthalmopathy. J. Clin. Endocrinol. Metab.88, 4246–4250 (2003).
  • Hiromatsu Y, Yang D, Bednarczuk T, Miyake I, Nonaka K, Inoue Y. Cytokine profiles in eye muscle tissue and orbital fat tissue from patients with thyroid-associated ophthalmopathy. J. Clin. Endocrinol. Metab.85, 1194–1199 (2000).
  • Masteller EL, Warner MR, Tang Q et al. Expansion of functional endogenous antigen-specific CD4+ CD25+ regulatory T cells from non-obese mice. J. Immunol.175, 3053–3059 (2005).
  • Bluestone JA. Regulatory T-cell therapy: is it ready for the clinic? Nat. Rev. Immunol.5, 343–349 (2005).
  • Kohm AP, Williams JS, Bickford AL et al. Treatment with nonmitogenic anti-CD3 monoclonal antibody induces CD4+ T cell unresponsiveness and functional reversal of established experimental autoimmune encephalomyelitis. J. Immunol.174, 4525–4534 (2005).
  • Earle KE, Tang Q, Zhou X et al.In vitro expanded human CD4+ CD25+ regulatory T cells suppress effector T cell proliferation. Clin. Immunol.115, 3–9 (2005).
  • Tang Q, Bluestone JA. Regulatory T-cell physiology and application to treat autoimmunity. Immunol. Rev.212, 217–237 (2006).
  • Chatenoud L, Bluestone JA. CD3-specific antibodies: a portal to the treatment of autoimmunity. Nat. Rev. Immunol.7, 622–632 (2007).
  • Scalapino KJ, Tang Q, Bluestone JA, Bonyhadi ML, Daikh DI. Suppression of disease in New Zealand Black/New Zealand White lupus-prone mice by adoptive transfer of ex vivo expanded regulatory T cells. J. Immunol.177, 1451–1459 (2006).
  • Sagoo P, Lombardi G, Lechler RI. Regulatory T cells as therapeutic cells. Curr. Opin. Organ Transplant.13, 645–653 (2008).
  • Pichurin P, Aliesky H, Chen CR, Nagayama Y, Rapoport B, McLachlan SM. Thyrotrophin receptor-specific memory T cell responses require normal B cells in a murine model of Graves’ disease. Clin. Exp. Immunol.134, 396–402 (2003).
  • Naik V, Khadavi N, Naik MN et al. Biologic therapeutics in thyroid-associated ophthalmopathy: translating disease mechanism into therapy. Thyroid18, 967–971 (2008).
  • Maloney DG, Grillo-Lopez AJ, White CA et al. IDEC-C3B8 (Rituximab) anti-CD20 monoclonal antibody therapy in patients with relapsed low-grade non-Hodgkin’s lymphoma. Blood90, 2188–2195 (1997).
  • Levine TD, Pestronk A. IgM antibody-related polyneuropathies: B-cell depletion chemotherapy using rituximab. Neurology52, 1701–1704 (1999).
  • Edwards JC, Cambridge G. Sustained improvement in rheumatoid arthritis following a protocol designed to deplete B lymphocytes. Rheumatology40, 205–211 (2001).
  • Salvi M, Vannucchi G, Campi I et al. Efficacy of rituximab treatment for thyroid-associated ophthalmopathy as a result of intraorbital B-cell depletion in one patient unresponsive to steroid immunosuppression. Eur. J. Endocrinol.154, 511–517 (2006).
  • El Fassi D, Nielsen CH, Hasselbach HC, Hegedus L. Treatment-resistant severe, active Graves’ ophthalmopathy successfully treated with B lymphocyte depletion. Thyroid16, 709–710 (2006).
  • El Fassi D, Nielsen CH, Bonnema SJ, Hasselbach HC, Hegedus L. B lymphocyte depletion with the monoclonal antibody rituximab in Graves’ disease: a controlled pilot study. J. Clin. Endocrinol. Metab.92, 1769–1772 (2007).
  • Salvi M, Vannucchi G, Campi I et al. Treatment of Graves’ disease and associated ophthalmopathy with the anti-CD20 monoclonal antibody rituximab: an open study. Eur. J. Endocrinol.156, 33–40 (2007).
  • Khanna D, Chong KK, Afifiyan NF et al. Increased generation of fibrocytes in thyroid-associated ophthalmopathy. J. Clin. Endocrinol. Metab.95, 430–438 (2010).
  • Heufelder AE, Bahn RS. Detection and localization of cytokine immunoreactivity in retro-ocular connective tissue in Graves’ ophthalmopathy. Eur. J. Clin. Invest.23, 10–17 (1993).
  • Durrani OM, Reuser TQ, Murray PI. Infliximab: a novel treatment for sight-threatening thyroid associated ophthalmopathy. Orbit24, 117–119 (2005).
  • Paridaens D, Van Den Bosch WA, Van Der Loos TL, Krenning EP, Van Hagen PM. The effect of etanercept on Graves’ ophthalmopathy: a pilot study. Eye19, 1286–1289 (2005).
  • Wakelkamp IM, Bakker O, Baldeschi L, Wiersinga WM, Prummel MF. TSH-R expression and cytokine profile in orbital tissue of active vs. inactive Graves’ ophthalmopathy patients. Clin. Endocrinol.58, 280–287 (2003).
  • Crisp MS, Lane C, Halliwell M, Wynford-Thomas D, Ludgate M. Thyrotropin receptor transcripts in human adipose tissue. J. Clin. Endocrinol. Metab.82, 2003–2005 (1997).
  • Smith TJ. The putative role of fibroblasts in the pathogenesis of Graves’ disease: evidence for the involvement of the insulin-like growth factor-1 receptor in fibroblast activation. Autoimmunity36, 409–415 (2003).
  • Pritchard J, Han R, Horst N, Cruikshank WW, Smith TJ. 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.170, 6348–6354 (2003).
  • Tsui S, Naik V, Hoa N 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.181, 4397–4405 (2008).
  • Hopkins A, Crowe PJ, Yang JL. Effect of type 1 insulin-like growth factor receptor targeted therapy on chemotherapy in human cancer and the mechanisms involved. J. Cancer Res. Clin. Oncol.136, 639–650 (2010).
  • Cao HJ, Han R, Smith TJ. Robust induction of PGHS-2 by IL-1 in orbital fibroblasts results from low levels of IL-1 receptor antagonist expression. Am. J. Physiol. Cell Physiol.284, 1429–1437 (2003).
  • Galligan CL, Siminovitch KA, Keystone EC, Bykerk V, Perez OD, Fish EN. Fibrocyte activation in rheumatoid arthritis. Rheumatology49, 640–651 (2010).
  • Strieter RM, Keeley EC, Hughes MA, Burdick MD, Mehrad B. The role of circulating mesenchymal progenitor cells (fibrocytes) in the pathogenesis of pulmonary fibrosis. J. Leukoc. Biol.86, 1111–1118 (2009).
  • Sakai N, Wada T, Matsushima K et al. The renin–angiotensin system contributes to renal fibrosis through regulation of fibrocytes. J. Hypertens.26, 780–790 (2008).
  • Douglas RS, Afifiyan NF, Hwang CJ et al. Increased generation of fibrocytes in thyroid-associated ophthalmopathy. J. Clin. Endocrinol. Metab.95, 430–438 (2010).
  • Hwang CJ, Afifiyan N, Sand D et al. Orbital fibroblasts from patients with thyroid-associated ophthalmopathy overexpress CD40: CD154 hyperinduces IL-6, IL-8, and MCP-1. Invest. Ophthalmol. Vis. Sci.50, 2262–2269 (2009).
  • Nanji SA, Hancock WW, Luo B et al. Costimulation blockade of both inducible costimulator and CD40 ligand induces dominant tolerance to islet allografts and prevents spontaneous autoimmune diabetes in the NOD mouse. Diabetes55, 27–33 (2006).
  • Liu Z, Geboes K, Colpaert S et al. Prevention of experimental colitis in SCID mice reconstituted with CD45RBhigh CD4+ T cells by blocking the CD40–CD154 interactions. J. Immunol.164, 6005–6014 (2000).

Website

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.