Next Article in Journal
The Role of Imaging Modalities in Estimating Myocardial Viability: A Narrative Review
Previous Article in Journal
The Role of Asprosin in Females in the Context of Fertility—An Exploratory Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Redefining Treatment Paradigms in Thyroid Eye Disease: Current and Future Therapeutic Strategies

by
Nicolò Ciarmatori
1,2,
Flavia Quaranta Leoni
3 and
Francesco M. Quaranta Leoni
1,4,5,*
1
Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
2
Department of Ophthalmology, Ospedali Privati Forlì “Villa Igea”, 47122 Forlì, Italy
3
Ophthalmology Unit, Department of Experimental Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy
4
Oftalmoplastica Roma, 00197 Rome, Italy
5
Orbital and Adnexal Service, Tiberia Hospital, GVM Care & Research, 00137 Rome, Italy
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2025, 14(15), 5528; https://doi.org/10.3390/jcm14155528
Submission received: 29 June 2025 / Revised: 29 July 2025 / Accepted: 31 July 2025 / Published: 6 August 2025
(This article belongs to the Section Ophthalmology)

Abstract

Background: Thyroid eye disease (TED) is a rare autoimmune orbital disorder predominantly associated with Graves’ disease. It is characterized by orbital inflammation, tissue remodeling, and potential visual morbidity. Conventional therapies, particularly systemic glucocorticoids, offer only partial symptomatic relief, failing to reverse chronic structural changes such as proptosis and diplopia, and are associated with substantial adverse effects. This review aims to synthesize recent developments in understandings of TED pathogenesis and to critically evaluate emerging therapeutic strategies. Methods: A systematic literature review was conducted using MEDLINE, Embase, and international clinical trial registries focusing on pivotal clinical trials and investigational therapies targeting core molecular pathways involved in TED. Results: Current evidence suggests that TED pathogenesis is primarily driven by the autoimmune activation of orbital fibroblasts (OFs) through thyrotropin receptor (TSH-R) and insulin-like growth factor-1 receptor (IGF-1R) signaling. Teprotumumab, a monoclonal IGF-1R inhibitor and the first therapy approved by the U.S. Food and Drug Administration for TED, has demonstrated substantial clinical benefit, including improvements in proptosis, diplopia, and quality of life. However, concerns remain regarding relapse rates and treatment-associated adverse events, particularly hearing impairment. Investigational therapies, including next-generation IGF-1R inhibitors, small-molecule antagonists, TSH-R inhibitors, neonatal Fc receptor (FcRn) blockers, cytokine-targeting agents, and gene-based interventions, are under development. These novel approaches aim to address both inflammatory and fibrotic components of TED. Conclusions: Teprotumumab has changed TED management but sustained control and toxicity reduction remain challenges. Future therapies should focus on targeted, mechanism-based, personalized approaches to improve long-term outcomes and patient quality of life.

1. Introduction

Thyroid eye disease (TED), also known as thyroid-associated orbitopathy (TAO) or Graves’ ophthalmopathy (GO), is a potentially sight-threatening autoimmune inflammatory disorder of the orbit, most commonly associated with Graves’ disease (GD). First described over a century ago, TED has evolved from a poorly understood complication of thyroid dysfunction to a distinct clinical entity with unique immunopathogenic mechanisms. It affects up to 40% of hyperthyroid patients and, although classically linked with hyperthyroidism, can also manifest in euthyroid or hypothyroid individuals [1,2].
With an estimated incidence of 5–20 per 100,000 person-years and a prevalence of approximately 0.25% in Western populations, TED is considered a rare disease [3]. Despite its rarity, it poses a significant socioeconomic burden as it often leads to significant physical disfigurement and psychosocial distress. Even patients with mild disease can experience substantial reductions in quality of life due to cosmetic concerns, chronic discomfort, and functional vision problems, leading to impaired daily functioning and productivity losses.
The disease typically follows a biphasic course, beginning with an active inflammatory phase lasting 1–3 years, characterized by signs such as eyelid retraction, chemosis, proptosis, and diplopia, and potentially progressing to dysthyroid optic neuropathy (DON), a sight-threatening complication. This is followed by a chronic, inactive phase during which residual fibrosis and tissue remodeling may persist.
Historically, TED management was primarily guided by clinical tools such as the Clinical Activity Score (CAS) and severity classifications like those from the European Group on Graves’ Orbitopathy (EUGOGO), which stratify disease as mild, moderate-to-severe, or sight-threatening. However, CAS remains limited by significant inter-observer variability, raising concerns about consistency in disease staging and treatment planning [4,5]. These limitations underscore the pressing need for more objective biomarkers and standardized assessment and treatment protocols.
Furthermore, traditional therapeutic approaches, including corticosteroids, orbital radiotherapy, and surgery, have been mostly supportive and symptomatic, limited by suboptimal efficacy in addressing chronic structural changes, and burdened by significant adverse effects. This highlights a critical unmet need for targeted mechanism-based therapeutic agents rather than symptom-based approaches.
Recently, the advent of targeted immunotherapies has represented a major paradigm shift in the management of TED, offering, for the first time, disease-modifying potential by directly interfering with the molecular pathways underlying inflammation and tissue remodeling. However, issues related to long-term results and relapse rates, safety profile, cost, and accessibility still limit their widespread adoption and remain under active investigation.
This review aims to provide an in-depth analysis of the evolving understanding of TED pathogenesis and to synthesize emerging evidence on the most recent therapeutic strategies. Analyzing these developments within the scope of established clinical frameworks underscores the persistent challenges and emerging opportunities in enhancing the quality of care for patients affected by this complex and debilitating condition.

2. Materials and Methods

A comprehensive literature search was conducted up to June 2025 using MEDLINE (via Ovid), Embase (via Ovid), and the US and EU Clinical Trials Registers, utilizing the following keywords: “Thyroid Eye Disease”, “Thyroid Orbitopathy”, “Graves’ Orbitopathy”, and “Graves’ Ophthalmopathy”. Boolean operators and MeSH terms were applied where appropriate.
The focus of the review was primarily on English language peer-reviewed articles, proof-of-concept studies, and publicly registered clinical trials addressing the pathophysiology of TED, pivotal clinical trials that have influenced current therapeutic strategies, and recent experimental and clinical investigations on novel therapies, including monoclonal antibodies and biologic agents. No restrictions concerning sex, age, follow-up time, setting, or number of participants were imposed.
Article selection was independently undertaken by the authors, involving title and abstract screening, followed by removal of duplicates and non-relevant publications. The reference lists of included articles were also manually reviewed to identify additional pertinent studies.

3. Results

3.1. Role of Glucocorticoids and Orbital Radiotherapy

Glucocorticoids (GCs) remain the first-line therapy for active TED in most European countries as per EUGOGO guidelines. However, the optimal treatment plan is still debated and protocols vary worldwide [6]. GCs modulate inflammation and immune responses via both direct and indirect genomic mechanisms, in addition to non-genomic pathways. This includes the induction of lipocortin synthesis, which subsequently inhibits leukotriene production, among other effects. Moreover, GCs mitigate the synthesis and release of cytokines, cell adhesion molecules, and glycosaminoglycans [7]. Consequently, GCs have been extensively utilized in the management of TED, although off-label, demonstrating the most substantial outcomes in immunosuppression when administered early during the active phase [8]. Intravenous methylprednisolone (IVMP) is favored over oral GCs for its higher bioavailability and faster onset [8].
However, recent evidence highlights the limited efficacy of GCs in addressing key clinical manifestations of TED. A meta-analysis reported a modest reduction in proptosis of 0.94 mm (95% CI: −1.57 to −0.32) at 12 weeks following IVMP therapy. Furthermore, a matching-adjusted indirect comparison found no statistically significant benefit of IVMP over a placebo in improving diplopia (odds ratio 2.69; 95% CI: 0.94–7.70) [9]. These findings underscore the limitations of GCs in targeting the fibrotic and structural changes underlying TED, leaving disfiguring symptoms such as proptosis and diplopia largely unresolved, with substantial implications for patients’ quality of life. While high-dose IVMP remains the mainstay in the management of DON, offering prompt visual improvement [10], its use is constrained by a significant adverse effect profile, including Cushingoid features, hypertension, hyperglycemia, psychiatric disturbances, and hepatotoxicity at cumulative doses exceeding 8 g [11].
Given the absence of robust, placebo-controlled trials supporting the usage of GCs, the necessity for novel pharmacological agents within the therapeutic arsenal for the treatment of TED is substantial and has consequently generated increasing interest in identifying new targets to disrupt the pathogenesis of TED.
Orbital radiotherapy (OR) is an established treatment for TED, primarily due to its anti-inflammatory effects and the radiosensitivity of infiltrating lymphocytes [12,13]. OR has demonstrated efficacy in the management of moderate-to-severe active TED, particularly in improving diplopia and ocular motility. A double-blind, sham-controlled RCT involving 60 patients confirmed the significant benefits of OR over sham treatment, while retrospective studies have reported superior outcomes when OR is combined with intravenous glucocorticoids compared to glucocorticoid monotherapy [14,15].
Longitudinal data further support the protective role of OR in disease progression: in a cohort of 351 patients with active TED, 17% of those treated with corticosteroids alone developed compressive optic neuropathy over 3.2 years, whereas no such cases were observed in patients receiving combined OR and corticosteroids, who also showed enhanced control of inflammation and ocular motility [16].
Combined with GCs in the active phase, OR reduces DON risk (0% versus 17%) [16]. Standard OR dosing is 20 Gy per orbit, administered as either 2 Gy over 10 days or 1 Gy over 20 days, with the latter showing better tolerability [17].
While generally well tolerated, OR may transiently exacerbate symptoms and is contraindicated in patients under 35 years of age or those with hypertensive or diabetic retinopathy due to potential long-term risks. Overall, OR remains an effective adjunctive therapy for TED, particularly during the active phase.

3.2. Role of Steroid-Sparing Agents

Nonsteroidal immunomodulatory agents have been explored as both adjunctive and standalone therapies in the treatment of moderate-to-severe active TED, with the goal of improving efficacy while minimizing the adverse effects associated with prolonged GC use.
Mycophenolate mofetil (MMF). MMF inhibits T and B cell proliferation through depletion of guanosine nucleotides. In a recent meta-analysis, MMF in combination with intravenous methylprednisolone (IVMP) demonstrated superior efficacy and tolerability compared to IVMP alone, with improvements in Clinical Activity Score (CAS) and reduced risk of adverse events [18]. The European Group on Graves’ Orbitopathy (EUGOGO) now recommends MMF plus IVMP as a first-line therapeutic option in moderate-to-severe active TED.
Cyclosporine. This calcineurin inhibitor suppresses IL-2 secretion and T cell activation. Several randomized controlled trials have shown that cyclosporine, when added to oral prednisolone, enhances therapeutic outcomes compared to prednisolone alone [6,19]. Based on these findings, EUGOGO recommends cyclosporine in combination with oral glucocorticoids as a second-line option for patients who fail to respond to IVGC monotherapy.
Methotrexate (MTX). MTX interferes with folate metabolism by inhibiting thymidylate synthase (TYMS) and dihydrofolate reductase (DHFR), thereby limiting the synthesis of nucleotides and the proliferation of inflammatory cells. Although robust RCTs are lacking, some studies have reported improvements in CAS; however, no significant effects on proptosis or diplopia have been demonstrated [20].
Azathioprine. Like MTX, azathioprine inhibits purine synthesis, impairing lymphocyte proliferation. Current evidence does not show significant long-term benefit over a placebo in TED, although it may play a role in reducing relapse rates following glucocorticoid tapering [21].
Sirolimus (rapamycin). Sirolimus is an mTOR pathway inhibitor with immunosuppressive and antiproliferative properties. Initial data were limited and often affected by prior interventions, but recent observational evidence suggests that sirolimus, when used in combination with IVMP, may offer superior outcomes in diplopia resolution compared to MMF from six months onward [22,23]. Its potential antifibrotic effect on extraocular muscles is of particular interest. The ongoing SIRGO trial is currently evaluating sirolimus as a first-line therapy for active TED [24].
Rituximab (RTX). RTX is a monoclonal antibody targeting CD20+ B cells. Early open-label and retrospective studies indicated reductions in CAS and TRAb levels, with some improvement in diplopia [25]. However, randomized trials have yielded inconsistent results, possibly due to heterogeneity in patient populations and disease stages. Consequently, RTX is generally reserved as a second-line treatment for corticosteroid-refractory cases.
Statins. Although primarily used for dyslipidemia, statins possess notable anti-inflammatory and immunomodulatory properties [26]. A recent systematic review reported that statin use was associated with reduced disease severity and slower progression of TED, suggesting a potential adjunctive role in its management [27].

3.3. Current Insights into the Pathogenesis of TED

TED’s multifactorial pathogenesis involves autoimmune activation of orbital fibroblasts (OFs) driving inflammation and extracellular matrix (ECM) remodeling. OFs exhibit phenotypic heterogeneity influenced by Thy-1 (CD90) expression: Thy-1+ OFs, in extraocular muscles, differentiate into myofibroblasts promoting fibrosis, while Thy-1 OFs, in connective tissue, act as preadipocytes promoting adipogenesis [28,29].
These subsets interact with immune cells, sustaining inflammation, ECM expansion, and fibrosis [30,31]. ECM remodeling persists into the chronic phase, challenging the classic biphasic model and underscoring the need for therapies addressing both inflammation and fibrosis [32].
Table 1 and Table 2 summarize current and emerging targeted therapies for TED.

3.4. Role of TSH-R and Its Inhibitors

The thyroid-stimulating hormone receptor (TSH-R), a G-protein-coupled receptor, is a key autoantigen in GD and TED. Normally expressed on thyroid cells, TSH-R is aberrantly found on orbital preadipocyte fibroblasts and myofibroblasts in TED, promoting hyaluronic acid (HA) production and fibroblast proliferation [33].
In murine models, human TSH-R transfection induced orbital adipose and connective tissue changes mimicking TED [34]. During the active phase of TED, TSH-R expression is notably higher, and activation by TSH, thyrotropin-receptor antibodies (TRAb), and interleukin-6 (IL-6) triggers downstream signaling cascades that drive the characteristic extracellular matrix expansion and cellular proliferation [35]. These findings support TSH-R as a promising therapeutic target in TED.

3.4.1. K1-70

K1-70 is a monoclonal antibody that inhibits TSH-R signaling. In a Phase I trial involving 18 stable TED patients, single intramuscular or intravenous doses led to complete CAS resolution and up to 8 mm proptosis reduction, with minimal adverse events (AEs) such as fatigue and diarrhea [36]. A Phase II Japanese randomized, double-blind, placebo-controlled trial is currently evaluating 50 mg and 100 mg doses in active TED to confirm efficacy and safety [37].

3.4.2. Small-Molecule TSH-R Antagonists

Small-molecule TSH-R antagonists are emerging as promising oral treatments for TED, offering favorable pharmacokinetics and potential cost benefits. These drugs act as allosteric inhibitors, meaning they interfere with TSH-R signaling inside the cell rather than blocking hormone binding at the receptor surface. For example, ANTAG3 reduced TRAb-induced cAMP and pAKT activity, as well as hyaluronic acid production, in OFs from TED patients [38].
Similarly, s37a inhibited cAMP buildup in TSH-R-expressing human embryonic kidney (HEK) cells stimulated with sera from TED patients [39]. In addition, TSH-R-targeted siRNA improved both clinical symptoms and biochemical markers in a mouse model of Graves’ disease [40].

3.5. Role of IGF-1R and Its Inhibitors

Insulin-like growth factor 1 receptor (IGF-1R), a receptor tyrosine kinase (RTK), is overexpressed and hyperfunctional in OFs and lymphocytes in TED, where it regulates fibroblast proliferation, adipogenesis, and lymphocyte activation via MAPK/ERK and PI3K/AKT pathways [41].
Although early studies suggested the existence of stimulating autoantibodies against IGF-1R similar to those targeting the TSH-R, their pathogenic relevance remains uncertain. IGF-1R antibody levels show no positive correlation with TED severity and may inversely associate with the CAS, indicating a potential protective role [42].
Current data support TSH-R/IGF-1R crosstalk as the primary driver of IGF-1R activation in TED [43]. The therapeutic relevance of this axis is underscored by the effectiveness of IGF-1R antagonists in suppressing TSH-R-induced hyaluronic acid (HA) production and cell proliferation and shows therapeutic potential in TED and oncology models by promoting apoptosis and reducing proptosis [44].

3.5.1. Teprotumumab

Teprotumumab (Tepezza), a fully human monoclonal antibody targeting IGF-1R, represents a significant advancement in TED therapy. Originally developed for oncology, it was repurposed after IGF-1R’s role in TED was identified [45]. Administered intravenously (10 mg/kg, then 20 mg/kg every 3 weeks for 8 doses) over 60–90 min, it showed efficacy in two pivotal RCTs: a Phase II trial by Smith et al. and the Phase III OPTIC trial, where 83% achieved ≥2 mm proptosis reduction compared to 10% on a placebo, with additional gains in diplopia and quality of life [46,47]. Following FDA approval in 2020, the 2022 OPTIC-X extension demonstrated sustained improvement in relapsing or non-responsive patients (89.2%) [48]. A subsequent RCT confirmed efficacy in chronic TED, leading to FDA indication extension to TED of any activity or duration [49].
Table 3 summarizes key clinical trials of teprotumumab in TED management.
Beyond regulatory milestones, further insights have emerged from imaging studies, real-world data, and subgroup analyses that deepen our understanding of teprotumumab’s mechanisms of action and clinical applications.
Imaging and real-world data confirmed orbital volume reductions [52,53] and DON improvement in up to 88% of cases [50], although relapse occurred in up to two-thirds within one year, suggesting some patients may require retreatment or combination with surgery [48,54,55]. Notably, a recent study found no significant association between the timing of post-teprotumumab orbital surgery and regression. However, delayed surgeries (≥180 days) were linked to greater disease activity and surgical burden [56].
Although teprotumumab has shown an acceptable safety profile, clinical trials and postmarketing surveillance highlighted an association with muscle spasms, hyperglycemia, inflammatory bowel disease, and hearing impairment, prompting routine audiometric evaluation [57].
Despite its transformative role in TED, the high cost and limited long-term data restrict its widespread use [58], although an upcoming Phase III trial of a subcutaneous formulation aims to improve convenience and adherence [59].
The 2022 ATA/ETA consensus recommends teprotumumab for patients with significant proptosis or diplopia, reflecting its established efficacy in moderate-to-severe active TED. In contrast, the EUGOGO guidelines currently consider teprotumumab a second-line option, citing the absence of head-to-head comparative data and the lack of European Medicines Agency (EMA) approval as of 2025 [4]. Teprotumumab is currently under regulatory review in Europe and Canada and has received approval in the United States, Australia, Brazil, Saudi Arabia, the United Arab Emirates, and Japan. The OPTIC-J trial has further confirmed its efficacy and safety in a Japanese cohort, supporting its potential for broader global implementation [51].
Overall, teprotumumab has significantly advanced TED management, particularly for proptosis and diplopia, though its optimal application requires careful patient selection, vigilant safety monitoring, and consideration of treatment cost and durability.

3.5.2. IBI311

IBI311, a recombinant anti-IGF-1R mAB under review by China’s NMPA, follows the teprotumumab dosing schedule. In the Phase III RESTORE-1 trial, 85.8% of Chinese TED patients achieved CAS improvement and ≥2 mm proptosis reduction at week 24 versus 3.8% with a placebo (p < 0.0001). Although trial data regarding AEs are pending, the safety profile was reported favorable [60].

3.5.3. Veligrotug (VRDN-001) and VRDN-003

Veligrotug (VRDN-001), a humanized anti-IGF-1R monoclonal antibody by Viridian Therapeutics, offers a shorter infusion schedule compared to teprotumumab, consisting of five 30-min intravenous infusions (10 mg/kg) every three weeks. Of the key clinical trials that are summarized in Table 4, two pivotal Phase III trials, THRIVE and THRIVE-2, have confirmed its efficacy and safety in active and chronic TED, respectively.
The THRIVE trial demonstrated that Veligrotug significantly improved outcomes in patients with active TED by week 15, with a proptosis responder rate (PRR) of 70% (64% placebo-adjusted). Diplopia resolved in 54% of treated patients (43% placebo-adjusted), and 64% achieved a CAS of 0 or 1 (46% placebo-adjusted). The safety profile was favorable, with no serious treatment-related adverse events; common side effects included muscle spasms (43%), headache (21%), and infusion reactions (17%). Hearing impairment occurred in 16% of treated patients versus 11% on a placebo (5.5% placebo-adjusted) [61].
In THRIVE-2, Veligrotug led to 2.34 mm proptosis reduction versus 0.46 mm with placebo, diplopia improvement in 56% versus 25%, and CAS ≤ 1 in 54% versus 24% (p < 0.01). Reported AEs were mild, including muscle spasms (36%), menstrual disorders (33%), and hearing impairment in 13% [62].
Veligrotug is currently under investigation in the STRIVE trial, a randomized, active-controlled study assessing efficacy, safety, and tolerability in TED patients irrespective of disease activity or duration [63]. Based on favorable Phase III outcomes, Viridian Therapeutics intends to submit a Biologics License Application (BLA) in the second half of 2025.
Furthermore, Viridian is advancing VRDN-003, a long-acting IGF-1R inhibitor intended for subcutaneous administration. Clinical regimens under evaluation include a 600 mg loading dose, followed by two 300 mg injections every eight weeks or five 300 mg injections every four weeks. VRDN-003 is currently being studied in the REVEAL-1 and REVEAL-2 Phase III trials for active and chronic TED, respectively, with results expected in the first half of 2026 [64].

3.5.4. Lonigutamab

Lonigutamab, a subcutaneous anti-IGF-1R monoclonal antibody by Acelyrin Inc., Agoura Hills, CA, USA demonstrated promising results in a Phase I/II trial for active TED. Two regimens, four 40 mg injections every 3 weeks versus a 50 mg loading dose followed by 25 mg weekly for 11 weeks, were well tolerated without ototoxicity. The 40 mg group showed a 50% proptosis response, a 25% improvement in diplopia, and consistent reductions in CAS, compared to no improvements in the placebo group. The weekly regimen yielded higher responses: 67% proptosis, 40% diplopia resolution, and 83% achieving ≥2-point CAS reduction, with slightly milder adverse events. These results support subcutaneous IGF-1R inhibition as a safe and effective TED therapy with sustained response and potential for extended dosing [65].

3.5.5. Linsitinib

Linsitinib is an oral dual inhibitor of IGF-1R and insulin receptor (IR) tyrosine kinase activity, disrupting PI3K/Akt and ERK signaling and TSH-R/IGF-1R crosstalk, which are central to TED pathogenesis. Preclinical murine models showed that Linsitinib inhibits OFs proliferation, hyaluronic acid secretion, and immune-driven tissue remodeling, while inducing apoptosis in IGF-1R/TSH-R-expressing cells [66,67].
In the Phase IIb LIDS trial involving 90 patients with active moderate-to-severe TED, linsitinib (150 mg twice daily) achieved a 52% proptosis responder rate at week 24, marking the first oral small molecule to demonstrate clinical efficacy in TED. Common adverse events included diarrhea, headache, nausea, fatigue, and elevated liver enzymes (~20), with no cases of hearing impairment or hyperglycemia reported [68].
A Phase III trial is planned for 2025 to evaluate long-term safety and efficacy in refractory or relapsing TED.

3.5.6. KRIYA-586

Kriya-586 is an investigational gene therapy by Kriya Therapeutics that delivers an IGF-1R-blocking antibody via a single peribulbar injection using an adeno-associated virus (AAV) vector. This one-time treatment allows sustained antibody expression, potentially eliminating the need for repeated dosing and significantly improving quality of life.
Preclinical in vivo studies have demonstrated effective suppression of IGF-1R-mediated signaling, with efficacy comparable to that of teprotumumab. First-in-human trials are expected to start in late 2025 [69].

3.6. Role of FcRn and Its Inhibitors

The neonatal Fc receptor (FcRn) extends the half-life of IgGs by protecting them from degradation in acidic endosomes and recycling them back to the bloodstream, a mechanism that also preserves pathogenic autoantibodies in autoimmune diseases [70].
FcRn inhibitors competitively bind to FcRn, promoting the lysosomal degradation of IgG and thereby selectively reducing their levels without affecting albumin, immune cells, cytokines, complement activity, or other immunoglobulins. This lowers the risk of infection compared to broad-spectrum immunosuppressants.
Targeting FcRn has recently emerged as a promising therapeutic approach for TED, with various drugs currently under evaluation for long-term safety and efficacy in TED, as summarized in Table 5.
Batoclimab (HBM9161), a human monoclonal antibody against FcRn, demonstrated a 64.8% reduction in serum IgG and a 56.7% decrease in anti-TSHR antibodies in the Phase IIa ASCEND-GO 1 trial involving patients with active moderate-to-severe TED, with predominantly mild, reversible adverse effects. However, the ASCEND-GO 2 trial was terminated early due to reversible hypercholesterolemia, raising safety concerns [71]. A Phase III trial is ongoing to further assess its efficacy and safety profile [72].
Efgartigimod, an engineered IgG1 Fc fragment with enhanced FcRn affinity, selectively reduces IgG while sparing IgM, IgA, and albumin. Originally approved by the FDA for myasthenia gravis, it has also shown benefits in other IgG-mediated diseases, including primary immune thrombocytopenia [75,76]. In TED, Efgartigimod is under investigation in the Phase III UplighTED trial, a randomized, double-masked, placebo-controlled study of patients with active moderate-to-severe disease, with a 2:1 randomization to drug or placebo, follow-up of up to 110 weeks, and an open-label extension. Preliminary data suggest a favorable safety profile, with mainly mild adverse events [73].
Viridian Therapeutics’ FcRn inhibitors, VRDN-006 and VRDN-008, have shown promising preclinical results. VRDN-006 demonstrated similar efficacy and safety to efgartigimod in non-human primates, without inducing hypercholesterolemia. Phase I human trials are planned for 2025. VRDN-008 offers an extended half-life and more durable IgG reduction compared to efgartigimod in murine models [74].

3.7. Role of Cytokines

Cytokines including IL-6, IL-11, IL-17, tumor necrosis factor alfa (TNFα), and transforming growth factor beta (TGF-β) play well-documented roles in the pathogenesis of TED [30,77].

3.7.1. IL-6

IL-6 is a key pro-inflammatory cytokine involved in chronic inflammation and autoimmune diseases, including TED. It signals through a hexameric complex composed of IL-6, IL-6R (both soluble and membrane-bound), and gp130, activating the JAK/STAT3, Ras/MAPK, and PI3K–Akt pathways to promote B and T cell responses [78,79].
Tocilizumab, a humanized monoclonal antibody targeting both soluble and membrane-bound IL-6R, prevents gp130 dimerization and downstream inflammatory signaling. Initially approved for rheumatoid arthritis, it has shown efficacy in corticosteroid-refractory TED in a small RCT (n = 32), where 86% of patients achieved CAS < 3, with a median proptosis reduction of 1.5 mm compared to no improvement in the placebo group [80]. Reported AEs included neutropenia and hypercholesterolemia. Observational studies and meta-analyses support its second-line use, though larger RCTs are needed [81,82]. The ongoing Phase II TOGO trial is comparing intravenous tocilizumab with methylprednisolone in moderate-to-severe TED [83].
Satralizumab is a subcutaneous anti-IL-6R IgG2 antibody with extended half-life and reduced dosing frequency. Initially approved for neuromyelitis optica spectrum disorder (NMOSD), it was developed through innovative antibody recycling technology, which extends its plasma half-life and facilitates subcutaneous administration. After being approved by the FDA in 2019 for the management of NMOSD, satralizumab is now being investigated in the context of TED. Currently, two global, Phase III, randomized, double-masked, placebo-controlled, multicenter studies, namely SatraGO-1 and SatraGO-2, are evaluating the drug’s safety and efficacy in adults with moderate-to-severe active and chronic inactive TED. In the initial six months of both studies, participants will receive subcutaneous injections every two weeks for the first three doses, followed by every four weeks for an additional five months (seven doses in total). The primary endpoint for these studies is the reduction in proptosis of at least 2 mm from baseline after the initial six months of treatment. This will be followed by a six-month follow-up period during which retreatment will be individualized based on proptosis response. The expected completion date for these studies is 2026 [84].

3.7.2. IL-11

IL-11 contributes to the development of fibrosis and inflammation in TED, and its parallel targeting in pulmonary fibrosis further underscores its potential as a therapeutic target in TED.
LASN01, a fully human monoclonal antibody targeting IL-11R, inhibits IL-11 signaling to reduce orbital fibrosis, inflammation, and remodeling, with preclinical models showing improvements in proptosis, CAS, and ocular motility. LASN01 is currently being evaluated intravenously in a Phase II trial, with CAS and proptosis reduction as primary endpoints [85].

3.7.3. IL-17

IL-17, mainly secreted by Th17 cells, promotes inflammation, fibrosis, and adipogenesis in TED via IL17R signaling in OFs. Elevated IL-17 levels correlate with higher CAS, and IGF-1R activity amplifies Th17-mediated orbital remodeling [86,87].
Vunakizumab (SHR-1314), a subcutaneously administered anti-IL-17A IgG1/κ antibody used in the treatment of psoriasis and ankylosing spondylitis, entered a TED trial but was discontinued early for commercial reasons [88].
Secukinumab, another anti-IL-17A IgG1κ monoclonal antibody, was evaluated in the Phase III ORBIT trial for the treatment of moderate-to-severe TED, which was halted early due to low efficacy despite acceptable safety [89].

4. Conclusions

TED is a debilitating, potentially sight-threatening autoimmune disorder that affects thousands annually. Historically, glucocorticoids have constituted the cornerstone of TED management and continue to serve as first-line therapy in many settings. While effective in mitigating orbital inflammation, glucocorticoids are limited in their capacity to address key manifestations such as proptosis and diplopia, and their long-term use is associated with considerable systemic toxicity. These limitations underscore the need for disease-modifying strategies that extend beyond symptomatic control.
Advances in the pathophysiological understanding of TED have catalyzed the development of precision-targeted therapies. Clinical trials and meta-analyses have shown that teprotumumab significantly reduces proptosis and diplopia, outperforming glucocorticoids in several domains, although direct head-to-head comparisons are still lacking [9,90]. EUGOGO management guidelines offer a structured, evidence-based framework for TED evaluation and treatment, and recommend a staged, activity- and severity-based approach, incorporating the CAS and risk stratification to guide therapeutic decision-making [6]. Although high-dose intravenous glucocorticoids remain the standard for moderate-to-severe active disease, EUGOGO acknowledges the shortcomings of conventional immunosuppression and endorses the incorporation of targeted therapies into the evolving treatment paradigm [91].
Beyond IGF-1R blockade, additional therapeutic targets are under investigation. TSH-R inhibitors aim to arrest autoimmune activation at its origin by blocking ligand–receptor interactions on orbital fibroblasts [36,37,38]. Cytokine inhibitors, particularly those targeting interleukins IL-6, IL-11, and IL-17, offer promise in modulating the inflammatory processes that drive TED. Elevated IL-6 levels correlate with disease activity, and IL-6 receptor blockade (e.g., tocilizumab) has demonstrated efficacy in reducing CAS, proptosis, and diplopia, particularly in glucocorticoid-refractory cases [82,92,93]. Emerging treatments such as statins and FcRn antagonists are also under active investigation. Statins, due to their immunomodulatory and anti-inflammatory properties, have been associated with decreased incidence and severity of TED in observational studies, likely through modulation of T-cell responses and fibroblast activity [94]. FcRn antagonists, by inhibiting IgG recycling, promote degradation of pathogenic thyroid-stimulating immunoglobulins, thereby reducing autoimmune stimulation [71,72,73].
Despite these advances, TED remains a clinically heterogeneous condition, characterized by variability in disease trajectory and therapeutic response. The future of TED management is likely to emphasize individualized, multimodal strategies. Integration of clinical, radiologic, serological, and molecular markers—such as TSH-R antibody titers, cytokine profiles, and fibroblast phenotypes—may enhance patient stratification and inform therapy selection, advancing the field toward a precision medicine framework.
As novel agents become available, combinatorial regimens—for example, concurrent IGF-1R and TSH-R inhibition or the addition of FcRn antagonists in relapsing cases—may enhance efficacy while mitigating toxicity. The development of long-acting and subcutaneous formulations (e.g., efgartigimod, lonigutamab) also holds potential for improving adherence and patient convenience. Nonetheless, high costs and limited global access to these biologics present substantial barriers. Ongoing cost-effectiveness analyses will be critical to inform reimbursement decisions and ensure equitable access. In parallel, the accumulation of real-world data and long-term safety profiles will be essential for broader clinical integration.
Clinicians should incorporate these novel therapies within established guidelines, including those from EUGOGO, ATA, and ETA. Targeted therapies may be particularly beneficial in patients with significant proptosis or recurrent disease, complementing traditional immunosuppression and surgical interventions. Multimodal approaches that combine immunomodulatory therapy with appropriately timed surgical management remain essential.
Surgical interventions—orbital decompression, strabismus correction, and eyelid procedures—continue to play a vital role in restoring function and appearance in the chronic or inactive phase. Their timing and extent should be guided by inflammatory activity and response to medical therapy [95]. Notably, early surgical planning in patients receiving agents such as teprotumumab may reduce fibrosis, procedural complexity, and enhance functional outcomes [96,97].
Ultimately, the evolution of TED management toward precision medicine will require not only therapeutic innovation but also standardized predictive tools and outcome metrics to guide treatment selection and monitor response. Addressing the economic and accessibility challenges in parallel with ongoing research will be crucial to optimizing long-term outcomes and quality of life in patients affected by this complex disorder.

Author Contributions

Conceptualization, F.M.Q.L.; methodology, N.C. and F.M.Q.L.; data curation, N.C., F.M.Q.L. and F.Q.L.; writing—original draft preparation, N.C., F.M.Q.L. and F.Q.L.; writing—review and editing, F.M.Q.L., N.C. and F.Q.L.; visualization, N.C.; supervision, F.M.Q.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The nature of the study means it did not require ethical approval.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

F.M.Q.L. serves as an advisor for Amgen; Argenx; Kriya Therapeutics; and Viridian Therapeutics, Inc. All other authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
TEDThyroid Eye Disease
TAOThyroid-Associated Orbitopathy
GOGraves’ Ophthalmopathy
GDGraves’ Disease
DONDysthyroid Optic Neuropathy
CASClinical Activity Score
GCGlucocorticoid
IVIntravenous
IVMPIntravenous Methylprednisolone
SCSubcutaneous
OROrbital Radiotherapy
MMFMycophenolate Mofetil
MTXMethotrexate
RTXRituximab
TRAbThyrotropin Receptor Antibodies
RCTRandomized Controlled Trial
ECMExtracellular Matrix
OFOrbital Fibroblast
THS-RThyroid-Stimulating Hormone Receptor
HAHyaluronic Acid
TSHThyroid-Stimulating Hormone
AEAdverse Event
HEKHuman Embryonic Kidney
IGF-1Insulin-like Growth Factor 1
IGF-1RInsulin-like Growth Factor 1 Receptor
RTKReceptor Tyrosine Kinase
FDAFood and Drug Administration
EMAEuropean Medicine Agency
ATAAmerican Thyroid Association
ETAEuropean Thyroid Association
EUGOGOEuropean Group on Graves’ Orbitopathy
mABMonoclonal Antibody
NMPANational Medical Products Administration
IRInsulin Receptor
FcRnNeonatal Fragment Crystallizable Receptor
TGF-ßTransforming Growth Factor Beta
PRRProptosis Responder Rate

References

  1. Smith, T.J.; Hegedüs, L. Graves’ Disease. N. Engl. J. Med. 2016, 375, 1552–1565. [Google Scholar] [CrossRef]
  2. Kahaly, G.J.; Grebe, S.K.G.; Lupo, M.A.; McDonald, N.; Sipos, J.A. Graves’ Disease: Diagnostic and Therapeutic Challenges (Multimedia Activity). Am. J. Med. 2011, 124, S2–S3. [Google Scholar] [CrossRef]
  3. Lazarus, J.H. Epidemiology of Graves’ orbitopathy (GO) and relationship with thyroid disease. Best. Pract. Res. Clin. Endocrinol. Metab. 2012, 26, 273–279. [Google Scholar] [CrossRef]
  4. Burch, H.B.; Perros, P.; Bednarczuk, T.; Cooper, D.S.; Dolman, P.J.; Leung, A.M.; Mombaerts, I.; Salvi, M.; Stan, M.N. Management of thyroid eye disease: A Consensus Statement by the American Thyroid Association and the European Thyroid Association. Eur. Thyroid. J. 2022, 11, e220189. [Google Scholar] [CrossRef]
  5. Kossler, A.L.; Stan, M.; Kazim, M.; Dosiou, C. Innovate Thyroid Eye Disease 2024 Multidisciplinary Symposium—April 26–28, 2024, Napa, California. Ophthalmic Plast. Reconstr. Surg. 2024, 40, 714. [Google Scholar] [CrossRef] [PubMed]
  6. Bartalena, L.; Kahaly, G.J.; Baldeschi, L.; Dayan, C.M.; Eckstein, A.; Marcocci, C.; Marinò, M.; Vaidya, B.; Wiersinga, W.M.; EUGOGO. The 2021 European Group on Graves’ orbitopathy (EUGOGO) clinical practice guidelines for the medical management of Graves’ orbitopathy. Eur. J. Endocrinol. 2021, 185, G43–G67. [Google Scholar] [CrossRef] [PubMed]
  7. Zang, S.; Ponto, K.A.; Kahaly, G.J. Intravenous Glucocorticoids for Graves’ Orbitopathy: Efficacy and Morbidity. J. Clin. Endocrinol. Metab. 2011, 96, 320–332. [Google Scholar] [CrossRef]
  8. Aktaran, S.; Akarsu, E.; Erbağci, I.; Araz, M.; Okumuş, S.; Kartal, M. Comparison of intravenous methylprednisolone therapy vs. oral methylprednisolone therapy in patients with Graves’ ophthalmopathy. Int. J. Clin. Pract. 2007, 61, 45–51. [Google Scholar] [CrossRef]
  9. Douglas, R.S.; Dailey, R.; Subramanian, P.S.; Barbesino, G.; Ugradar, S.; Batten, R.; Qadeer, R.A.; Cameron, C. Proptosis and Diplopia Response With Teprotumumab and Placebo vs the Recommended Treatment Regimen With Intravenous Methylprednisolone in Moderate to Severe Thyroid Eye Disease: A Meta-analysis and Matching-Adjusted Indirect Comparison. JAMA Ophthalmol. 2022, 140, 328–335. [Google Scholar] [CrossRef] [PubMed]
  10. Currò, N.; Covelli, D.; Vannucchi, G.; Campi, I.; Pirola, G.; Simonetta, S.; Dazzi, D.; Guastella, C.; Pignataro, L.; Beck-Peccoz, P.; et al. Therapeutic Outcomes of High-Dose Intravenous Steroids in the Treatment of Dysthyroid Optic Neuropathy. Thyroid® 2014, 24, 897–905. [Google Scholar] [CrossRef]
  11. Marinó, M.; Morabito, E.; Brunetto, M.R.; Bartalena, L.; Pinchera, A.; Marocci, C. Acute and Severe Liver Damage Associated with Intravenous Glucocorticoid Pulse Therapy in Patients with Graves’ Ophthalmopathy. Thyroid® 2004, 14, 403–406. [Google Scholar] [CrossRef]
  12. Tanda, M.L.; Bartalena, L. Efficacy and Safety of Orbital Radiotherapy for Graves’ Orbitopathy. J. Clin. Endocrinol. Metab. 2012, 97, 3857–3865. [Google Scholar] [CrossRef]
  13. Rajendram, R.; Taylor, P.N.; Wilson, V.J.; Harris, N.; Morris, O.C.; Tomlinson, M.; Yarrow, S.; Garrott, H.; Herbert, H.M.; Dick, A.D.; et al. Combined immunosuppression and radiotherapy in thyroid eye disease (CIRTED): A multicentre, 2×2 factorial, double-blind, randomised controlled trial. Lancet Diabetes Endocrinol. 2018, 6, 299–309. [Google Scholar] [CrossRef] [PubMed]
  14. Kim, J.W.; Han, S.H.; Son, B.J.; Rim, T.H.; Keum, K.C.; Yoon, J.S. Efficacy of combined orbital radiation and systemic steroids in the management of Graves’ orbitopathy. Graefe’s Arch. Clin. Exp. Ophthalmol. 2016, 254, 991–998. [Google Scholar] [CrossRef] [PubMed]
  15. Oeverhaus, M.; Witteler, T.; Lax, H.; Esser, J.; Führer, D.; Eckstein, A. Combination Therapy of Intravenous Steroids and Orbital Irradiation is More Effective Than Intravenous Steroids Alone in Patients with Graves’ Orbitopathy. Horm. Metab. Res. 2017, 49, 739–747. [Google Scholar] [CrossRef]
  16. Shams, P.N.; Ma, R.; Pickles, T.; Rootman, J.; Dolman, P.J. Reduced Risk of Compressive Optic Neuropathy Using Orbital Radiotherapy in Patients with Active Thyroid Eye Disease. Am. J. Ophthalmol. 2014, 157, 1299–1305. [Google Scholar] [CrossRef]
  17. Sobel, R.K.; Aakalu, V.K.; Vagefi, M.R.; Foster, J.A.; Tao, J.P.; Freitag, S.K.; Wladis, E.J.; McCulley, T.J.; Yen, M.T. Orbital Radiation for Thyroid Eye Disease: A Report by the American Academy of Ophthalmology. Ophthalmology 2022, 129, 450–455. [Google Scholar] [CrossRef] [PubMed]
  18. Kahaly, G.J.; Riedl, M.; König, J.; Pitz, S.; Ponto, K.; Diana, T.; Kampmann, E.; Kolbe, E.; Eckstein, A.; Moeller, L.C.; et al. Mycophenolate plus methylprednisolone versus methylprednisolone alone in active, moderate-to-severe Graves’ orbitopathy (MINGO): A randomised, observer-masked, multicentre trial. Lancet Diabetes Endocrinol. 2018, 6, 287–298. [Google Scholar] [CrossRef] [PubMed]
  19. Prummel, M.F.; Mourits, M.P.; Berghout, A.; Krenning, E.P.; van der Gaag, R.; Koornneef, L.; Wiersinga, W.M. Prednisone and Cyclosporine in the Treatment of Severe Graves’ Ophthalmopathy. N. Engl. J. Med. 1989, 321, 1353–1359. [Google Scholar] [CrossRef] [PubMed]
  20. Strianese, D.; Iuliano, A.; Ferrara, M.; Comune, C.; Baronissi, I.; Napolitano, P.; D’Alessandro, A.; Grassi, P.; Bonavolontà, G.; Bonavolontà, P.; et al. Methotrexate for the Treatment of Thyroid Eye Disease. J. Ophthalmol. 2014, 2014, 128903. [Google Scholar] [CrossRef]
  21. Taylor, P.; Rajendram, R.; Hanna, S.; Wilson, V.; Pell, J.; Li, C.; Cook, A.; Gattamaneni, R.; Plowman, N.; Jackson, S.; et al. Factors Predicting Long-term Outcome and the Need for Surgery in Graves Orbitopathy: Extended Follow-up from the CIRTED Trial. J. Clin. Endocrinol. Metab. 2023, 108, 2615–2625. [Google Scholar] [CrossRef]
  22. Lanzolla, G.; Maglionico, M.N.; Comi, S.; Menconi, F.; Piaggi, P.; Posarelli, C.; Figus, M.; Marcocci, C.; Marinò, M. Sirolimus as a second-line treatment for Graves’ orbitopathy. J. Endocrinol. Invest. 2022, 45, 2171–2180. [Google Scholar] [CrossRef]
  23. Lai, K.K.H.; Wang, Y.; Pang, C.P.; Chong, K.K. Sirolimus versus mycophenolate mofetil for triple immunosuppression in thyroid eye disease patients with recent-onset intractable diplopia: A prospective comparative case series. Clin. Exp. Ophthalmol. 2023, 51, 878–881. [Google Scholar] [CrossRef]
  24. Marinò, M. Sirolimus for Graves’ Orbitopathy (SIRGO). 2022. Available online: https://clinicaltrials.gov/study/NCT04598815 (accessed on 13 June 2025).
  25. Kang, S.; Hamed Azzam, S.; Minakaran, N.; Ezra, D.G. Rituximab for thyroid-associated ophthalmopathy. Cochrane Database Syst. Rev. 2022, 2022, CD009226. [Google Scholar] [CrossRef] [PubMed]
  26. Lund University. Treatment of Graves’ Ophthalmopathy with Simvastatin (GO-S). 2017. Available online: https://clinicaltrials.gov/study/NCT03131726 (accessed on 13 June 2025).
  27. Malboosbaf, R.; Maghsoomi, Z.; Emami, Z.; Khamseh, M.E.; Azizi, F. Statins and thyroid eye disease (TED): A systematic review. Endocrine 2024, 85, 11–17. [Google Scholar] [CrossRef]
  28. Shu, X.; Shao, Y.; Chen, Y.; Zeng, C.; Huang, X.; Wei, R. Immune checkpoints: New insights into the pathogenesis of thyroid eye disease. Front. Immunol. 2024, 15, 1392956. [Google Scholar] [CrossRef]
  29. Smith, T.J. New advances in understanding thyroid-associated ophthalmopathy and the potential role for insulin-like growth factor-I receptor. F1000Research 2018, 7, 134. [Google Scholar] [CrossRef]
  30. Łacheta, D.; Miśkiewicz, P.; Głuszko, A.; Nowicka, G.; Struga, M.; Kantor, I.; Poślednik, K.B.; Mirza, S.; Szczepański, M.J. Immunological Aspects of Graves’ Ophthalmopathy. BioMed Res. Int. 2019, 2019, 7453260. [Google Scholar] [CrossRef] [PubMed]
  31. Basak, M.; Sanyal, T.; Kar, A.; Bhattacharjee, P.; Das, M.; Chowdhury, S. Peripheral blood mononuclear cells—Can they provide a clue to the pathogenesis of Graves’ Orbitopathy? Endocrine 2022, 75, 447–455. [Google Scholar] [CrossRef] [PubMed]
  32. Lehmann, G.M.; Feldon, S.E.; Smith, T.J.; Phipps, R.P. Immune Mechanisms in Thyroid Eye Disease. Thyroid® 2008, 18, 959–965. [Google Scholar] [CrossRef]
  33. Smith, T.J.; Janssen, J.A.M.J.L. Insulin-like Growth Factor-I Receptor and Thyroid-Associated Ophthalmopathy. Endocr. Rev. 2019, 40, 236–267. [Google Scholar] [CrossRef] [PubMed]
  34. Xia, N.; Ye, X.; Hu, X.; Song, S.; Xu, H.; Niu, M.; Wang, H.; Wang, J. Simultaneous induction of Graves’ hyperthyroidism and Graves’ ophthalmopathy by TSHR genetic immunization in BALB/c mice. PLoS ONE 2017, 12, e0174260. [Google Scholar] [CrossRef]
  35. Woeller, C.F.; Roztocil, E.; Hammond, C.; Feldon, S.E. TSHR Signaling Stimulates Proliferation Through PI3K/Akt and Induction of miR-146a and miR-155 in Thyroid Eye Disease Orbital Fibroblasts. Investig. Ophthalmol. Vis. Sci. 2019, 60, 4336–4345. [Google Scholar] [CrossRef]
  36. Furmaniak, J.; Sanders, J.; Sanders, P.; Li, Y.; Rees Smith, B. TSH receptor specific monoclonal autoantibody K1-70TM targeting of the TSH receptor in subjects with Graves’ disease and Graves’ orbitopathy—Results from a phase I clinical trial. Clin. Endocrinol. 2022, 96, 878–887. [Google Scholar] [CrossRef]
  37. KIKUCHIT A Phase II Randomized, Double-Blind, Placebo-Controlled, Parallel-Group, Multicenter Study to Evaluate K1-70 in Japanese Patients with Active Thyroid Eye Disease. 2024. Available online: https://rctportal.mhlw.go.jp/en/detail?trial_id=jRCT2031240330 (accessed on 13 June 2025).
  38. Turcu, A.F.; Kumar, S.; Neumann, S.; Coenen, M.; Iyer, S.; Chiriboga, P.; Gershengorn, M.C.; Bahn, R.S. A Small Molecule Antagonist Inhibits Thyrotropin Receptor Antibody-Induced Orbital Fibroblast Functions Involved in the Pathogenesis of Graves Ophthalmopathy. J. Clin. Endocrinol. Metab. 2013, 98, 2153–2159. [Google Scholar] [CrossRef] [PubMed]
  39. Marcinkowski, P.; Hoyer, I.; Specker, E.; Furkert, J.; Rutz, C.; Neuenschwander, M.; Sobottka, S.; Sun, H.; Nazare, M.; Berchner-Pfannschmidt, U.; et al. A New Highly Thyrotropin Receptor-Selective Small-Molecule Antagonist with Potential for the Treatment of Graves’ Orbitopathy. Thyroid 2019, 29, 111–123. [Google Scholar] [CrossRef]
  40. Wang, X.; Liu, W.; Rui, Z.; Zheng, W.; Tan, J.; Li, N.; Yu, Y. Immunotherapy with a biologically active ICAM-1 mAb and an siRNA targeting TSHR in a BALB/c mouse model of Graves’ disease. Endokrynol. Pol. 2021, 72, 592–600. [Google Scholar] [CrossRef]
  41. Truong, T.; Silkiss, R.Z. The Role of Insulin-like Growth Factor-1 and Its Receptor in the Eye: A Review and Implications for IGF-1R Inhibition. Ophthalmic Plast. Reconstr. Surg. 2023, 39, 4. [Google Scholar] [CrossRef]
  42. Marinò, M.; Rotondo Dottore, G.; Ionni, I.; Lanzolla, G.; Sabini, E.; Ricci, D.; Sframeli, A.; Mazzi, B.; Menconi, F.; Latrofa, F.; et al. Serum antibodies against the insulin-like growth factor-1 receptor (IGF-1R) in Graves’ disease and Graves’ orbitopathy. J. Endocrinol. Investig. 2019, 42, 471–480. [Google Scholar] [CrossRef]
  43. Krieger, C.C.; Boutin, A.; Neumann, S.; Gershengorn, M.C. Proximity ligation assay to study TSH receptor homodimerization and crosstalk with IGF-1 receptors in human thyroid cells. Front. Endocrinol. 2022, 13, 989626. [Google Scholar] [CrossRef] [PubMed]
  44. Morshed, S.A.; Ma, R.; Latif, R.; Davies, T.F. Mechanisms in Graves Eye Disease: Apoptosis as the End Point of Insulin-Like Growth Factor 1 Receptor Inhibition. Thyroid® 2022, 32, 429–439. [Google Scholar] [CrossRef]
  45. Pappo, A.S.; Vassal, G.; Crowley, J.J.; Bolejack, V.; Hogendoorn, P.C.; Chugh, R.; Ladanyi, M.; Grippo, J.F.; Dall, G.; Staddon, A.P.; et al. A phase 2 trial of R1507, a monoclonal antibody to the insulin-like growth factor-1 receptor (IGF-1R), in patients with recurrent or refractory rhabdomyosarcoma, osteosarcoma, synovial sarcoma, and other soft tissue sarcomas: Results of a Sarcoma Alliance for Research Through Collaboration study. Cancer 2014, 120, 2448–2456. [Google Scholar] [CrossRef]
  46. Smith, T.J.; Kahaly, G.J.; Ezra, D.G.; Fleming, J.C.; Dailey, R.A.; Tang, R.A.; Harris, G.J.; Antonelli, A.; Salvi, M.; Goldberg, R.A.; et al. Teprotumumab for Thyroid-Associated Ophthalmopathy. N. Engl. J. Med. 2017, 376, 1748–1761. [Google Scholar] [CrossRef]
  47. Douglas, R.S.; Kahaly, G.J.; Patel, A.; Sile, S.; Thompson, E.H.Z.; Perdok, R.; Fleming, J.C.; Fowler, B.T.; Marcocci, C.; Marinò, M.; et al. Teprotumumab for the Treatment of Active Thyroid Eye Disease. N. Engl. J. Med. 2020, 382, 341–352. [Google Scholar] [CrossRef]
  48. Douglas, R.S.; Kahaly, G.J.; Ugradar, S.; Elflein, H.; Ponto, K.A.; Fowler, B.T.; Dailey, R.; Harris, G.J.; Schiffman, J.; Tang, R.; et al. Teprotumumab Efficacy, Safety, and Durability in Longer-Duration Thyroid Eye Disease and Re-treatment: OPTIC-X Study. Ophthalmology 2022, 129, 438–449. [Google Scholar] [CrossRef] [PubMed]
  49. Douglas, R.S.; Couch, S.; Wester, S.T.; Fowler, B.T.; Liu, C.Y.; Subramanian, P.S.; Tang, R.; Nguyen, Q.T.; Maamari, R.N.; Ugradar, S.; et al. Efficacy and Safety of Teprotumumab in Patients with Thyroid Eye Disease of Long Duration and Low Disease Activity. J. Clin. Endocrinol. Metabolism. 2024, 109, 25–35. [Google Scholar] [CrossRef]
  50. Sears, C.M.; Wang, Y.; Bailey, L.A.; Turbin, R.; Subramanian, P.S.; Douglas, R.; Cockerham, K.; Kossler, A.L. Early efficacy of teprotumumab for the treatment of dysthyroid optic neuropathy: A multicenter study. Am. J. Ophthalmol. Case Rep. 2021, 23, 101111. [Google Scholar] [CrossRef]
  51. Hiromatsu, Y.; Ishikawa, E.; Kozaki, A.; Takahashi, Y.; Tanabe, M.; Hayashi, K.; Imagawa, Y.; Kaneda, K.; Mimura, M.; Dai, X.; et al. A randomised, double-masked, placebo-controlled trial evaluating the efficacy and safety of teprotumumab for active thyroid eye disease in Japanese patients. Lancet Reg. Health West. Pac. 2025, 55, 101464. [Google Scholar] [CrossRef]
  52. Ugradar, S.; Parunakian, E.; Zimmerman, E.; Malkhasyan, E.; Raika, P.; Douglas, R.N.; Kossler, A.L.; Douglas, R.S. Clinical and Radiologic Predictors of Response to Teprotumumab: A 3D Volumetric Analysis of 35 Patients. Ophthalmic Plast. Reconstr. Surg. 2024, 41, 408–414. [Google Scholar] [CrossRef] [PubMed]
  53. Jain, A.P.; Gellada, N.; Ugradar, S.; Kumar, A.; Kahaly, G.; Douglas, R. Teprotumumab reduces extraocular muscle and orbital fat volume in thyroid eye disease. Br. J. Ophthalmol. 2022, 106, 165–171. [Google Scholar] [CrossRef] [PubMed]
  54. Hwang, C.J.; Rebollo, N.P.; Mechels, K.B.; Perry, J.D. Reactivation After Teprotumumab Treatment for Active Thyroid Eye Disease. Am. J. Ophthalmol. 2024, 263, 152–159. [Google Scholar] [CrossRef] [PubMed]
  55. Ugradar, S.; Parunakian, E.; Malkhasyan, E.; Chiou, C.A.; Walsh, H.L.; Tolentino, J.; Wester, S.T.; Freitag, S.K.; Douglas, R.S. The Rate of Re-treatment in Patients Treated with Teprotumumab: A Multicenter Study of 119 Patients with 1 Year of Follow-up. Ophthalmology 2025, 132, 92–97. [Google Scholar] [CrossRef]
  56. Walsh, H.L.; Clauss, K.D.; Meyer, B.I.; Parunakian, E.; Yasar, C.; Chiou, C.A.; Johnson, T.E.; Ugradar, S.; Kossler, A.L.; Freitag, S.K.; et al. Surgical Timing for Patients with Thyroid Eye Disease Treated With Teprotumumab: A Collaborative Multicenter Study. Ophthalmic Plast. Reconstr. Surg. 2025, 41, 320. [Google Scholar] [CrossRef] [PubMed]
  57. Huang, J.; Su, A.; Yang, J.; Zhuang, W.; Li, Z., Sr. Postmarketing Safety Concerns of Teprotumumab: A Real-World Pharmacovigilance Assessment. J. Clin. Endocrinol. Metab. 2025, 110, 159–165. [Google Scholar] [CrossRef] [PubMed]
  58. Brito, J.P.; Nagy, E.V.; Singh Ospina, N.; Zˇarković, M.; Dosiou, C.; Fichter, N.; Lucarelli, M.J.; Hegedüs, L. A Survey on the Management of Thyroid Eye Disease Among American and European Thyroid Association Members. Thyroid® 2022, 32, 1535–1546. [Google Scholar] [CrossRef] [PubMed]
  59. Amgen. A Phase 3, Randomized, Double-Masked, Placebo-Controlled, Parallel-Group, Multicenter Trial to Evaluate the Efficacy, Safety and Tolerability of Subcutaneous Teprotumumab in Participants with Moderate-to-Severe Active Thyroid Eye Disease. 2025. Available online: https://clinicaltrials.gov/study/NCT06248619 (accessed on 13 June 2025).
  60. Innovent Biologics. Innovent Announces Primary Endpoint Met in the Phase 3 Clinical Trial (RESTORE-1) of IBI311 (Anti-IGF-1R Antibody) in Treating Thyroid Eye Disease and Plans to Submit NDA to the NMPA. 2024. Available online: https://www.prnewswire.com/news-releases/innovent-announces-primary-endpoint-met-in-the-phase-3-clinical-trial-restore-1-of-ibi311-anti-igf-1r-antibody-in-treating-thyroid-eye-disease-and-plans-to-submit-nda-to-the-nmpa-302065096.html (accessed on 13 June 2025).
  61. Viridian Therapeutics, Inc. THRIVE Readout Data. 2024. Available online: https://s203.q4cdn.com/807813999/files/doc_presentations/2024/Sep/THRIVE-readout-data-deck-FINAL.pdf (accessed on 13 June 2025).
  62. Rodien, P.; Schittkowski, M.; Konuk, O.; Eckstein, A.; Csutak, A.; Eade, E.; Uddin, J.; Lee, V.; Sales Sanz, M.; Conroy, W.; et al. THRIVE-2 phase 3 trial of Veligrotug (VRDN-001) in chronic thyroid eye disease (TED): Efficacy and safety at 15 weeks. Endocr. Abstr. 2025, 110, 29. [Google Scholar] [CrossRef]
  63. Viridian Therapeutics, Inc. A Randomized, Controlled, Safety and Tolerability Study of VRDN-001, a Humanized Monoclonal Antibody Directed Against the IGF-1 Receptor, in Participants with Thyroid Eye Disease (TED). 2025. Available online: https://clinicaltrials.gov/study/NCT06384547 (accessed on 13 June 2025).
  64. Newell, R.; Zhao, Y.; Bedian, V. Design and Preclinical Characterization of VRDN-003, a Next-Generation, Half-life Extended Antibody to IGF-1 Receptor in Development for Thyroid Eye Disease (TED). Investig. Ophthalmol. Vis. Sci. 2023, 64, 4044. [Google Scholar]
  65. Ugradar, S.; Kostick, D.A.; Spadaro, J.; Grover, A.; Imm, S.; Chesler, S.; Mpofu, S.; Khong, J. Preliminary Safety and Efficacy of Subcutaneous Lonigutamab (anti-IGF-1R) from a Phase 1/2 Proof of Concept Study in Patients with Thyroid Eye Disease. J. Endocr. Soc. 2024, 8, bvae163.2040. [Google Scholar] [CrossRef]
  66. Boutin, A.; Eliseeva, E.; Templin, S.; Marcus-Samuels, B.; Anderson, D.E.; Gershengorn, M.C.; Neumann, S. Linsitinib Decreases Thyrotropin-Induced Thyroid Hormone Synthesis by Inhibiting Crosstalk Between Thyroid-Stimulating Hormone and Insulin-Like Growth Factor 1 Receptors in Human Thyrocytes In Vitro and In Vivo in Mice. Thyroid® 2025, 35, 216–224. [Google Scholar] [CrossRef]
  67. Luffy, M.; Ganz, A.L.; Wagner, S.; Wolf, J.; Ropertz, J.; Zeidan, R.; Kent, J.D.; Douglas, R.S.; Kahaly, G.J. Linsitinib inhibits proliferation and induces apoptosis of both IGF-1R and TSH-R expressing cells. Front. Immunol. 2024, 15, 1488220. [Google Scholar] [CrossRef]
  68. Sling Therapeutics. Sling Therapeutics Announces Positive Topline Results from Phase 2b/3 LIDS Clinical Trial of Oral Small Molecule Linsitinib in Patients with Thyroid Eye Disease. 2025. Available online: https://live-vasaragen.pantheonsite.io/2025/01/14/sling-therapeutics-announces-positive-topline-results-from-phase-2b-3-lids-clinical-trial-of-oral-small-molecule-linsitinib-in-patients-with-thyroid-eye-disease/ (accessed on 13 June 2025).
  69. Kriya Therapeutics. Kriya Highlights Positive Preclinical Data for KRIYA-586, a Gene Therapy Product Candidate for Thyroid Eye Disease (TED), at ESOPRS Annual Meeting. 2024. Available online: https://kriyatherapeutics.com/news/kriya-highlights-positive-preclinical-data-for-kriya-586-a-gene-therapy-product-candidate-for-thyroid-eye-disease-ted-at-esoprs-annual-meeting/ (accessed on 13 June 2025).
  70. Remlinger, J.; Madarasz, A.; Guse, K.; Hoepner, R.; Bagnoud, M.; Meli, I.; Feil, M.; Abegg, M.; Linington, C.; Shock, A.; et al. Antineonatal Fc Receptor Antibody Treatment Ameliorates MOG-IgG–Associated Experimental Autoimmune Encephalomyelitis. Neurol. Neuroimmunol. Neuroinflamm. 2022, 9, e1134. [Google Scholar] [CrossRef]
  71. Kahaly, G.J.; Dolman, P.J.; Wolf, J.; Giers, B.C.; Elflein, H.M.; Jain, A.P.; Srinivasan, A.; Hadjiiski, L.; Jordan, D.; Bradley, E.A.; et al. Proof-of-concept and Randomized, Placebo-controlled Trials of an FcRn Inhibitor, Batoclimab, for Thyroid Eye Disease. J. Clin. Endocrinol. Metab. 2023, 108, 3122–3134. [Google Scholar] [CrossRef]
  72. Immunovant Sciences GmbH. A Phase 3, Multi-Center, Randomized, Quadruple-Masked, Placebo-Controlled Study of Batoclimab for the Treatment of Participants with Active Thyroid Eye Disease (TED). 2025. Available online: https://clinicaltrials.gov/study/NCT05517421 (accessed on 13 June 2025).
  73. Argenx. A Phase 3, Randomized, Double-Masked, Placebo-Controlled, Multicenter Study to Evaluate the Efficacy, Safety, Tolerability, Pharmacokinetics, Pharmacodynamics, and Immunogenicity of Efgartigimod PH20 SC Administered by Prefilled Syringe in Adult Participants with Thyroid Eye Disease. 2025. Available online: https://clinicaltrials.gov/study/NCT06307626 (accessed on 12 June 2025).
  74. Viridian Therapeutics, Inc. VRDN Q1 2024 Corporate Presentation. 2024. Available online: https://s203.q4cdn.com/807813999/files/doc_events/2024/May/08/VRDN-Q1-2024-Deck-May-13-Final.pdf (accessed on 13 June 2025).
  75. Heo, Y.-A. Efgartigimod: First Approval. Drugs 2022, 82, 341–348. [Google Scholar] [CrossRef]
  76. Broome, C. Efgartigimod alfa for the treatment of primary immune thrombocytopenia. Ther. Adv. Hematol. 2023, 14, 20406207231172831. [Google Scholar] [CrossRef] [PubMed]
  77. Gupta, V.; Hammond, C.L.; Roztocil, E.; Gonzalez, M.O.; Feldon, S.E.; Woeller, C.F. Thinking inside the box: Current insights into targeting orbital tissue remodeling and inflammation in thyroid eye disease. Surv. Ophthalmol. 2022, 67, 858–874. [Google Scholar] [CrossRef] [PubMed]
  78. Kaur, S.; Bansal, Y.; Kumar, R.; Bansal, G. A panoramic review of IL-6: Structure, pathophysiological roles and inhibitors. Bioorganic Med. Chem. 2020, 28, 115327. [Google Scholar] [CrossRef] [PubMed]
  79. Kulbay, M.; Tanya, S.M.; Tuli, N.; Dahoud, J.; Dahoud, A.; Alsaleh, F.; Arthurs, B.; El-Hadad, C. A Comprehensive Review of Thyroid Eye Disease Pathogenesis: From Immune Dysregulations to Novel Diagnostic and Therapeutic Approaches. Int. J. Mol. Sci. 2024, 25, 11628. [Google Scholar] [CrossRef]
  80. Perez-Moreiras, J.V.; Gomez-Reino, J.J.; Maneiro, J.R.; Perez-Pampin, E.; Romo Lopez, A.; Rodríguez Alvarez, F.M.; Castillo Laguarta, J.M.; Del Estad Cabello, A.; Gessa Sorroche, M.; España Gregori, E.; et al. Efficacy of Tocilizumab in Patients with Moderate-to-Severe Corticosteroid-Resistant Graves Orbitopathy: A Randomized Clinical Trial. Am. J. Ophthalmol. 2018, 195, 181–190. [Google Scholar] [CrossRef]
  81. Sánchez-Bilbao, L.; Martínez-López, D.; Revenga, M.; López-Vázquez, Á.; Valls-Pascual, E.; Atienza-Mateo, B.; Valls-Espinosa, B.; Maiz-Alonso, O.; Blanco, A.; Torre-Salaberri, I. Anti-IL-6 Receptor Tocilizumab in Refractory Graves’ Orbitopathy: National Multicenter Observational Study of 48 Patients. J. Clin. Med. 2020, 9, 2816. [Google Scholar] [CrossRef]
  82. Duarte, A.F.; Xavier, N.F.; Sales Sanz, M.; Cruz, A.A.V. Efficiency and Safety of Tocilizumab for the Treatment of Thyroid Eye Disease: A Systematic Review. Ophthalmic Plast. Reconstr. Surg. 2024, 40, 367. [Google Scholar] [CrossRef]
  83. Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico. Multicenter, Randomized, Observer-Blind, Controlled Study of the Anti-IL-6 Receptor Antibody Tocilizumab (TCZ) or Methylprednisolone (MP) Treatment in Patients with Active Moderate-Severe Graves’ Orbitopathy. 2022. Available online: https://clinicaltrials.gov/study/NCT04876534 (accessed on 13 June 2025).
  84. Ezra, D. Interleukin-6 (IL-6) Receptor Signalling Inhibition with Satralizumab in Thyroid Eye Disease (TED): Phase 3 SatraGO-1 and SatraGO-2 Trial Design. 2024. Available online: https://medically.roche.com/global/en/ophthalmology/esoprs-2024/medical-material/ESOPRS-2024-presentation-ezra-interleukin-6-IL-6-receptor-pdf.html (accessed on 13 June 2025).
  85. Lassen Therapeutics, Inc. A Study to Evaluate the Efficacy and Safety of LASN01 in Patients with Thyroid Eye Disease. 2025. Available online: https://clinicaltrials.gov/study/NCT06226545?intr=NCT06226545&rank=1 (accessed on 13 June 2025).
  86. Fang, S.; Huang, Y.; Wang, S.; Zhang, Y.; Luo, X.; Liu, L.; Zhong, S.; Liu, X.; Li, D.; Liang, R. IL-17A Exacerbates Fibrosis by Promoting the Proinflammatory and Profibrotic Function of Orbital Fibroblasts in TAO. J. Clin. Endocrinol. Metab. 2016, 101, 2955–2965. [Google Scholar] [CrossRef]
  87. Kim, S.E.; Yoon, J.S.; Kim, K.H.; Lee, S.Y.; Kim, S.E.; Yoon, J.S.; Kim, K.H.; Lee, S.Y. Increased serum interleukin-17 in Graves’ ophthalmopathy. Graefes Arch. Clin. Exp. Ophthalmol. 2012, 250, 1521–1526. [Google Scholar] [CrossRef]
  88. Suzhou Suncadia Biopharmaceuticals Co. Ltd. A Single-Arm, Open-Label, Prospective Phase II Study to Evaluate the Efficacy and Safety of SHR-1314 by Subcutaneous Injection in Active Moderate to Severe Graves’ Orbitopathy Patients. 2024. Available online: https://clinicaltrials.gov/study/NCT05394857 (accessed on 13 June 2025).
  89. Novartis Pharmaceuticals. A Two-Year Multi-Center Phase 3 Study to Investigate the Efficacy and Safety of Secukinumab in Adult Patients with Active, Moderate to Severe Thyroid Eye Disease (ORBIT), with a Randomized, Parallel-Group, Double-Blind, Placebo-Controlled, 16-Week Treatment Period, and a Follow-Up/Retreatment Period. 2025. Available online: https://clinicaltrials.gov/study/NCT04737330 (accessed on 13 June 2025).
  90. Lin, F.; Yao, Q.; Yu, B.; Deng, Z.; Qiu, J.; He, R. The Efficacy and Safety of Teprotumumab in Thyroid Eye Disease: Evidence from Randomized Controlled Trials. Int. J. Clin. Pract. 2023, 2023, 6638089. [Google Scholar] [CrossRef]
  91. Moledina, M.; Damato, E.M.; Lee, V. The changing landscape of thyroid eye disease: Current clinical advances and future outlook. Eye 2024, 38, 1425–1437. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  92. Farde, K.; Träisk, F. Tocilizumab—A disease-modulating treatment for thyroid associated ophthalmopathy? Orbit 2025, 44, 415–419. [Google Scholar] [CrossRef] [PubMed]
  93. Murdock, J.; Nguyen, J.; Hurtgen, B.J.; Andorfer, C.; Walsh, J.; Lin, A.; Tubbs, C.; Erickson, K.; Cockerham, K. The role of IL-6 in thyroid eye disease: An update on emerging treatments. Front. Ophthalmol. 2025, 5, 1544436. [Google Scholar] [CrossRef]
  94. Lanzolla, G.; Comi, S.; Cosentino, G.; Pakdel, F.; Marinò, M. Statins in Graves Orbitopathy: A New Therapeutic Tool. Ophthalmic Plast. Reconstr. Surg. 2023, 39, S29–S39. [Google Scholar] [CrossRef]
  95. Silkiss, R.Z.; Kim, S.; Bilyk, J.R. Treatment of corticosteroid-resistant thyroid eye disease. Can. J. Ophthalmol. 2021, 56, 312–318. [Google Scholar] [CrossRef]
  96. Dallalzadeh, L.O.; Villatoro, G.A.; Chen, L.; Sim, M.S.; Movaghar, M.; Robbins, S.L.; Karlin, J.N.; Khitri, M.R.; Velez, F.G.; Korn, B.S.; et al. Teprotumumab for Thyroid Eye Disease-related Strabismus. Ophthalmic Plast. Reconstr. Surg. 2024, 40, 434–439. [Google Scholar] [CrossRef] [PubMed]
  97. Topilow, N.J.; Penteado, R.C.; Ting, M.; Al-Sharif, E.; Villatoro, G.A.; Yoon, J.S.; Liu, C.Y.; Korn, B.S.; Kikkawa, D.O. Orbital decompression following treatment with teprotumumab for thyroid eye disease. Can. J. Ophthalmol. 2025, 60, e59–e64. [Google Scholar] [CrossRef] [PubMed]
Table 1. Current and emerging IGF-1R-targeted therapies for TED.
Table 1. Current and emerging IGF-1R-targeted therapies for TED.
DrugDeveloperDrug
Type
Initially
Developed for
Cohort and
Clinical Trial
Phase in TED
Dosing RegimenMost
Effective on
Side Effects
in TED
Teprotumumab
(TEPEZZA®,
2020 FDA
Approval)
Horizon/
Amgen, Thousand Oaks, CA, USA
Fully human
mAb
Solid cancerActive and Chronic TED
(FDA approved/Phase IV)
Eight Q3W IV inj.
(1st: 10 mg/kg,
then 20 mg/kg)
CAS
Proptosis Diplopia
Hearing Impairment, Hyperglycemia,
Muscle Spasms, Nausea/Diarrhea, Teratogenicity
IBI311Innovent, Mountain View, CA, USARecombinant
mAb
TEDActive TED
(Phase IIb)
Eight Q3W IV inj.
(1st: 10 mg/kg,
then 20 mg/kg)
CAS
Proptosis
Not Yet Reported, Expected Similar to Teprotumumab
Veligrotug (VRDN-001)Viridian
Therapeutics, Waltham, MA, USA
Humanized
mAb
TEDActive and Chronic TED (Phase III)Five Q3W IV inj.
(10 mg/kg)
CAS
Proptosis Diplopia
Muscle Spasms,
Headache,
Mild Hearing Issues
VRDN-003Viridian
Therapeutics, Waltham, MA, USA
Next-gen,
half-life extended
Humanized
mAb
TEDActive and Chronic TED (Phase III)Loading dose (600 mg) +
Two Q8W or Five Q4W
SC inj. (300 mg)
Not yet
reported
Not Reported,
Expected Similar to Veligrotug
LonigutamabValenzaBIO, Bethesda, MD, USA/Acelyrin Inc., Agoura Hills, CA, USAHumanized mAbTEDActive TED
(Phase Ib)
Four Q3W (40 mg)
or
Twelve QW SC inj.
(1st 50 mg, then 25 mg)
CAS
Proptosis Diplopia
Muscle Spasms,
Headache,
No Hearing Impairment
LinsitinibSling
Therapeutics, Ann Arbor, MI, USA
Small moleculeCancerActive TED
(Phase III)
75 mg or 150 mg
tablets BID for 24 weeks
ProptosisDiarrhea, Headache, Nausea, Fatigue, Elevated Liver Enzymes
Kriya-586Kriya
Therapeutics, Morrisville, NC, USA
AAV Gene
Therapy
TEDNot specified
(Phase I)
Single peribulbar
injection
Not yet reportedNot Yet Reported
Table 2. Other current and emerging targeted therapies for TED.
Table 2. Other current and emerging targeted therapies for TED.
TargetDrugDeveloperInitially
Developed for
Cohort and Clinical
Trial Phase in TED
Dosing RegimenMost
Effective on
Side Effects
in TED
TSH-RK1-70RSR Ltd., Lancashire, UKThyroid CancerActive TED
(Phase I)
Single dose 25 mg IM inj
or
Single dose 50/150 mg IV inj
CAS
Proptosis
Fatigue,
Diarrhea
FcRnBatoclimabImmunovant, New York, NY, USAMGActive TED
(Phase III)
Six QW SC inj.
(2 × 680 mg + 4 × 340 mg)
Lower TRAB CASHypercholesterolemia, Hypoalbuminemia, Headache
EfgartigimodArgenx, Amsterdam, The NetherlandsMGActive TED
(Phase III)
Twentyfour QW SC inj.
(1 g prefilled syringe)
Not yet
reported
Not Yet
Reported
IL6RTocilizumabHoffmann-
La Roche, Basel, Switzerland
Rheumatoid ArthritisActive TED
(Phase II)
Four Q4W IV inj.
(8 mg/kg)
CAS
Proptosis (low)
Mild AEs, Neutropenia, Hypercholesterolemia
SatralizumabHoffmann-
La Roche
NMOSDActive and Chronic TED
(Phase III)
Three Q2W + Five Q4W SC inj. (120 mg)Not yet
reported
Not Yet
Reported
PacibekitugTourmaline Bio, Inc., New York, NY, USAASCVDActive TED
(Phase IIb)
Three Q8W SC inj.
(20 mg or 50 mg)
Not yet
reported
Not Yet
Reported
IL11RLASN01Lassen
Therapeutics, Inc., San Diego, CA, USA
Pulmonary
fibrosis
Active TED
(Phase II)
Four Q4W IV inj.
(multiple ascending dose)
Not yet
reported
Not Yet
Reported
IL17RVunakizumabSuzhou Suncadia
Biopharmaceuticals Co., Ltd., Beijing, China
Anti-rheumatic
and Psoriasis
Active TED
(Phase II—
terminated early)
SC inj.
(Details not reported)
Not
reported
Not Reported
SecukinumabNovartis, Basel, SwitzerlandAnti-rheumatic
and Psoriasis
Active TED
(Phase III—
terminated early)
Four QW inj. + Two Q4W SC inj. (300 mg each)Primary endpoint not metNot Reported
Table 3. Key clinical trials of Teprotumumab.
Table 3. Key clinical trials of Teprotumumab.
StudyStudy
Population
Follow-Up DurationPrimary EndpointKey FindingsMain
Adverse
Events
Smith et al., 2017 [46]RCT on
moderate-to-severe active TED;
average CAS ≥ 4 and
disease duration < 9 months
24 weeksProptosis
reduction
≥ 2 mm
Proptosis response: 71% vs. 20%
CAS 0–1: 59% vs. 21%
Hyperglycemia in
Diabetic Patients
Douglas et al., 2020
(OPTIC) [47]
RCT on
moderate-to-severe active TED;
average CAS ≥ 4 and
disease duration 6.3 months
24 weeksProptosis
reduction
≥ 2 mm
Proptosis response: 83% vs. 10%
CAS 0–1: 59% vs. 21%
Diplopia response: 68% vs. 29%
GO-QOL Score Improvement:
13.79 vs. 4.43
Muscle Spasms (25%) Nausea (17%)
Alopecia (13%)
Diarrhea (13%)
Hearing Impairment (10%)
Hyperglycemia (8%)
Sears et al., 2021 [50]Retrospective observational
case series
15 weeksUnspecifiedBCVA improvement: 0.87 logMAR
Proptosis reduction: 4.7 mm
CAS improvement: 5.25 points
Diplopia improvement: 0.75 points
RAPD resolution/improvement: 100%
Color normalization/improvement: 100%
Not Reported
Douglas et al., 2022
(OPTIC-X) [48]
Open-label extension study on non-responders or relapsed from OPTIC48 weeksProptosis
reduction
≥ 2 mm
Proptosis response: 89%
CAS 0–1: 65.6%
Diplopia response: 63%
GO-QOL Score Improvement: 11.7
Proptosis recurrence at 6 m: 26%
Similar Profile
to OPTIC
Douglas et al., 2024 [49]RCT on
inactive/Chronic TED;
average CAS ≤ 1 and
disease duration 2–10 years
24 weeksProptosis
Reduction
≥ 2 mm
Proptosis response: 61.9% vs. 25%
Diplopia Response not significant
(study not powered enough)
Muscle Spasms (41.5%) Hearing Impairment (22%) Hyperglycemia (14.6%)
Hiromatsu et al., 2025 (OPTIC-J) [51]RCT on
Japanese patients with
moderate-to-severe active TED;
average CAS ≥ 3 and
disease duration < 9 months
24 weeksProptosis
reduction
≥ 2 mm
Proptosis improvement: 89% vs. 11%
CAS 0–1: 59% vs. 22%
Diplopia improvement: 64% vs. 45% (not significant)
Hyperglycemia (22%)
Hearing Impairment (15%)
Table 4. Key clinical trials of Veligrotug.
Table 4. Key clinical trials of Veligrotug.
StudyStudy PopulationFollow-Up DurationPrimary EndpointKey FindingsMain Adverse
Events
THRIVE [61]
(2024)
RCT on
moderate-to-severe
active TED;
average CAS ≥ 4 and
disease duration < 8 months
15 weeksProptosis reduction
≥ 2 mm
Proptosis response:
70% vs. 5%

CAS 0–1:
64% vs. 18%

Diplopia response:
63% vs. 20%

Diplopia complete
resolution:
54% vs. 12%
Muscle spasms (43%)

Headache
(21%)

Hearing
impairment
(16%)

Hyperglycemia (15%)
THRIVE-2 [62]
(2024)
RCT on
chronic TED;
average CAS < 3 and
disease duration > 5 years
15 weeks for
primary endpoint,
extended up to 52 weeks
Proptosis reduction
≥ 2 mm
Proptosis response: 56% vs. 8%

CAS 0–1:
54% vs. 24%

Diplopia response:
56% vs. 25%

Diplopia complete
resolution:
32% vs. 14%
Muscle spasms (6%)

Menstrual
disorders
(33%)

Headache
(13%)

Hearing
impairment
(13%)

Diarrhea
(10%)

Hyperglycemia (5%)
STRIVE
(ongoing) [63]
RCT on
TED of any
severity or duration
52 weeksIncidence of treatment-emergent
adverse events
(TEAEs)
Expected late 2025Not yet
reported
Table 5. Anti-FcRn drugs in clinical development for TED.
Table 5. Anti-FcRn drugs in clinical development for TED.
DrugStudyPopulation
Tested
Follow-Up
Duration
Treatment
Regimen
Primary
Endpoint
Key FindingsMain
Adverse
Events
Batoclimab (HBM9161)ASCEND-GO 1 (Phase IIa) [71]RCT on
moderate-to-severe
active TED
6 weeks5 QW SC injections
(680 mg 1st and 2nd, then 340 mg)
Changes in serum levels of anti-TSH-R Abs and total IgGSignificant reductions in serum IgG (64.8%)
and
anti-TSHR antibodies (56.7%)
Mild Reversible
Hypoalbuminemia, Headache
Batoclimab (HBM9161)ASCEND-GO 2 (Phase IIb) [71]RCT on
moderate-to-severe
active TED
13 weeks12 QW SC injections,
various doses tested
(680 mg, 340 mg, 255 mg)
Proptosis reduction ≥ 2 mmDespite early termination due to unexpected
increases in cholesterol
levels, significant
reductions in
pathogenic antibodies
Were observed
Reversible Increase
in Serum
Cholesterol Levels
Batoclimab (HBM9161)NCT05517421
(Phase III) [72]
RCT on
moderate-to-severe
active TED
24 weeks24 QW SC injections
(twelve 680 mg inj.
+
twelve 340 mg inj.)
Incidence of
treatment-emergent adverse events (TEAEs)
Expected in 2027Not Yet
Reported
EfgartigimodUplighTED
(Phase III) [73]
RCT on
moderate-to-severe
active TED
24 weeks for
primary
endpoint;
extended
up to
110 weeks
24 QW SC injections
(1 g prefilled syringe)
Proptosis
reduction ≥ 2 mm
Expected in 2027
(75–90% IgG reduction
in preclinical
murine models)
No Impact on IgM, IgA, Cholesterol or
Albumin in Other
Conditions Tested
VRND-006Phase I [74]Healthy
volunteers
Not yet reportedNot yet
reported
Not yet
reported
Expected late 2025
Similar to Efgartigimod in preclinical trials.
Safety Profile
Similar to
Efgartigimod in Preclinical Trials
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ciarmatori, N.; Quaranta Leoni, F.; Quaranta Leoni, F.M. Redefining Treatment Paradigms in Thyroid Eye Disease: Current and Future Therapeutic Strategies. J. Clin. Med. 2025, 14, 5528. https://doi.org/10.3390/jcm14155528

AMA Style

Ciarmatori N, Quaranta Leoni F, Quaranta Leoni FM. Redefining Treatment Paradigms in Thyroid Eye Disease: Current and Future Therapeutic Strategies. Journal of Clinical Medicine. 2025; 14(15):5528. https://doi.org/10.3390/jcm14155528

Chicago/Turabian Style

Ciarmatori, Nicolò, Flavia Quaranta Leoni, and Francesco M. Quaranta Leoni. 2025. "Redefining Treatment Paradigms in Thyroid Eye Disease: Current and Future Therapeutic Strategies" Journal of Clinical Medicine 14, no. 15: 5528. https://doi.org/10.3390/jcm14155528

APA Style

Ciarmatori, N., Quaranta Leoni, F., & Quaranta Leoni, F. M. (2025). Redefining Treatment Paradigms in Thyroid Eye Disease: Current and Future Therapeutic Strategies. Journal of Clinical Medicine, 14(15), 5528. https://doi.org/10.3390/jcm14155528

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop