Home Can chimeric antigen receptors – based therapy bring a gleam of hope for thyroid-associated ophthalmopathy and other autoimmune diseases?
Article Open Access

Can chimeric antigen receptors – based therapy bring a gleam of hope for thyroid-associated ophthalmopathy and other autoimmune diseases?

  • Weiyi Zhou ORCID logo , Xinyu Zhu and Yongquan Shi EMAIL logo
Published/Copyright: April 14, 2025

Abstract

Thyroid-associated ophthalmopathy (TAO), also known as Graves’ ophthalmopathy (GO) is an autoimmune disease (AD) with abnormal thyroid function typically. Currently, intravenous glucocorticoid therapy remains the first-line treatment for moderate-to-severe active TAO. Second-line treatments, including immunosuppressants and biological agents, are being explored in depth. However, like other ADs, the adverse effects of these therapies, little impact on long-term sequelae, and the irreversible progression of the disease remain significant limitations. As a result, the development of new therapeutic strategies for TAO is essential. Chimeric antigen receptors (CAR)-based adoptive cell therapy has emerged as an innovative approach for ADs treatment, capitalizing on its principles of genetically modifying immune cells to specifically target pathogenic cells. This approach aims to reduce autoimmune response or eliminate effective cells, CAR-based therapies of both T-cell-mediated and B-cell-mediated ADs have shown promising results in wide clinical trial. CAR-based therapy obviously become a rising star on refractory and relapsed ADs. TAO is no exception in terms of the potential for improvement through CAR-based therapy. However, the success of CAR-based therapy in TAO depends critically on identifying appropriate targets. Selected targets need to be coverage to ensure the therapeutic efficiency while specificity to preserve safety. Furthermore, the target cells must be relevant to the pathogenesis of TAO. Except target selection, adopting advanced and effective strategies for CAR design is also crucial. For example, dual-target approaches involving thyroid-stimulating hormone receptor (TSHR) or insulin-like growth factor-1 receptor (lGF-1R), off-the-shelf CAR-based cells, or leveraging artificial intelligence (Al) to predict optimal targets could enhance the specificity and effectiveness of CAR-based, therapies in TAO treatment.

Therapeutics of TAO in the present state and its tight corner

Thyroid-associated ophthalmopathy (TAO), also known as Graves’ ophthalmopathy (GO). It is a representative extrathyroidal manifestation of Graves’ disease (GD). TAO is usually associated with hyperthyroidism due to GD, while 10 % of TAO patients had normal thyroid function or hypothyroidism [1]. The prevalence of TAO is estimated to be between 90 and 305 cases per 100,000 people [2]. The annual incidence of TAO is 16/100,000 in female and 3/100,000 in male [3]. The research of risk factors for TAO is significant for the assessment and treatment of the disease. Female, older age, history of smoking, or history of radioactive iodine therapy (RAI) are risk factors for TAO [4]. In addition, the risk of TAO is known to rise with later diagnosis and longer duration of GD [5]. The most typical clinical signs and symptoms of TAO are: eyelid recession in more than 90 % of patients, eyeball protrusion in 60 %, restrictive extraocular myopathy in 40 %, and ocular swelling and pain or discomfort in 30 % [6]. TAO exerts a significant impact on the quality of life, affecting individuals even when the disease is mild [7], [8]. In addition, TAO creates a significant burden on public health due to associated direct and indirect costs [9]. TAO is a complex autoimmune disease and its exact pathological mechanisms have not been fully explained, so its treatment remains challenging.

Glucocorticoids with other agents

Intravenous glucocorticoids (IVGC) have been recommended as first-line treatment for moderate-to-severe and active TAO due to their anti-inflammatory and immunosuppressive effects, which are more effective and better tolerated than oral glucocorticoids [10]. In a multicenter, randomized, double-blind trial conducted by L. Bartalena et al. [11], 159 participants were randomized to three different cumulative doses of intravenous methylprednisolone, 2.25 g, 4.98 g and 7.47 g. Overall ocular improvement was more common with the 7.47 g dose (52 %) than with 4.98 g (35 %; p<0.03) and 2.25 g (28 %; p<0.01). Clinical activity scores (CAS) decreased in all groups, with the high-dose regimen showing the most significant improvements in objectively measured ocular motility and CAS. However, the improvement of IVGC on proptosis and extraocular muscle dysfunction was limited [12]. The dose of IVGC therapy needs to be closely monitored, and excessive doses may lead to adverse effects such as hepatotoxicity [13]. glucocorticoids should not be used in patients with the following contraindications: recent hepatitis, hepatic insufficiency, uncontrolled hypertension, and severe cardiovascular disease [14]. Relapse rates were high, with 33 % in the 7.47 g group, 21 % in the 4.98 g group, and 40 % in the 2.25 g group of patients with 12th week improvement in GO experiencing recurrence of orbital disease after glucocorticoid withdrawal at the exploratory 24th week visit [11].

In addition, glucocorticoids can also be combined with immunosuppressive agents for TAO treatment. Cyclosporine is a robust immunosuppressive agent that inhibit the calcineurin pathway, and reduce T-cell proliferation and IL-2 secretion. In an random clinical trials (RCT) study [15], the response rate to the combination of prednisone and cyclosporine was 59 % in participants who did not respond to monotherapy. It was better tolerated in the cyclosporine group.

Azathioprine (AZA) is an antiproliferative agent, frequently used in autoimmune diseases for reducing steroid dosage. AZA is not an effective treatment for moderately severe TAO alone [16]. But evidence from retrospective research demonstrates that AZA as a steroid-sparing agent may reduce the overall cumulative steroid dose and associated side effects [17]. Methotrexate is a widely used antimetabolic agent. In a small scale non-randomized trial in active TAO, adjunctive IVMP therapy with methotrexate is an effective and safe treatment for moderately to severely active TAO, significantly reducing disease activity and total steroid dose. And participants received 50 % fewer IVGC treatments [18]. Cyclophosphamide (CYC) is an alkylating agent that inhibits the proliferation of immune cells by disrupting cell replication and division through an alkylation reaction with DNA. Combined CYC/GCs therapy may be a therapy option for patients with TAO who do not respond well to IVGCs therapy and who relapse after withdrawal of IVGCs [19]. Of note, there are no clinical evidence-based studies comparing those immunosuppressive agents alone with IVGC for the treatment of TAO.

Mycophenolate mofetil

Mycophenolate mofetil (MMF) inhibits the proliferation of T and B cells as well as antibody production by inhibiting inosine dehydrogenase. In addition, MMF is known to inhibit fibroblast proliferation [20]. In a multicenter, randomized, observer-masked trial (MINGO) study, 164 patients with active and moderate to severe TAO were enrolled and were randomly assigned to two groups, methylprednisolone alone and methylprednisolone with a combination of glucocorticoids. In 24th week, 38 of 72 participants (53 %) in the alone treatment group responded, and 53 (71 %) of 75 participants in the combination treatment group responded (OR=2.16, 95 % CI 1.09–4.25, p=0.026). Although no statistically significant differences were observed in response rates at 12th week and recurrence rates at 24th and 36th week, post hoc analyses suggest that the addition of MMF to GCs treatment in patients with active moderate to severe TAO improves the response rate at 24th week and improves the quality of life of patients [21]. In a meta-analysis conducted in 2022, the combination of GCs and MMF was more effective, with a more significant decrease in CAS compared with GCs monotherapy (WMD=0.29, 95 % CI 0.10–0.48; p=0.002) and a lower incidence of adverse events in the MMF treatment group (OR=0.2, 95 % CI 0.06–0.72; p=0.01) [22]. At present, the sample size of relevant studies is too small. More multicenter, randomized, controlled, double-blind trials are required to confirm these results.

Orbital radiotherapy

Orbital radiotherapy (ORT) has been used as an adjuvant treatment for active TAO for more than 60 years due to its anti-inflammatory effects and radiosensitivity of orbital lymphocytes. Given its long-term use, many prospective and retrospective studies have been performed, but the efficacy of TAO remains controversial. Two large retrospective studies have shown that radiotherapy, either with or without corticosteroids, is effective in preventing or treating compressive optic neuropathy (CON) [23]. Newly evaluated studies have further proven that ORT improves examination findings and clinical activity, including ocular mobility restrictions and CAS [24]. ORT also helps reduce the side effects of long-term glucocorticoids use, such as diabetes, liver disease, adrenal suppression, and infections [25]. However, there is level 1 evidence that ocular proptosis, eyelid retraction and soft tissue damage do not improve with ORT [26]. Although ORT is generally safe, there is a potential risk of side effects such as cataracts or radiation retinopathy.

Teprotumumab and other IGF-1R-targeting agents

IGF-1R plays an important role in the pathogenesis of TAO, so it is important to develop new drugs targeting IGF-1R. Teprotumumab is a human monoclonal antibody that binds to the extracellular portion of IGF-1R and blocks its activation. In an RCT [27], patients with active, moderately severe TAO were included and randomly assigned in a 1:1 ratio to either the teprotumumab group or the placebo group. Injections every 3 weeks for a total of eight doses in 24 weeks. 83 % (34 patients) in the teprotumumab group had a significant reduction in proptosis in 24th week, while only 10 % (4 patients) in the placebo group showed such a reduction (p<0.001). All secondary endpoints were significantly superior in the teprotumumab group compared to the placebo group, including overall response (78 % vs. 7 %), a clinical activity score of 0 or 1 (59 % vs. 21 %), a mean change in proptosis (−2.82 mm vs. −0.54 mm), diplopia response (68 % vs. 29 %), and a mean change in overall GO-QoL scores (13.79 vs. 4.43 points). In OPTIC-X study [28], it was found that patients who had an insufficient response to initial treatment or who had disease relapse may benefit from additional treatment with teprotumumab. The efficacy of teprotumumab was consistent across patient subgroups [29]. And it allows for long-term maintenance of response in most patients [29]. The majority of adverse events with teprotumumab were mild to moderate, with the greatest difference in risk of muscle cramps, hearing loss, and high glucose in the teprotumumab group compared to the placebo group [30]. The disadvantages are that teprotumumab, a newer treatment, is expensive and not available in all regions.

Linsitinib is a highly selective small molecule dual inhibitor of IGF-1R and insulin receptor. It binds to the structural domain of cytoplasmic tyrosine kinase, thereby inhibiting the intrinsic tyrosine kinase activity of IGF-1R [31]. Studies in animal models have shown that linsitinib was effective in preventing autoimmune hyperthyroidism in the early treatment group and in reducing immune infiltration in the orbit in the late treatment group. Suggesting its potential as a new approach for the treatment of TAO [32]. A phase 2b RCT is currently underway to study the efficacy and safety of linsitinib in active TAO (ClinicalTrials.gov identifier: NCT05276063). Subcutaneous injection of IGF-1R antibodies is being actively researched, a phase 1–2 RCT comparing lonigutamab with placebo is ongoing (ClinicalTrials.gov identifier: NCT05683496). Clinical trials of VRDN-001, a full antagonist of IGF-1R, are also underway, including two phase 3 RCTs in patients with active TAO (ClinicalTrials.gov identifier: NCT05176639) and patients with inactive TAO (ClinicalTrials.gov identifier: NCT06021054).

Rituximab

Rituximab (RTX) is a human-mouse chimeric monoclonal antibody that targets the antigen CD20 on B cells leading to B cell depletion. An RCT conducted in Italy compared the efficacy of rituximab and methylprednisolone in 31 participants [33]. In 24th week, the RTX group showed a 100 % reduction in CAS, which proved to be more effective than the GCs group, which showed a 69 % reduction. A meta-analysis including 152 patients with TAO showed that, compared to baseline, it reduced CAS and TSHR Antibody (TRAb) levels. While rituximab did not affect proptosis [34]. In contrast, a RCT conducted at the Mayo Clinic in the United States found no effect of rituximab compared with placebo in improving CAS, including proptosis and diplopia [35]. Comparing these two RCTs, TAO duration averaged 4.5 months in Italy [33], while it averaged 12 months in the United States [35]. Therefore, rituximab may be a more effective treatment for patients with TED of shorter duration. Injection reactions were mild in the RTX group compared to the IVMP and placebo groups, but adverse events occurred more commonly. The current evidence is inadequate to support the use of RTX in patients with TAO. Further multicenter studies are required to enroll enough participants to adequately evaluate the efficacy and safety of this new therapy [36].

Tocilizumab and other IL-6-targeting agents

IL-6 is a pro-inflammatory cytokine that can activate T and B cells and promote TSHR expression in orbital fibroblasts. In an RCT, which enrolled 32 moderate to severe TAO participants with glucocorticoids resistance, it was observed that tocilizumab was effective in reducing CAS compared to the placebo group (93.3 % vs. 58.8 %, in 16th week) [37]. In addition, at both 16th and 40th week, proptosis was reduced by 1.5 mm in the tocilizumab group, while there was no change in the placebo group. However, no significant difference in CAS was observed in 40th week. Among the adverse events, infections and headaches occupied a high proportion, and neutropenia and high cholesterol were also observed. A phase 2 RCT comparing tocilizumab and IVGCs is ongoing (ClinicalTrials.gov identifier: NCT04876534). While a Phase 3 multinational RCT is also underway to evaluate the efficacy and safety of satralizumab, an anti-IL-6 monoclonal antibody (ClinicalTrials.gov identifier: NCT05987423). A novel developed long-term anti-IL - 6 antibody, TOUR006, is currently undergoing a phase 2 RCT to compare the efficacy of TOUR006 20 mg, TOUR006 50 mg, and placebo in patients with active TAO (ClinicalTrials.gov identifier: NCT06088979).

Sirolimus

Sirolimus is a macrolide immunosuppressant used to prevent organ rejection after transplantation. It targets the mammalian target of rapamycin protein (mTOR), modulates cell growth and proliferation, and has antifibrotic characteristics by acting on GCs [38]. It can inhibit mTOR complex 1 (mTORC1), which is involved in adipogenesis, and also has a role in reducing adipogenesis [39]. In one case report, diplopia and field of binocular single vision (BSV) improved over a period of several months after sirolimus treatment, and no adverse events directly attributable to the treatment were observed [40]. A recent observational single-center clinical study compared the efficacy of sirolimus with GCs in moderate to severe active TAO [41]. The proportion of TAO responders at 24th week was significantly higher in the sirolimus group than in the GCs group (86.6 % vs. 26.6 %), and GO-QoL scores were also higher in the sirolimus group. Advantage of sirolimus is that it rarely causes side effects due to its relatively small dose. However, its efficacy has not been confirmed as the results of RCT studies are not yet available. Further randomized clinical trials are required to confirm these observations. At present, two phase 2 RCTs are underway to compare sirolimus and GCs for efficacy (ClinicalTrials.gov identifier: NCT04598815 and NCT04936854).

Batoclimab

The neonatal fragment crystallizable receptor (FcRN) has the potential to protect immunoglobulin G (IgG) from intracellular degradation and increase its half-life. Batoclimab blocks the recirculation of IgG by competitive binding to FcRN, thereby reducing pathogenic TRAb [42]. In a randomized, double-blind, placebo-controlled trial, there was a significant difference in the rate of proptosis response with Batoclimab compared to placebo at multiple early time points, but not at the primary endpoint of 12th week, which may be due to data insufficient [42]. A multicenter, quadruple-masked phase 3 study is currently underway to observe the effects of batoclimab on proptosis (ClinicalTrials.gov identifiers: NCT05517421 and NCT05524571). A Phase 3 RCT was also conducted to evaluate the efficacy and safety of efgartigimod (FcRN antagonist) compared to placebo in patients with active and moderate-to-severe TAO (ClinicalTrials.gov identifiers: NCT06307626 and NCT06307613).

However, like other ADs, the adverse effects of these therapies, little impact on long-term sequelae, and the irreversible progression of the disease remain significant limitations (Table 1). As a result, the development of new therapeutic strategies for TAO is essential.

Table 1:

Current TAO treatments and its efficacy.

Method Target Efficacy Adverse effects
Cyclosporine [15] Inhibits the calcineurin pathway The response rate to the combination of prednisone and cyclosporine was 59 % in participants who did not respond to monotherapy. Hypertension
Diarrhea
Hirsutism
Azathioprine [17] Inhibits the proliferation of lymphocyte Evidence from retrospective research demonstrates that AZA as a steroid-sparing agent may reduce the overall cumulative steroid dose and associated side effects; Nausea
Leukopenia
Methotrexate [18] DHFR Adjunctive IVMP therapy with methotrexate is an effective and safe treatment for moderately to severely active TAO, significantly reducing disease activity and total steroid dose. Participants received 50 % fewer IVGC treatments. Nausea
Vomiting
Oral ulcers
Diarrhea
Abnormal liver function
Bone marrow suppression
Cyclophosphamide [19] Alkylation reaction with DNA Combined CYC & GCs therapy is an option for patients with TAO who do not respond well to IVGCs therapy and who relapse after withdrawal. Infection risk
Bone marrow suppression
Reproductive system
Hemorrhagic cysts and pulmonary fibrosis
Mycophenolate mofetil [21] Inhibits the proliferation of T and B cells The combination of GCs and MMF was more effective, with a more significant decrease in CAS compared with GCs monotherapy. Nausea
Vomiting
Diarrhea
Abdominal pain
Orbital radiotherapy [24] Anti-inflammatory effects and radiosensitivity ORT improves examination findings and clinical activity, including ocular mobility restrictions and CAS Cataracts
Radiation retinopathy
Teprotumumab [27], [29] IGF-1R The efficacy was consistent across patient subgroups. Long-term maintenance of response in most patients. Muscle cramps
Hearing loss
Hyperglycemia
Linsitinib [32]

(NCT05276063)
IGF-1R Animal study showed that linsitinib was effective in preventing autoimmune hyperthyroidism in the early treatment group and in reducing immune infiltration in the orbit in the late treatment group. A phase 2b RCT is ongoing. RCT is ongoing
Lonigutamab

(NCT05683496)
IGF-1R Subcutaneous injection of IGF-1R antibodies is being actively researched, a phase 1 and 2 RCT comparing lonigutamab with placebo is ongoing. RCT is ongoing
VRDN-001

(NCT05176639, NCT06021054)
IGF-1R Clinical trials of VRDN-001, a full antagonist of IGF-1R, are also underway, including two phase 3 RCTs in patients with active TAO and patients with inactive TAO. RCT is ongoing
Rituximab [33], [34], [35] CD20 In Italy RCT, RTX group reduced CAS and TRAb levels, did not affect proptosis. In contrast, an US RCT found no effect of rituximab compared with placebo in improving CAS, including proptosis and diplopia. Diarrhea
Cytokine release syndrome
Retrobulbar hemorrhage, hypertension
Hyperglycemia
Weight gain
Serious infections
Tocilizumab [37]

(NCT04876534)
IL-6 Tocilizumab was effective in reducing CAS compared to the placebo group. In addition, at both 16th and 40th week, proptosis was reduced by 1.5 mm in the tocilizumab group, while there was no change in the placebo group. Infections
Headaches
Neutropenia
High cholesterol
Satralizumab

(NCT05987423)
IL-6 While a phase 3 multinational RCT is also underway to evaluate the efficacy and safety of satralizumab. RCT is ongoing
TOUR006

(NCT06088979)
IL-6 Undergoing a phase 2 RCT to compare the efficacy of TOUR006 20 mg, TOUR006 50 mg, and placebo in patients with active TAO. RCT is ongoing
Sirolimus [41]

(NCT04598815, NCT04936854)
mTOR The proportion of TAO responders at 24th week was significantly higher in the sirolimus group than in the GCs group (86.6 % vs. 26.6 %), and GO-QoL scores were also higher in the sirolimus group. Rare side effects due to small dose
Batoclimab [42]

(NCT06307626, NCT06307613)
FcRN There was a significant difference in the rate of proptosis response with Batoclimab compared to placebo at multiple early time points, but not at the primary endpoint of 12th week, which may be due to data insufficient. Fatigue
Muscle cramps
Peripheral edema
Serum albumin decreased
High cholesterol
Nausea
High intraocular pressure
  1. AZA, azathioprine; DHFR, dihydrofolate Reductase; TAO, thyroid-associated ophthalmopathy; IVGC, intravenous glucocorticoids; CYC, cyclophosphamid; CAS, clinical activity scores; ORT, orbital radiotherapy; RCT, random clinical trials; RTX, rituximab; TRAb, thyroid-stimulating hormone receptor antibody; FcRN, neonatal fragment crystallizable receptor.

In autoimmune disease the CAR-based therapy bursts into life

Contemporary therapeutic strategies for ADs encompass the administration of immunosuppressants, glucocorticoids, and monoclonal antibodies while presenting several limitations, including heightened susceptibility to opportunistic infections and an absence of long-lasting therapeutic response. As the advancement of personalization and precision medicine, chimeric antigen receptor (CAR)-based adoptive cell therapy has paved a novel field as an innovative approach for AD treatment, it harnesses the inherent attack capabilities of immune cells, which are activated through engineered receptors that bind to target cells. Currently, over 70 registered clinical trials are carried out [43], bringing immense hope to desperate patients and the uncertain future of AD research.

Binding to targets on the surface of cells specifically, CAR-based therapy eliminates pathogenesis cells to alleviate uncontrol autoimmune therapy. In terms of T or B cell-mediated ADs, target selection is distinctive (Figure 1). In B cell-mediated ADs, such as systemic lupus erythematosus (SLE), Neuromyelitis Optica Spectrum Disorders (NMOSD), Sjogren’s Syndrome, and so on, CD19, CD20, BAFF-R, and BCMA have turned out to be effective and popular targets in CAR-based trials depleting B cells in ADs. By contrast with targeting generally expressed B cell markers, targeting specifically autoreactive B cells will leave patients avoid to be vulnerable to infections and impair long-term immune function. So, chimeric autoantibody receptor (CAAR)-based therapy raised to the occasion. CAAR-based therapy replaces the CAR’s scFv domain with an autoantigen. This mimics B cell receptor (BCR)-antigen recognition, enabling autoactivated B cells to bind to CAAR-modified cells and leading to the destruction of memory B cells and plasma cells. In T cell-mediated ADs, clinical researches on T cell target CAR-based therapies are limited compared to B cell-targeting therapies. One approach is targeting general T cell markers like CD5 and CD7. Similarly, a more refined strategy involves modifying the CAR to recognize T cells via MHC-T cell receptor (TCR) interaction. For example, Fishman [44] et al. developed a CAR that targets autoimmune CD8+ T cells in NOD mice by presenting the InsB15-23 peptide with MHC-I. This therapy reduced insulin secretion and prevented diabetes in these mice.

Figure 1: 
Target and modified cell selection in current CAR-based therapy of autoimmune diseases (ADs). In CAR-based therapies for ADs, target selection depends on whether T cells or B cells primarily mediate the disease’s pathogenesis. One approach is to target broadly expressed surface antigens, while another involves mimicking the process of B cells or T cells recognizing antigens. Additionally, the choice of CAR-modified cells is diverse, with current strategies primarily focusing on NK cells, T cells, and T regulatory (Treg) cells. ADs, autoimmune diseases; CAR, chimeric antigen receptor; NK, natural killer.
Figure 1:

Target and modified cell selection in current CAR-based therapy of autoimmune diseases (ADs). In CAR-based therapies for ADs, target selection depends on whether T cells or B cells primarily mediate the disease’s pathogenesis. One approach is to target broadly expressed surface antigens, while another involves mimicking the process of B cells or T cells recognizing antigens. Additionally, the choice of CAR-modified cells is diverse, with current strategies primarily focusing on NK cells, T cells, and T regulatory (Treg) cells. ADs, autoimmune diseases; CAR, chimeric antigen receptor; NK, natural killer.

Another question is, on which cells are the CAR loaded? Most current trials are concentrated on T cells, but NK cells, with their advantages of a lower risk of autoimmunity and graft-versus-host disease (GVHD), can also be utilized as carriers for CARs in ADs. In addition, CAR-regulatory T cells (Treg) therapy modifies Tregs to target autoreactive immune cells, using suppressive cytokines and regulatory functions to restore immune tolerance and reduce inflammation in autoimmune diseases without directly killing the target cells.

Advanced innovation has provided us with substantial approaches to expand CAR-based therapy to more ADs, but we should hold a firm belief that it’s vital to choose appropriate approaches in terms of the unique pathogenesis of diseases themselves.

Hit the TAO’s eye

Attempts to apply CAR-based therapies in ADs have already begun, and TAO is no exception – it too presents a compelling opportunity for the exploration of CAR-based therapies as a potential innovative treatment. However, the key to success, also the premise, lies in selecting the most appropriate “weapon” to hit the TAO at its core. The most significant part of the “weapon” is identifying the right target, selected from the pathogenesis of TAO. Selected targets should provide broad coverage to ensure therapeutic efficiency while maintaining specificity to preserve safety [45].

The autoimmune response in TAO is complex, involving orbital fibroblasts, T cells, B cells, and a range of cytokines [46]. T and B lymphocytes infiltrate from the peripheral circulation into orbital tissues, beginning with recognizing TSHR or lGF-1R. Among them, T cells which are considered as the predominant link differentiate into Th1, Th2, and Th17 subsets, secreting cytokines that drive inflammation. They are seen to be a better target to curtail or eliminate abnormal immune responses from origin. In addition, CD34+ fibrocytes migrate into the orbital environment, differentiating into orbital fibroblasts (OFs). These OFs further differentiate into myofibroblasts and adipocytes, and also accumulate hyaluronic acid, leading to orbital tissue expansion (Figure 2). So, targeting fibroblasts directly is possible to improve the bad outcomes closely related to clinical manifestation to some extent. CD34 may serve as a specific target for the selective elimination of OFs. However, because CD34 is widely expressed in stem cells, targeting it alone may cause substantial off-target effects, risking a “Pyrrhic victory” where the harm to healthy tissues outweighs the therapeutic benefits. Given the complexity of disease pathogenesis in TAO and other ADs, it is challenging to identify an optimal target that balances broad efficacy with safety. To overcome these obstacles, advanced strategies can be leveraged to equip our CAR weapons.

Figure 2: 
Pathogenesis of thyroid-associated ophthalmopathy. The pathogenesis of TAO begins with the aberrant recognition of the TSHR and its interaction with the IGF-1R. This leads to the infiltration of T and B lymphocytes from the peripheral circulation into orbital tissues. Upon receiving activation signals from APCs, T cells differentiate into Th1, Th2, and Th17 subsets, which secrete cytokines that drive inflammation. Concurrently, B cells are stimulated, transforming into plasma cells that produce antibodies binding to TSHR. CD34+fibrocytes migrate into the orbital environment, differentiating into orbital fibroblasts. Depending on Thy-1 expression, these OFs further differentiate into myofibroblasts and adipocytes, contributing to orbital tissue expansion. Additionally, the accumulation of hyaluronic acid exacerbates this process. The interaction between T cells and OFs plays a crucial role in the pathogenesis of TAO. TAO, thyroid-associated ophthalmopathy; TSHR, thyrotropin receptor; IGF-1R, insulin-like growth factor 1 receptor; APCS, antigen-presenting cells; Th1, T helper 1; Th2, T helper 2; Th17, T helper 17; Thy-1, CD90; OFs,orbital fibroblasts.
Figure 2:

Pathogenesis of thyroid-associated ophthalmopathy. The pathogenesis of TAO begins with the aberrant recognition of the TSHR and its interaction with the IGF-1R. This leads to the infiltration of T and B lymphocytes from the peripheral circulation into orbital tissues. Upon receiving activation signals from APCs, T cells differentiate into Th1, Th2, and Th17 subsets, which secrete cytokines that drive inflammation. Concurrently, B cells are stimulated, transforming into plasma cells that produce antibodies binding to TSHR. CD34+fibrocytes migrate into the orbital environment, differentiating into orbital fibroblasts. Depending on Thy-1 expression, these OFs further differentiate into myofibroblasts and adipocytes, contributing to orbital tissue expansion. Additionally, the accumulation of hyaluronic acid exacerbates this process. The interaction between T cells and OFs plays a crucial role in the pathogenesis of TAO. TAO, thyroid-associated ophthalmopathy; TSHR, thyrotropin receptor; IGF-1R, insulin-like growth factor 1 receptor; APCS, antigen-presenting cells; Th1, T helper 1; Th2, T helper 2; Th17, T helper 17; Thy-1, CD90; OFs,orbital fibroblasts.

Dual-target CAR-based therapy enhances specificity by targeting multiple surface antigens. For instance, CD34 can be targeted alongside TSHR and IGF-1R as co-targets in a CAR-based therapy, since these antigens are co-expressed on OFs. Off-the-shelf CAR cells, pre-manufactured from healthy donors, offer quicker and more affordable treatment options. It has already been explored in a clinical trial for myositis and systemic sclerosis, showing promising results [47]. Furthermore, the suicide gene strategy, which involves incorporating genes that allow CAR cells to be selectively eliminated in case of adverse effects, could add an extra layer of safety to these therapies.

Discussion

Currently, in terms of CAR-based therapy for TAO, there is no related research reported, which is an indication of a novel but challenging field. The key limitation is to define one or more accurate and efficient targets to construct CAR cells, and it also needs practice to choose ideal strategies and carried cells. Nonetheless, research on CAR-based therapy for Grave’s disease and thyroid cancer has been in process, regarding TSHR as targets [48], [49], [50]. IGF-1R targeted CAR-based therapy has also been explored [51]. These results will cast light on the attempt of CAR-based therapy on TAO.

Among the application of CAR-based therapy for TAO, accuracy, and safety are two vital aspects of CAR-based therapy evaluation. Therefore, identifying precise targets and mitigating adverse effects such as cytokine release syndrome present importance which is the rationale behind our proposition to equip the CAR with strategies. Dual-target CAR design has shown good results in the treatment of thyroid cancer [49] and can be used to design CAR-based cells of TAO. The potential of applying CAR-based therapy to TAO and other ADs treatments is both exciting and challenging. However, we must acknowledge that CAR-based therapies are not exclusive to oncology; they hold potential advantages in treating ADs as well. More efforts and innovations should be pursued to break the longstanding challenge of treating TAO and other autoimmune diseases.


Corresponding author: Yongquan Shi, Department of Endocrinology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China, E-mail:
Weiyi Zhou and Xinyu Zhu contributed equally to the work.

Award Identifier / Grant number: 23SG33

Acknowledgments

The figures were created with Biorender.com by XZ and WZ.

  1. Research ethics: Not applicable.

  2. Informed consent: Not applicable.

  3. Author contributions: All authors have accepted responsibility for the entire content of this manuscript and approved its submission.

  4. Use of Large Language Models, AI and Machine Learning Tools: None declared.

  5. Conflict of interest: The authors state no conflict of interest.

  6. Research funding: This study was supported by the “Dawn” Program of Shanghai Education Commission, China and the Distinguished Young Scholars of the National Defense Biotechnology Foundation (23SG33).

  7. Data availability: Not applicable.

References

1. Hoang, TD, Stocker, DJ, Chou, EL, Burch, HB. 2022 Update on clinical management of graves disease and thyroid eye disease. Endocrinol Metab Clin North Am. 2022;51:287–304, https://doi.org/10.1016/j.ecl.2021.12.004.Search in Google Scholar PubMed PubMed Central

2. Bartalena, L, Gallo, D, Tanda, ML, Kahaly, GJ. Thyroid eye disease: epidemiology, natural history, and risk factors. Ophthalmic Plast Reconstr Surg 2023;39:S2–8. https://doi.org/10.1097/iop.0000000000002467.Search in Google Scholar

3. Smith, TJ, Hegedüs, L. Graves’ disease. N Engl J Med 2016;375:1552–65. https://doi.org/10.1056/nejmra1510030.Search in Google Scholar PubMed

4. Smith, TJ. Understanding pathogenesis intersects with effective treatment for thyroid eye disease. J Clin Endocrinol Metab 2022;107:S13–26. https://doi.org/10.1210/clinem/dgac328.Search in Google Scholar PubMed PubMed Central

5. Khong, JJ, Finch, S, De Silva, C, Rylander, S, Craig, JE, Selva, D, et al.. Risk factors for graves’ orbitopathy; the Australian thyroid-associated orbitopathy research (ATOR) study. J Clin Endocrinol Metab. 2016;101:2711–20, https://doi.org/10.1210/jc.2015-4294.Search in Google Scholar PubMed

6. Bartley, GB. The epidemiologic characteristics and clinical course of ophthalmopathy associated with autoimmune thyroid disease in Olmsted County, Minnesota. Trans Am Ophthalmol Soc 1994;92:477–588.Search in Google Scholar

7. Schovanek, J, Radvansky, M, Karhanova, M, Bolacka, M, Pekarova, K, Dohnal, R, et al.. Long-term impact of thyroid eye disease on quality of life: insights from a retrospective cohort study. Endocr Pract. 2025;S1530-891X:0004-2.Search in Google Scholar

8. Wang, C, Qiao, J, Liu, S, Piao, S, Zhou, Y, Hu, Y, et al.. Selenium in the treatment of mild-to-moderate Graves’ orbitopathy: a 5-year prospective controlled cohort study. Endocrine 2024;84:1072–80. https://doi.org/10.1007/s12020-023-03672-5.Search in Google Scholar PubMed

9. Ponto, KA, Merkesdal, S, Hommel, G, Pitz, S, Pfeiffer, N, Kahaly, GJ. Public health relevance of Graves’ orbitopathy. J Clin Endocrinol Metab 2013;98:145–52. https://doi.org/10.1210/jc.2012-3119.Search in Google Scholar PubMed

10. Zang, S, Ponto, KA, Kahaly, GJ. Clinical review: intravenous glucocorticoids for Graves’ orbitopathy: efficacy and morbidity. J Clin Endocrinol Metab 2011;96:320–32. https://doi.org/10.1210/jc.2010-1962.Search in Google Scholar PubMed

11. Bartalena, L, Krassas, GE, Wiersinga, W, Marcocci, C, Salvi, M, Daumerie, C, et al.. Efficacy and safety of three different cumulative doses of intravenous methylprednisolone for moderate to severe and active Graves’ orbitopathy. J Clin Endocrinol Metab 2012;97:4454–63. https://doi.org/10.1210/jc.2012-2389.Search in Google Scholar PubMed

12. Wiersinga, WM. Immunosuppressive treatment of Graves’ ophthalmopathy. Trends Endocrinol Metab 1990;1:377–81. https://doi.org/10.1016/1043-2760(90)90095-k.Search in Google Scholar PubMed

13. Marinó, M, Morabito, E, Brunetto, MR, Bartalena, L, Pinchera, A, Marocci, C. Acute and severe liver damage associated with intravenous glucocorticoid pulse therapy in patients with Graves’ ophthalmopathy. Thyroid 2004;14:403–6. https://doi.org/10.1089/105072504774193276.Search in Google Scholar PubMed

14. Längericht, J, Krämer, I, Kahaly, GJ. Glucocorticoids in Graves’ orbitopathy: mechanisms of action and clinical application. Ther Adv Endocrinol Metab 2020;11. https://doi.org/10.1177/2042018820958335.Search in Google Scholar PubMed PubMed Central

15. Prummel, MF, Mourits, MP, Berghout, A, Krenning, EP, van der Gaag, R, Koornneef, L, et al.. Prednisone and cyclosporine in the treatment of severe Graves’ ophthalmopathy. N Engl J Med 1989;321:1353–9. https://doi.org/10.1056/nejm198911163212002.Search in Google Scholar PubMed

16. Perros, P, Weightman, DR, Crombie, AL, Kendall-Taylor, P. Azathioprine in the treatment of thyroid-associated ophthalmopathy. Acta Endocrinol (Copenh) 1990;122:8–12. https://doi.org/10.1530/acta.0.1220008.Search in Google Scholar PubMed

17. Chalvatzis, NT, Tzamalis, AK, Kalantzis, GK, El-Hindy, N, Dimitrakos, SA, Potts, MJ. Safety and efficacy of combined immunosuppression and orbital radiotherapy in thyroid-related restrictive myopathy: two-center experience. Eur J Ophthalmol 2014;24:953–9. https://doi.org/10.5301/ejo.5000463.Search in Google Scholar PubMed

18. Sipkova, Z, Insull, EA, David, J, Turner, HE, Keren, S, Norris, JH. Early use of steroid-sparing agents in the inactivation of moderate-to-severe active thyroid eye disease: a step-down approach. Clin Endocrinol (Oxf) 2018;89:834–9. https://doi.org/10.1111/cen.13834.Search in Google Scholar PubMed

19. Xiang, Q, Yang, M, Luo, W, Cao, Y, Shuai, S, Wei, X, et al.. Combined glucocorticoids and cyclophosphamide in the treatment of Graves’ ophthalmopathy: a systematic review and meta-analysis. BMC Endocr Disord 2024;24:12. https://doi.org/10.1186/s12902-024-01545-0.Search in Google Scholar PubMed PubMed Central

20. Azzola, A, Havryk, A, Chhajed, P, Hostettler, K, Black, J, Johnson, P, et al.. Everolimus and mycophenolate mofetil are potent inhibitors of fibroblast proliferation after lung transplantation. Transplantation 2004;77:275–80. https://doi.org/10.1097/01.tp.0000101822.50960.ab.Search in Google Scholar PubMed

21. Kahaly, GJ, Riedl, M, König, J, Pitz, S, Ponto, K, Diana, T, 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;6:287–98. https://doi.org/10.1016/s2213-8587(18)30020-2.Search in Google Scholar

22. Feng, W, Hu, Y, Zhang, C, Shi, H, Zhang, P, Yang, Y, et al.. Efficacy and safety of mycophenolate mofetil in the treatment of moderate to severe Graves’ orbitopathy: a meta-analysis. Bioengineered 2022;13:14719–29. https://doi.org/10.1080/21655979.2022.2101191.Search in Google Scholar PubMed PubMed Central

23. Shams, PN, Ma, R, Pickles, T, Rootman, J, Dolman, PJ. Reduced risk of compressive optic neuropathy using orbital radiotherapy in patients with active thyroid eye disease. Am J Ophthalmol 2014;157:1299–305. https://doi.org/10.1016/j.ajo.2014.02.044.Search in Google Scholar PubMed

24. Choi, JH, Lee, JK. Efficacy of orbital radiotherapy in moderate-to-severe active graves’ orbitopathy including long-lasting disease: a retrospective analysis. Radiat Oncol 2020;15:220. https://doi.org/10.1186/s13014-020-01663-8.Search in Google Scholar PubMed PubMed Central

25. Hahn, E, Laperriere, N, Millar, BA, Oestreicher, J, McGowan, H, Krema, H, et al.. Orbital radiation therapy for Graves’ ophthalmopathy: measuring clinical efficacy and impact. Pract Radiat Oncol 2014;4:233–9. https://doi.org/10.1016/j.prro.2014.02.008.Search in Google Scholar PubMed

26. Bradley, EA, Gower, EW, Bradley, DJ, Meyer, DR, Cahill, KV, Custer, PL, et al.. Orbital radiation for graves ophthalmopathy: a report by the American Academy of Ophthalmology. Ophthalmology 2008;115:398–409. https://doi.org/10.1016/j.ophtha.2007.10.028.Search in Google Scholar PubMed

27. Douglas, RS, Kahaly, GJ, Patel, A, Sile, S, Thompson, EHZ, Perdok, R, et al.. Teprotumumab for the treatment of active thyroid eye disease. N Engl J Med 2020;382:341–52. https://doi.org/10.1056/nejmoa1910434.Search in Google Scholar PubMed

28. Douglas, RS, Kahaly, GJ, Ugradar, S, Elflein, H, Ponto, KA, Fowler, BT, et al.. Teprotumumab efficacy, safety, and durability in longer-duration thyroid eye disease and Re-treatment: OPTIC-X study. Ophthalmology 2022;129:438–49. https://doi.org/10.1016/j.ophtha.2021.10.017.Search in Google Scholar PubMed

29. Kahaly, GJ, Douglas, RS, Holt, RJ, Sile, S, Smith, TJ. Teprotumumab for patients with active thyroid eye disease: a pooled data analysis, subgroup analyses, and off-treatment follow-up results from two randomised, double-masked, placebo-controlled, multicentre trials. Lancet Diabetes Endocrinol 2021;9:360–72. https://doi.org/10.1016/s2213-8587(21)00056-5.Search in Google Scholar

30. Douglas, RS, Couch, S, Wester, ST, Fowler, BT, Liu, CY, Subramanian, PS, et al.. Efficacy and safety of teprotumumab in patients with thyroid eye disease of long duration and low disease activity. J Clin Endocrinol Metab 2023;109:25–35. https://doi.org/10.1210/clinem/dgad637.Search in Google Scholar PubMed PubMed Central

31. Wong, PC, Christ, DD, Timmermans, PB. Enhancement of losartan (DuP 753)-induced angiotensin II receptor antagonism by PD123177 in rats. Eur J Pharmacol 1992;220:267–70. https://doi.org/10.1016/0014-2999(92)90758-v.Search in Google Scholar PubMed

32. Gulbins, A, Horstmann, M, Daser, A, Flögel, U, Oeverhaus, M, Bechrakis, NE, et al.. Linsitinib, an IGF-1R inhibitor, attenuates disease development and progression in a model of thyroid eye disease. Front Endocrinol (Lausanne) 2023;14:1211473. https://doi.org/10.3389/fendo.2023.1211473.Search in Google Scholar PubMed PubMed Central

33. Salvi, M, Vannucchi, G, Currò, N, Campi, I, Covelli, D, Dazzi, D, et al.. Efficacy of B-cell targeted therapy with rituximab in patients with active moderate to severe Graves’ orbitopathy: a randomized controlled study. J Clin Endocrinol Metab 2015;100:422–31. https://doi.org/10.1210/jc.2014-3014.Search in Google Scholar PubMed PubMed Central

34. Chen, J, Chen, G, Sun, H. Intravenous rituximab therapy for active Graves’ ophthalmopathy: a meta-analysis. Hormones (Athens) 2021;20:279–86. https://doi.org/10.1007/s42000-021-00282-6.Search in Google Scholar PubMed

35. Shen, WC, Lee, CH, Loh, EW, Hsieh, AT, Chen, L, Tam, KW. Efficacy and safety of rituximab for the treatment of Graves’ orbitopathy: a meta-analysis of randomized controlled trials. Pharmacotherapy 2018;38:503–10. https://doi.org/10.1002/phar.2111.Search in Google Scholar PubMed

36. Kang, S, Hamed Azzam, S, Minakaran, N, Ezra, DG. Rituximab for thyroid-associated ophthalmopathy. Cochrane Database Syst Rev 2022;6:CD009226. https://doi.org/10.1002/14651858.CD009226.pub3.Search in Google Scholar PubMed PubMed Central

37. Perez-Moreiras, JV, Gomez-Reino, JJ, Maneiro, JR, Perez-Pampin, E, Romo Lopez, A, Rodríguez Alvarez, FM, et al.. Efficacy of tocilizumab in patients with moderate-to-severe corticosteroid-resistant graves orbitopathy: a randomized clinical trial. Am J Ophthalmol 2018;195:181–90. https://doi.org/10.1016/j.ajo.2018.07.038.Search in Google Scholar PubMed

38. Molina-Molina, M, Machahua-Huamani, C, Vicens-Zygmunt, V, Llatjós, R, Escobar, I, Sala-Llinas, E, et al.. Anti-fibrotic effects of pirfenidone and rapamycin in primary IPF fibroblasts and human alveolar epithelial cells. BMC Pulm Med 2018;18:63. https://doi.org/10.1186/s12890-018-0626-4.Search in Google Scholar PubMed PubMed Central

39. Zhang, L, Grennan-Jones, F, Lane, C, Rees, DA, Dayan, CM, Ludgate, M. Adipose tissue depot-specific differences in the regulation of hyaluronan production of relevance to Graves’ orbitopathy. J Clin Endocrinol Metab 2012;97:653–62. https://doi.org/10.1210/jc.2011-1299.Search in Google Scholar PubMed

40. Roos, JCP, Murthy, R. Sirolimus (rapamycin) for the targeted treatment of the fibrotic sequelae of Graves’ orbitopathy. Eye (Lond). 2019;33:679–82. https://doi.org/10.1038/s41433-019-0340-3.Search in Google Scholar PubMed PubMed Central

41. Lanzolla, G, Maglionico, MN, Comi, S, Menconi, F, Piaggi, P, Posarelli, C, et al.. Sirolimus as a second-line treatment for Graves’ orbitopathy. J Endocrinol Invest 2022;45:2171–80. https://doi.org/10.1007/s40618-022-01862-y.Search in Google Scholar PubMed PubMed Central

42. Kahaly, GJ, Dolman, PJ, Wolf, J, Giers, BC, Elflein, HM, Jain, AP, et al.. Proof-of-concept and randomized, placebo-controlled trials of an FcRn inhibitor, batoclimab, for thyroid eye disease. J Clin Endocrinol Metab 2023;108:3122–34. https://doi.org/10.1210/clinem/dgad381.Search in Google Scholar PubMed PubMed Central

43. Cheever, A, Kang, CC, O’Neill, KL, Weber, KS. Application of novel CAR technologies to improve treatment of autoimmune disease. Front Immunol 2024;15:1465191. https://doi.org/10.3389/fimmu.2024.1465191.Search in Google Scholar PubMed PubMed Central

44. Fishman, S, Lewis, MD, Siew, LK, De Leenheer, E, Kakabadse, D, Davies, J, et al.. Adoptive transfer of mRNA-transfected T cells redirected against diabetogenic CD8 T cells can prevent diabetes. Mol Ther 2017;25:456–64. https://doi.org/10.1016/j.ymthe.2016.12.007.Search in Google Scholar PubMed PubMed Central

45. Wei, J, Han, X, Bo, J, Han, W. Target selection for CAR-T therapy. J Hematol Oncol 2019;12:62. https://doi.org/10.1186/s13045-019-0758-x.Search in Google Scholar PubMed PubMed Central

46. Bahn, RS. Graves’ ophthalmopathy. N Engl J Med 2010;362:726–38. https://doi.org/10.1056/nejmra0905750.Search in Google Scholar

47. Wang, X, Wu, X, Tan, B, Zhu, L, Zhang, Y, Lin, L, et al.. Allogeneic CD19-targeted CAR-T therapy in patients with severe myositis and systemic sclerosis. Cell 2024;S0092–8674:00701–3.Search in Google Scholar

48. TSHR-based chimeric antigen receptor T cell specifically deplete auto-reactive B lymphocytes for treatment of autoimmune thyroid disease - PubMed [Internet]. [cited 2025 Feb 17]. Available from: https://pubmed.ncbi.nlm.nih.gov/37690235/.Search in Google Scholar

49. Ding, J, Li, D, Liu, X, Hei, H, Sun, B, Zhou, D, et al.. Chimeric antigen receptor T-cell therapy for relapsed and refractory thyroid cancer. Exp Hematol Oncol 2022;11:59. https://doi.org/10.1186/s40164-022-00311-z.Search in Google Scholar PubMed PubMed Central

50. Zhou, J, Zhang, C, Mao, W, Zhu, Y, Zhao, H, Han, R, et al.. Development of TSHR-CAR NK-92 cells for differentiated thyroid cancer. Mol Cell Endocrinol 2024;589:112251. https://doi.org/10.1016/j.mce.2024.112251.Search in Google Scholar PubMed

51. IGF1R- and ROR1-specific CAR T cells as a potential therapy for high risk sarcomas - PubMed [Internet]. [cited 2025 Feb 17]. Available from: https://pubmed.ncbi.nlm.nih.gov/26173023/.Search in Google Scholar

Received: 2024-12-18
Accepted: 2025-03-03
Published Online: 2025-04-14

© 2025 the author(s), published by De Gruyter, Berlin/Boston

This work is licensed under the Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License.

Downloaded on 28.9.2025 from https://www.degruyterbrill.com/document/doi/10.1515/mr-2024-0099/html
Scroll to top button