You are on page 1of 16

Drugs

https://doi.org/10.1007/s40265-018-1045-9

THERAPY IN PRACTICE

Current and Emerging Treatment Strategies for Graves’ Orbitopathy


Natalia Genere1 · Marius N. Stan1 

© Springer Nature Switzerland AG 2019

Abstract
Graves’ orbitopathy is a debilitating disorder which occurs in patients with autoimmune thyroid disease, mainly Graves’ dis-
ease, and adds layers of complexity to management of both conditions. We conducted a comprehensive review of literature for
publications relating to established and new management options for Graves’ orbitopathy and have summarized key articles
in this review. Initial evaluation of patients with Graves’ disease should also include clinical evaluation for orbitopathy. If
eye disease is present, patients are best managed by a multi-specialty team including an endocrinologist and ophthalmolo-
gist. All patients with Graves’ orbitopathy benefit from risk factor modification and normalization of thyroid function tests.
Patients with active, mild disease generally benefit from local therapies and selenium, while patients with moderate-to-severe
disease usually require the addition of intravenous glucocorticoid therapy. If there is an inadequate response to glucocor-
ticoid therapy, several second-line therapies have been investigated for use, including orbital radiotherapy (with additional
glucocorticoids), rituximab, cyclosporine, mycophenolate mofetil, and methotrexate. Use of new biologic agents, mainly
teprotumumab and tocilizumab, have demonstrated impressive reductions in disease activity and severity. If these results are
confirmed, the treatment paradigm is likely to change in the future. Finally, there are several novel immunotherapies being
investigated for Graves’ disease, which may have treatment implications for Graves’ orbitopathy as well. Overall, there are
many encouraging advances in the therapy of Graves’ orbitopathy that are making the future more promising for patients
suffering from this disease.

1 Introduction
Key Points 
Graves’ orbitopathy (GO) is an autoimmune, inflamma-
The evaluation of Graves’ orbitopathy should determine tory disorder of the orbit and represents the most common
its clinical activity, its severity, and the impact it has on a extra-thyroidal manifestation of Graves’ disease, present in
patient’s quality of life. anywhere between 25 and 70% of cases, depending on the
Current standard of managing Graves’ orbitopathy spans diagnostic modality employed (clinical vs CT imaging) [1].
from local supportive measures and selenium supple- Onset of GO commonly concurs with onset of autoimmune
mentation to intravenous glucocorticoids and rehabilita- thyroid disease, but subclinical eye disease may sometimes
tive surgery. be present before the clinical syndrome becomes evident.
While most cases of GO are associated with hyperthyroid-
Multiple new treatments are being explored for Graves’ ism, a minority of patients (about 10%) may be hypothy-
orbitopathy and Graves’ disease, mainly focusing on roid or euthyroid [2]. Typically, the natural history of the
immunomodulatory approaches. Teprotumumab and tocili- disease includes an active, inflammatory stage (ranging
zumab are leading the way, but there are many other prom- 3–6 months), followed by clinical stabilization (ranging
ising options at various levels of development, exploiting 1–3 years) and quiescence with return towards baseline ocu-
the now better understood pathophysiology of the disease. lar status [3, 4]. This paradigm has been illustrated by Run-
dle’s curve, named after the physician who first described
the progression of GO in two of his patients. For this review,
* Marius N. Stan
stan.marius@mayo.edu we focus on available and emerging medical therapies for
patients with active GO.
1
Division of Endocrinology, Diabetes, Metabolism
and Nutrition, Mayo Clinic, 200 1st St SW, Rochester,
MN 55905, USA

Vol.:(0123456789)
N. Genere, M. N. Stan

2 Evaluation of Patient with Graves inflammation of the orbit [7]. Consequently, the CAS score
Orbitopathy (GO) is an important predictor of response to anti-inflammatory
therapies. The CAS measurement awards one point for each
Crucial to timely and effective management of GO is the of the following criteria: spontaneous retrobulbar pain, pain
clinician’s familiarity with manifestations of orbital dis- on attempted upwards or downwards gaze, redness of the
ease and their grading [5, 6]. All patients diagnosed with eyelids, redness of the conjunctiva, swelling of the caruncle
Graves’ disease should be evaluated clinically for presence or plica, swelling of the eyelids and swelling of the con-
or absence of orbitopathy. If orbitopathy is present, its evalu- junctiva (Table 1). Scores of CAS < 3 suggest inactive GO
ation should include three important elements: (1) degree of while scores ≥3 suggest active GO. An expanded, 10-point
activity, (2) degree of severity, and (3) impacts on a patient’s CAS scoring system is applicable for patients who have
quality of life. The algorithm for approaching a patient with subsequent assessments, with additional points allocated for
GO is summarized in Fig. 1. decreases in visual acuity, worsening diplopia, and increas-
ing proptosis, with scores ≥4 indicative of clinically active
2.1 Assessing Disease Activity disease [7].

GO activity can be evaluated using a validated scoring sys- 2.2 Assessing Disease Severity
tem called the clinical activity score (CAS), which repre-
sents a quantitative measurement of the degree of active GO severity is qualified by categories of mild, moderate-
to-severe, and sight-threatening, based on the presence and

Fig. 1  Algorithm for the treatment of Graves’ orbitopathy (GO). IVGC intravenous glucocorticoid, QOL quality of life
Current and Emerging Treatment Strategies for Graves’ Orbitopathy

degree of lid retraction, soft tissue involvement, proptosis, 2.3 Quality of Life Evaluation
diplopia, corneal exposure, and optic nerve involvement
(Table 2) [5]. Mild GO is the most common form of GO and GO-related impairments can be substantial and carry sub-
encompasses approximately 75% of the impacted group [8]. stantial psychosocial and financial implications for impacted
Mild GO is defined by absent or minimal features in severity patients [9, 10]. Several questionnaires have been validated
factors, and absent signs of corneal or optic nerve involve- specifically for use in GO patients, in an effort to estab-
ment. Patients with moderate-to-severe GO have increased lish quantifiable markers for impacts of orbitopathy on a
soft tissue involvement with at least one advanced sever- patient’s quality of life [10–12]. While specific questions
ity parameter, but without sight-threatening complications. vary amongst questionnaires, key concepts of importance
Finally, very severe GO, or sight-threatening GO, includes include impact of GO on daily visual functioning, impacts
any patient with dysthyroid optic neuropathy, corneal break- on appearance and self-perception, and impacts on mood
down, or evidence of globe subluxation. Sight-threatening and overall perceptions of wellness.
disease, whether active or inactive, requires immediate oph- While we review the management options categorized,
thalmologic evaluation and consideration of urgent medical based on levels of severity, it is important to include qual-
and surgical therapies. ity-of-life status and patient preferences in determining the

Table 1  Graves’ orbitopathy activity assessment according to CAS

Clinical activity parameter Points score


 Spontaneous retrobulbar pain 1
 Pain on attempted upward or downward gaze 1
 Redness of eyelid 1
 Redness of conjunctiva 1
 Swelling of caruncle and plica 1
 Swelling on eyelids 1
 Swelling on conjunctiva (chemosis) 1
Scoring of initial CAS (of 7)
CAS <3 = inactive GO
CAS ≥3 = active GO
Clinical activity subsequent evaluation Additional points
 Increase in proptosis of ≥2 mm during a period of 1–3 months 1
 Decrease of eye movements in any direction ≥ 5° during a period of 1–3 months 1
 Decrease of visual acuity of ≥1 line(s) on the Snellen chart (using a pinhole) during a period of 1–3 months 1
Scoring of subsequent CAS (of 10)
CAS <4 = inactive GO
CAS ≥4 = active GO

CAS clinical activity score, GO Graves’ orbitopathy

Table 2  Graves’ orbitopathy severity assessment according to EUGOGO


Degree of severity Lid retraction Soft tissue involve- Proptosisa Diplopia Corneal exposure Optic nerve status
ment

Mild (≥ 1 of the fol- < 2 mm Mild < 3 mm Absentb or intermit- Absent Normal
lowing) tent
Moderate-to-severe ≥ 2 mm Moderate or severe ≥ 3 mm Inconstantb or Mild Normal
(≥ 1 of the follow- constant
ing)
Sight threatening Not contributory Not contributory Not contributory Not contributory Ulceration Compromised
(one of the last 2
categories)
a
 Proptosis refers to variation from the normal for each race and gender or to the patient’s baseline, if applicable
b
 Transient diplopia refers to presence of symptoms at times of fatigue; inconstant diplopia refers to presence of symptoms at extremes of gaze;
constant diplopia is presence of symptoms at all times
N. Genere, M. N. Stan

appropriate treatment plan. For example, a patient with mild measures to help with symptom management [27]. Current
GO may still benefit from corticosteroid treatment if the ret- therapies for GO and their mechanisms of action are sum-
roocular or eye-movement induced ocular pain prohibits her marized in Fig. 2.
from successfully completing her occupation as an account- Local therapies alleviate symptoms associated with mild
ant. Now, we will turn to reviewing medical management orbitopathy and can be useful in both active and inactive
options for active GO and its supporting literature; selected GO [5]. Ocular lubrication with artificial tears or gels can
prominent studies of therapeutic interventions for GO are alleviate dryness and foreign body sensation. This is particu-
highlighted in Table 3. larly helpful for patients with a large palpebral aperture due
to lid retraction. For patients who are particularly troubled
by morning symptomatology due to lagophthalmos, thicker
3 Risk Factor Modification for Management ointments or gels can be protective from nocturnal ocular
of GO drying. Prisms or eye patching (if divergence is large) can
be beneficial for patients with symptoms of diplopia. Small
All patients at risk for or with established GO, regardless studies have used sub-conjunctival botulinum toxin A injec-
of disease status, benefit significantly from modification of tions for reduction of upper eyelid retraction, but the benefits
their risk factors for onset and progression of GO. While seem to be clinically small and transient [28–30].
some risk factors are not readily altered, like age, sex, and In addition to local measures, patients with mild, active
serum thyrotropin receptor antibody (TRAb) level, modifi- GO may benefit from oral selenium supplementation, par-
cation of smoking status and thyroid function are crucial in ticularly those in selenium-deficient areas. Selenium is a
management of the condition [8, 13, 14]. trace element that contributes to normal thyroid function
Cigarette smoking has been strongly linked to risk for devel- [31]. In 2011, Marcocci et al, under the European Group
opment and progression of GO. Amongst patients with Graves’ on GO (EUGOGO) umbrella, conducted a randomized,
disease, smoking patients were more likely to have orbital placebo-controlled trial in 5 European countries, compar-
involvement compared to non-smokers [8, 15]. GO patients ing the efficacy of selenium (100 µg twice daily), pen-
who smoke have been found to follow a more severe clinical toxifylline, and placebo for ocular symptoms of mild GO
course as well as a poorer responsiveness to standard therapies [32]. Euthyroid patients were treated with 6 months of
[15–18]. Severity of orbital symptoms seems to be dependent on supplementation followed by 6 months of observation.
the dose of the exposure to cigarettes, and active smokers have At the completion of the study, investigators found that
significantly more symptoms compared to former smokers [17, there were significantly more patients in the selenium
19]. Therefore, discussion of smoking cessation is of crucial group with symptomatic improvement (61% vs 36% in
importance and is emphasized by major GO guidelines [5, 20]. placebo group), and significantly fewer patients with dis-
Normalization of thyroid function is important in mitigat- ease progression (7% vs 26% in placebo group). No sig-
ing the severity of GO. The majority of patients with GO are nificant adverse effects were noted in the selenium group.
initially hyperthyroid and require normalization of thyroid Of note, the study was conducted in six European centers
function. The degree of a patient’s hyperthyroidism has been with various degrees of selenium sufficiency, and whether
associated with the perceived severity of GO [21]. Euthy- this impacted the generalizability of the study results is
roidism can typically be achieved by use of anti-thyroid unclear. A retrospective review of 84 patients with GO
medications, but surgery is also a valid option after under- from Germany, a majority of whom were selenium deplete,
standing a patient’s values and preferences and it might be suggested that GO severity did not correlate with selenium
preferred in a subset of cases [22]. Use of radioactive iodine or selenoprotein P concentrations [33]. The role of sele-
(RAI) in patients with active GO is associated with a small nium supplementation in selenium-sufficient areas, like
but significant risk for worsening of GO, more prominently the USA, is still unclear, but is a low-risk intervention for
in smokers, and can be possibly circumvented with concur- patients with mild, active GO.
rent use of steroids [23, 24]. Care must be used to actively For most patients with mild GO, local therapies and sele-
prevent development of overt hypothyroidism, which has nium are helpful in supporting patients through the natu-
also been associated with disease progression [25, 26]. ral disease course. However, for patients who progress and
develop significant impairments to quality of life, consid-
eration should be given to approaching them in a manner
4 Management of Mild Disease similar to that of patients with moderate-to-severe GO. Dis-
cussion of risks and benefits to therapeutic options should
Because more than half of patients with mild GO experience be discussed prior to initiation of therapy. These will be
spontaneous resolution of ocular symptoms within about discussed further in the next section.
6 months, management of mild GO is focused on low-risk
Table 3  Prominent studies of medical therapies of Graves’ orbitopathy
Study [References] Year of Intervention and comparator Type of study Num- Outcome measures Result
Publica- ber of
tion patients

Marcocci et al. [32] 2011 Selenium vs RCT, double-blind 159 Ophthalmic ­indexa,b Selenium is superior to placebo in
pentoxifylline vs GO-QOLb mild GO (improved symptoms and
placebo CAS prevented disease progression)
Gorman diplopia score
Kahaly et al. [36] 2005 IV vs oral GC RCT, single-blind 70 Ophthalmic ­indexa,b IV MP was superior to oral daily pred-
QL- SF-36b nisone in patients with M-T-S GO
CAS
Bartalena et al. [39] 2012 IV GC at cumulative doses of 7.47 g RCT, double-blind 159 Ophthalmic ­indexa,b Moderate dose if IV MP (5 g) has best
vs 4.98 g vs 2.25 g GO-QOLb risk-benefit profile compared with
CAS high dose (7.5 g) or low dose (2.3 g)
Gorman diplopia score in patients with M-T-S GO
Prummel et al. [44] 2004 Retrobulbar orbital radiotherapy vs RCT, double-blind 88 Ophthalmic ­indexa,b Orbital radiotherapy superior to pla-
placebo CAS cebo, mainly for diplopia (no impact
GO-QOL on QOL) in patients with mild GO
Cost effectiveness
Current and Emerging Treatment Strategies for Graves’ Orbitopathy

Need for follow-up


Oeverhaus et al. [50] 2017 IV GC with vs without orbital Retrospective 148 Ophthalmic ­indexa Orbital radiotherapy + IV MP had
radiotherapy CAS higher improvement in severity,
NOSPECS particularly in ocular motility, in
M-T-S GO
Salvi et al. [51] 2015 Rituximab vs IV GC RCT, double-blind 32 CASb Rituximab was superior to IV MP at
Ophthalmic ­indexa reaching clinically inactive disease in
NOSPECS M-T-S GO of short disease duration
Gorman diplopia score
GO-QOL
Stan et al. [52] 2015 Rituximab vs placebo RCT, double-blind 25 CASb Rituximab was equivalent to placebo,
Ophthalmic ­indexa regarding CAS reductions but had
Gorman diplopia score more frequent serious adverse events
SF-12 QOL in M-T-S GO of median 1-year
disease duration
Kahaly et al. [54] 1986 Cyclosporine with oral GC vs oral RCT, blinding not mentioned 40 Ophthalmic ­indexa Cyclosporine + prednisone was
GC alone Orbital CT superior to prednisone alone regard-
ing CAS reductions in patients with
M-T-S GO
Ye et al. [56] 2017 MMF vs IV/oral GC RCT, single-blind 74 Ophthalmic ­indexa,b MMF was superior to GC in reducing
CAS ophthalmic involvement and CAS in
patients with M-T-S GO

Table 3  (continued)
Study [References] Year of Intervention and comparator Type of study Num- Outcome measures Result
Publica- ber of
tion patients

Kahaly et al. [58] 2018 MMF with IV GC vs IV GC alone RCT, single-blind 164 Ophthalmic ­indexa,b Addition of MMF to standard IV
CAS GC therapy did not meaning-
GO-QOL fully improve clinical response at
12 weeks, but may improve certain
symptoms and QOL, in patients with
M-T-S GO
Strianese et al. [61] 2014 Methotrexate Retrospective 36 Ophthalmic ­indexa In patients with GC intolerance,
CAS methotrexate monotherapy was asso-
ciated with reductions in CAS and
improvements in motility in patients
with M-T-S GO
Rajendram et al. [64] 2018 Azathioprine + oral GC vs orbital RCT, double-blind, 2 × 2 factorial 126 Ophthalmic ­indexa,b Addition of azathioprine to GC did not
radiotherapy + oral GC vs azathio- CASb improve GO clinical severity param-
prine + orbital radiotherapy +oral eters in patients with M-T-S GO
GC vs oral GC alone
Baschieri et al. [65] 1997 IVIG vs IV GC Prospective, non-randomized, single- 65 Ophthalmic ­indexa IVIG and IV MP had similar efficacy
blinded NOSPECS in GO, with a safer profile in the
Orbital CT IVIG therapy group
Stan et al. [69] 2006 Octreotide long-acting release vs RCT, double-blind 29 CAS,b Octreotide LAR resulted in small
placebo Ophthalmic ­indexa reductions in CAS and in lid retrac-
Orbital CT tion, at the cost of gastrointestinal
side effects in a minority of patients
Smith et al. [73] 2017 Teprotumumab vs placebo RCT, double-blind 88 Ophthalmic ­indexa,b Teprotumumab was superior to pla-
CAS cebo at reducing CAS and proptosis
GO-QOL in patients with M-T-S GO
Perez-Moreiras et al. [78] 2018 Tocilizumab vs placebo RCT, double-blind 32 CASb Tocilizumab was superior to placebo
Ophthalmic ­indexa at reducing CAS and proptosis in
EUGOGO severity corticosteroid-resistant patients with
GO-QOL M-T-S GO
Ayabe et al. [80] 2014 Adalimumab, with varied prior Retrospective 10 Ophthalmic ­indexa Adalimumab was not associated with
treatments significant reductions in inflamma-
tory parameters, and recurrence rate
was high after drug discontinuation

CAS clinical activity score [7], EUGOGO European Group on Graves’ Orbitopathy, GC glucocorticoids, GO Graves’ orbitopathy, GO-QOL Graves’ Ophthalmopathy Quality of Life assessment
[12], IVIG intravenous immune globulin, MMF mycophenolate mofetil, M-T-S moderate-to-severe, MP methylprednisolone, NOSPECS classification for severity of eye disease [5], RCT​ rand-
omized controlled trial, QL-SF-36 and SF-12 QOL are both non-disease specific, validated quality of life questionnaires
a
 Ophthalmic index varied between studies but commonly included several variables such as proptosis, eyelid aperture, soft tissues involvement, diplopia/motility. Adverse events were men-
tioned in multiple studies, and these results are mentioned in results, where pertinent
b
 Primary outcome, if listed
N. Genere, M. N. Stan
Current and Emerging Treatment Strategies for Graves’ Orbitopathy

Fig. 2  Summary of mecha-
nisms of action for therapies
for Graves’ orbitopathy. IGF-1r
Ab insulin-like growth factor 1
receptor antibody, IL-6 inter-
leukin-6, IL-6R interleukin-6
receptor, MMF mycophenolate
mofetil, TNFα tumor necrosis
factor alpha, TNFαR tumor
necrosis factor alpha recep-
tor, TSHr thyrotropin receptor,
TSHr Ab thyrotropin receptor
antibody

5 Management of Moderate‑to‑Severe GO the interpretation of the efficacy of treatment are: (1) lack
of a uniform outcome assessment, with studies employing
Patients with active, moderate-to-severe GO have sufficient CAS, severity measures (diplopia, proptosis etc.), quality
symptoms and orbital changes to warrant immunosuppres- of life or a mixture of all these in analyzing response, mak-
sive treatments but do not have evidence for sight-threaten- ing between-studies comparison difficult and (2) lack of a
ing disease. As mentioned above, patients with significant control group in many studies that would account for the
impairment in quality of life may warrant more aggressive natural history of the disease, raising questions about the
treatment compared to those with less severe manifesta- actual impact of the specific intervention tested.
tions. The trend of disease progression over the most recent
3 months is used by most clinicians as guide to patient’s 5.1 Glucocorticoids
natural history and as basis for deciding on the risk-bene-
fit ratio of any intervention. GCs are considered first-line The mainstay of treatment for moderate-to-severe GO are
therapy for management of active, moderate-to-severe GO. systemic GC therapies, which can be delivered orally or
We will present available data regarding nuances of ster- via intravenous (IV) administration. Multiple randomized
oid management, followed by alternate therapies in cases of controlled trials (RCTs) have looked at the impact of route
GC failure or intolerance. Two elements which complicate of administration with respect to improvements in clinical
N. Genere, M. N. Stan

activity and markers of severity of GO [34, 35]. Stiebel- group; after the 12-week protocol, there were three patients
Kalish et al. conducted a systematic review and meta-analy- in the high-dose group with rebound DON after completion
sis of 33 RCTs, including 1367 patients comparing efficacies of treatment. Other adverse events occurred more commonly
of available treatments of GO [35]. Of these, there were 4 in the high-dose group, including severe psychiatric mani-
trials, including 206 patients, specifically comparing efficacy festations, new diabetes, and severe infections requiring hos-
of oral versus IV GC therapies in patients with active GO of pitalization. None of the patients in this study experienced
varying severity. In patients with moderate-to-severe GO, IV severe hepatotoxicity. Based on these findings, the authors
pulse steroids were significantly more effective at reducing concluded that the moderate dose (5.0 g) provided the best
CAS [standardized mean difference − 0.64, 95% confidence benefit-to-risk ratio for the majority of patients.
interval (CI) − 1.11 to − 0.17]. At the same cumulative dose, weekly administrations
Furthermore, IV GCs were associated with significantly of IV GCs appear to be superior to daily administrations,
lower rates of adverse events compared to oral GCs (OR 0.12, and there was no significant difference between weekly and
95% CI 0.05–0.26) [35]. Side effect profiles between the two monthly dosing [40, 41]. Early deterioration despite pulse
routes also varied. Patients who received oral GCs experi- IV GCs at 6 weeks of therapy may be predictive of future
enced weight gain (26%), hypertension (8%), and cushingoid GC failure, and other treatment options should be considered
features (7%) while patients in the IV group experienced [42]. European Group on Graves Orbitopathy (EUGOGO)
symptoms within 24 h of infusion consisting of flushing guidelines currently recommend IV GCs as first-line therapy
(20%), transient dyspepsia (15%), and palpitations (8%), and for active, moderate-to-severe GO [5]. Cumulative doses
fewer metabolic complications (3–4%). Insomnia is a com- across treatments should not exceed 8 g and it is important
monly reported problem with IV steroids the night after the that physicians are attuned to the multiple contraindications
infusion. Treatment was discontinued in four patients in the for pulse corticosteroids. The most commonly employed reg-
oral corticosteroid group and none in the IV corticosteroids imen remains the one tested by Kahaly et al. and described
group. The use of IV GCs was also associated with improved above (methylprednisolone 0.5 g weekly for 6 weeks, fol-
quality of life and fewer subsequent surgeries [36]. There have lowed by 0.25 g weekly for 6 weeks, for cumulative dose of
been reports of acute, fatal hepatotoxicity in patients treated 4.5 g) [36]. The most severe cases may benefit from high-
with high doses of IV GCs, with most patients receiving dose therapy, of 0.75 g/infusion for 6 weeks, followed by
cumulative doses of >8 g or daily doses of ≥ 589 mg meth- 0.5 g/infusion for an additional 6 weeks (7.5 g cumulative).
ylprednisolone [37]. Currently available data suggest that IV Overall, significant progress has been made in the last
steroids are the preferred method for treatment of active GO, decade with regard to optimal use of steroids in GO. IV
although one must account also for cost and convenience dif- GCs represent the standard of care, and first-line treatment
ferences between these two routes when deciding manage- of moderate-to-severe, active GO. However, in cases of
ment for a given patient. Therefore, attention to the different poor response or unacceptable side effects from IV GCs, a
regimens employed over time in the various studies is needed, number of second-line therapies have been proposed. These
and likely will identify a certain degree of heterogeneity. treatment options have been far less tested than steroid-based
Historically, oral prednisone was used at starting doses therapies and evidence for these treatments is rarely based
of 40–100  mg/day with tapering over 10–24  weeks for on adequately powered RCTs.
cumulative doses of 2–6 g [38]. The preferred regimen of
administering IV GCs has been the topic of more recent 5.2 Second‑Line Therapies for Moderate‑To‑Severe
investigation. After Kahaly et al. identified IV methylpred- GO
nisolone at 6 weekly doses of 500 mg followed by another
6 weekly doses of 250 mg to be superior to oral daily pred- 5.2.1 Orbital Radiotherapy
nisone (for a similar total amount of GCs), Bartalena et al. with and without Glucocorticoids
randomized 159 patients with moderate-to-severe GO to
receive cumulative doses of methylprednisolone 2.3 g, 5.0 g, Radiotherapy has been employed extensively for GO man-
or 7.5 g over 12 weekly infusions [36, 39]. At 12 weeks, agement, although its use has not been without controversy.
there was significantly more ophthalmic improvement (CAS While one RCT conducted by Gorman et al. found no evi-
and ocular mobility) in the high-dose group (52%), com- dence of benefit using 20 Gy of external beam radiation ther-
pared to the moderate- and low-dose groups (35% and 28%, apy compared to sham radiation, most other trials have found
respectively). These differences between the groups were benefit of radiation therapy to some GO parameters [43–45].
not evident by the 24-week follow-up, suggesting that the A meta-analysis conducted by Bradley et al. found that radi-
superiority of the high-dose therapy was short lived. During ation therapy to the orbits is likely to lead to improvement in
the study, six patients developed dysthyroid optic neuropathy diplopia but no other consistent effects could be identified in
(DON), three in the low-dose and three in the moderate-dose the group of trials that used this approach [46].
Current and Emerging Treatment Strategies for Graves’ Orbitopathy

Oral and IV GCs have been combined with orbital radio- rituximab group developing DON and the IV GC-controlled
therapy with the hopes of improving severity parameters trial reported two patients in the rituximab group with acute
and also limiting cumulative GC exposure. Combination visual loss related to an acute cytokine release reaction
therapy with GCs and orbital radiotherapy may be superior (which resolved within hours following administration of
to monotherapy. Ng et al. compared 52-week outcomes of 100 mg of hydrocortisone). A number of other side effects
15 patients treated with GC regimen (three doses of IV fol- were reported in both studies, including many infusion reac-
lowed by an oral taper for approximately 3 months) to those tions. Overall, it appears that rituximab may show promise
who received the same steroid course, as well as 20 Gy over in a select group of patients with short disease duration, but
2 weeks [47]. Patients completing combined therapy had further trials are warranted to confirm this and to explore the
significant improvements in activity and severity measures concerns regarding DON development. EUGOGO guide-
compared to the monotherapy group, without any severe lines currently recommend use of rituximab as one of sev-
adverse events. These results concurred with prior studies of eral options in those patients who do not have a satisfactory
combination therapy with oral GCs [48, 49]. More recently, response to IV GC therapy [5].
Oeverhaus et al. retrospectively analyzed 148 patients with
active, moderate-to-severe GO treated with IV GCs versus 5.2.3 Cyclosporine
IV GCs in addition to orbital radiation, and again found
that combined approach to treatment was associated with Cyclosporine has been tested extensively in transplanta-
more reductions in severity [50]. While those who received tion medicine due to its ability to inhibit the calcineurin
combined treatment had more severe disease at the start pathway, thus decreasing the IL-2 secretion from T cells.
of treatment, subsequent subgroup analysis with matched The combination of cyclosporine with GCs may also be an
severity had similar results. These studies point to the effec- effective treatment for patients with moderate-to-severe GO
tiveness of a multi-modality approach with GCs (oral or IV) disease, while cyclosporine monotherapy is not superior to
and orbital radiotherapy, particularly in patients with more GC therapy alone. Two randomized controlled trials have
severe disease. However, whether this combination therapy compared combination therapy with cyclosporine (starting
is able to avoid rehabilitative surgeries or significantly doses ranging 5–7.5 mg/kg) plus oral GCs (starting doses
improved quality of life remains to be seen. ranging 50–100 mg, tapering down per protocol) compared
to monotherapy, and found superior reduction in GO severity
5.2.2 Rituximab in the combined therapy group after 10–12 weeks of inter-
vention [54, 55]. The first study used combination therapy
Rituximab was the first of several biologic therapies applied in moderate-to-severe GO patients who had not had treat-
to treatment of active GO. After early case series suggested ment to date, while the second offered combination therapy
substantial improvement in CAS in patients with prior GC to non-responders to single drug therapy. Besides clinical
failure, two randomized controlled trials compared the effi- improvement in GO signs, combined therapy was associ-
cacy of rituximab to either placebo or IV GCs [51, 52]. Stan ated with a reduction in relapses of GO, but perhaps with
et al. found no difference of rituximab (2 g total dose) com- adverse renal and infectious side effects, gingival hyperpla-
pared to placebo in clinical disease activity, but Salvi et al. sia and reversible elevations in blood pressure. Given this
[51] found that rituximab (at both 0.5 g and 2 g dosing) had profile, it can probably be considered as add-on combination
improved disease activity (CAS declined from 4.4 ± 0.7 to GCs in patients who had a partial response to that agent
at baseline to 0.6 ± 0.3 by 24 weeks; p < 0.01) and led to as monotherapy.
increased disease inactivation compared to high-dose (7.5 g)
IV GCs (CAS ≤ 3 achieved in 100% rituximab patients vs 5.2.4 Mycophenolate Mofetil
68.7% corticosteroid patients; p = 0.04). Both studies tar-
geted patients with moderate-to-severe and active GO with Mycophenolate mofetil (MMF) is another agent that has an
CAS ≥ 4; however, the cohort in the placebo-controlled immunosuppresive effect, achieved through depleting the
study had significantly longer disease duration, was older, guanosine nucleotides from the T and B cells. Due to this
had higher proportions of males, and had higher TRAb lev- property, MMF was studied in patients with active, moder-
els, all of which may have contributed to a lower respon- ate-to-severe GO both as monotherapy and as combination
siveness to therapy in this group [53]. Of note, there was an therapy with GCs. Ye et al. randomized 174 patients to receive
increased percentage of smokers in the IV GC-controlled 24 weeks of therapy, either 1 g daily of MMF or with a com-
study, which predicted a reduced response to immunosup- bination of IV (3 g over 2 weeks) and oral GC therapy (start-
pressive therapy in previous trials, yet interestingly it did ing at 60 mg/day) tapering over the treatment duration [56].
not seem to diminish the response to rituximab in this trial. Patients in both groups had ocular symptoms for approxi-
The placebo-controlled trial reported two patients in the mately 6 months, had mean CAS scores of 5.2, and most had
N. Genere, M. N. Stan

at least one prior therapy. There was a higher response rate to history of the disease in contrast to the true impact of ther-
therapy in the MMF group (91% vs 68%) at the 24th week, apy. Another retrospective study included 23 patients with
and degree of CAS reduction was higher in the MMF group active, moderate-to-severe GO who were treated with MTX
as well (reduction to CAS 1.9 ± 0.8 vs 2.5 ± 1.4). Rate of and IV GCs. Approximately half needed at least one addi-
adverse events was substantially lower in the MMF group tional steroid-sparing agent during the time of observation
(5.0% vs 28.2%) and the reported events were not serious [62]. Authors report an average cumulative dose of IV GCs
(mild hypokalemia, mild transaminitis). Other reports of MMF of 2.72 g in patients receiving combination therapy, which is
use in GO describe somewhat higher rate of adverse events, significantly lower than the EUGOGO protocol of cumula-
including the occurrence of optic neuropathy [57]. Recently, tive grams. However, the study is limited by the variety of
the EUGOGO group published results of the MINGO trial, regimens used for patients who did not respond to or did not
which randomized patients with active moderate-to-severe tolerate MTX, which may mask the true efficacy of combina-
GO to receive IV GCs versus IV GCs with MMF [58]. In tion therapy. A few other retrospective studies also suggest
both groups, IV GCs were delivered per current EUGOGO that use of MTX is associated with successful tapering off of
recommendations, a total of 4.5 g methylprednisolone over GC therapy. However, more randomized prospective data are
12 weeks. Mycophenolate was administered at 360 mg twice needed to delineate the utility of MTX as monotherapy, or as
daily. Ocular evaluation as performed at 12 weeks, 24 weeks, combination therapy for treatment of active GO.
and 36 weeks. Response rates at week 12 (one of the primary
outcomes), were not significantly different between the two 5.3 Additional Agents
groups (49% monotherapy vs 63% combined therapy, OR 1.8,
95% CI 0.9–3.4). Relapse rates at weeks 24 and 36 (the other 5.3.1 Azathioprine
primary outcomes) were also similar between groups. How-
ever, a post-hoc analysis suggested that at week 24 there were Azathioprine is a purine analog that works as an antime-
significantly more responders in the combined therapy group tabolite and is also used mainly in transplantation medicine
(53% monotherapy vs 71% combination, 95% CI 1.09–4.25). but has proved useful in some autoimmune disorders (e.g.
Adverse events happened at similar rates in both treatment rheumatoid arthritis, inflammatory bowel disease). Unfor-
groups. The authors concluded that addition of MMF to IV tunately, use of azathioprine as monotherapy has not gained
GCs was not impactful in terms of initial response to therapy momentum in treatment of GO since this agent was not
and rate of relapse, but that there may be benefit with longer effective in improving GO compared to control patients [63].
duration of therapy. It is also worth mentioning that combi- Recently, a large double-blinded, randomized controlled trial
nation therapy had promising signs in terms of improvement re-tested this hypothesis and compared in a 2 × 2 factorial
in QOL and ocular symptoms. These somewhat conflicting design, the use of azathioprine, orbital radiotherapy, both
results are likely related to a combination of factors: the rapid therapies, or neither in parallel with an oral GC course [64].
response expected in the MINGO trial (as opposed to what There was a surprisingly high discontinuation rate (>50%)
might be a positive response at 6 months) and on the other and, in the intention-to-treat analysis, the addition of aza-
hand the substandard GC regimen used by Ye et al. Based on thioprine was not found to be beneficial for reducing clinical
these results, one would hope that a multicenter RCT with activity compared to GC therapy alone.
adequate blinding will be performed in order to understand
the potential utility of MMF for GO. 5.3.2 Intravenous Immunoglobulins

5.2.5 Methotrexate Intravenous immunoglobulins (IVIG) have been found to be


as effective as some corticosteroid regimens in treatment of
Methotrexate (MTX) has also been evaluated for possible active, moderate-to-severe GO, as measured by clinical and
utility in GO management [59–61]. Strianese et al. [61] pub- radiographic assessments [65, 66]. Kahaly et al. compared
lished a retrospective trial of 36 patients with intolerance to IVIG 1 g/kg given for 2 days every 3 weeks to oral predniso-
GCs, who were subsequently started on oral MTX therapy, lone tapered from 100 mg/day, each for 20 weeks. Baschieri
at doses of 7.5–10 mg, based on weight. In this open-label, et al. compared a different dosing schedule of 400 mg/kg
uncontrolled study, 67% of patients had a clinical response daily for 5 days, repeated every 3 weeks for 5 months, com-
to treatment at 3 months, with 94% having clinical improve- pared to tapering doses of daily IV methylprednisolone
ment at 1 year. At 3 months, mean CAS score reduced from from 80 mg/day. Response rate was found to be between 62
4.1 ± 1.1 to 2.5 ± 1.1. More than half of the patients were and 76%, with low rates of adverse events seen in the IVIG
smokers and had a median time of 8 months since their prior group. Of note, neither study compared IVIG to weekly,
attempt at treatment. It is important to note that the study moderate dose pulses of IV GCs, which are superior to dos-
cannot differentiate between improvements due to the natural ing regimens included. However, due to the difficult logistics
Current and Emerging Treatment Strategies for Graves’ Orbitopathy

and costs associated with this approach, there has been no the remaining 7 doses. The study enrolled adult patients
further research since the late 1990s and currently this agent who developed GO within 9 months of the start of the
is not being utilized in GO. trial, had CAS ≥ 4 and had not received any prior therapy
with the exception of oral GCs. The trial was funded by
5.3.3 Somatostatin Analogs the manufacturer. In the intention-to-treat group, 20% of
the placebo group and 69% of the teprotumumab group
Somatostatin analogs appeared to be an attractive treat- experienced significant clinical improvement (CAS score
ment option for GO because of their immunomodulatory reduction ≥ 2 and proptosis reduction ≥T2 mm) by week
effects as well as the presence of somatostatin receptors on 24 (OR 8.9; p < 0.01). Patients treated with teprotumumab
orbital fibroblasts. Four randomized, placebo-controlled were also more likely to respond early and to have more
trials compared long-acting release formulations of octreo- pronounced responses to therapy. GO-QOL visual func-
tide or lanreotide versus placebo, with treatment durations tion score increased significantly throughout the trial, but
of 3–8 months and follow-up durations of up to 14 months appearance score did not. Adverse events were generally
[67–70]. The results were fairly consistent, and a subsequent mild and self-resolving, but 5 patients developed more
meta-analysis concluded that there was a small, statistically serious complications (diarrhea, inflammatory bowel
significant reduction in CAS (mean − 0.63, 95% CI − 0.098 disease, Escherichia coli sepsis, autoimmune encepha-
to − 0.28) that was unlikely to be clinically relevant. Further- lopathy, urinary retention). One patient in the placebo
more, there was no benefit in other clinical outcomes such group developed DON. Hyperglycemia was noted in both
as diplopia, proptosis, and lid aperture [35]. Additionally, the teprotumumab and placebo groups (5 and 2 patients,
about 60% of patients treated with somatostatin developed respectively), with more significant hyperglycemia occur-
significant gastrointestinal side effects. Based on the small ring only in the teprotumumab group, in patients with pre-
overall impact in somatostatin treatments and concerns over existing diabetes, and was easily managed with adjust-
adverse effects, somatostatin analogs are not considered ment of anti-hyperglycemic medications. Overall, the trial
helpful options for treatment of active GO. showed strong data for teprotumumab being a safe and
effective treatment for patients with active, moderate-to-
severe GO of recent onset. Therefore, presently there is a
6 Emerging Therapies for GO second RCT ongoing (NCT03298867) that aims to validate
these findings, focusing primarily on the expected benefit
New therapies for GO are targeting specific pathophysi- in proptosis of ≥2 mm.
ologic mechanisms involved in GO. The known pathways
are summarized in Fig. 2. Pharmaceutically targeting these 6.2 Tocilizumab
molecular mechanisms of GO could provide more effec-
tive therapy without the undesired side effects of some of Tocilizumab (TCZ) is a recombinant humanized monoclo-
the non-GO specific approaches. We will review here the nal antibody directed against the IL-6 receptor. IL-6 is a
biologic therapies currently under investigation for GO. pro-inflammatory cytokine, which is also overexpressed in
GO orbital tissues, so blockade of this cytokine was hypoth-
esized to reduce the ongoing inflammation in GO [74]. TCZ
6.1 Teprotumumab has been successfully used to treat resistant rheumatoid
arthritis and other rheumatologic conditions, with few seri-
Orbital fibroblasts express not only thyrotropin receptors, ous adverse effects [75]. Perez-Moreiras et al. published the
but also insulin-like growth factor 1 receptors (IGF-1R). first prospective non-randomized study of 18 patients with
IGF-1R is actually overexpressed in orbital fibroblasts, clinically active (CAS ≥ 4) and GC-resistant disease and
and lymphocytes, of patients with GO [71, 72]. Tepro- treated them with TCZ 8 mg/kg every 4 weeks, until CAS
tumumab is a human monoclonal blocking antibody that ≤ 1 or thyroid-stimulating immunoglobulins were negative
binds to the extracellular portion of the IGF-1R. IGF-1R [76]. Patients in this study were predominantly women,
normally increases thyrotropin action synergistically at the mean age of 48 ± 8.6 years, and 50% were smokers. Fol-
cellular level. After several promising in vitro studies, the low-up period was at least 9 months but ranged from 9 to
first clinical trial of teprotumumab for GO was published 27 months. During the study period, clinical activity scores
in 2017 [73]. The trial was a multicenter, randomized, decreased from 6.5 to 0.6, with specific improvements noted
placebo-controlled study of 88 patients who were rand- in eye motility, diplopia, and visual acuity. TRAb decreased
omized to receive teprotumumab or placebo infusion every by approximately 40% as well. Side effects were generally
3 weeks for a total of 8 IV infusions. Teprotumumab dose mild (fatigue, musculoskeletal complaints), and did not
was 10 mg/kg for the first dose, followed by 20 mg/kg for require discontinuation of therapy. Two patients developed
N. Genere, M. N. Stan

neutropenia and required a slight dose reduction. Atienza- be necessary to determine if these agents could play a role
Mateo et al. recently presented an abstract of 29 patients in the management of GO.
with similar improvements in disease activity and sever-
ity noted in a multicenter study of apparently open-label 6.4 Immunotherapy for Graves’ Disease
TCZ (reductions in CAS from 5.0 ± 1.7 to 1.0 ± 0.9 over with Potential Translation to GO
the course of 1 year) [77]. The first RCT with TCZ was
recently published, and after 4 monthly infusions of TCZ, A number of other agents aiming to reshape the immune
there was improvement in CAS by at least 2 points in 93% of response in GD and GO are making their way in early tri-
TCZ-treated patients compared with 59% of placebo-treated als. Most such agents are being tested first in Graves’ dis-
patients (CI 1.3–73.2) [78]. Proptosis also improved by a ease given the larger affected population, better understood
median of 1.5 mm (compared with 0 mm in the placebo pathophysiology as well as the more straightforward out-
group). The number of adverse effects was double in the come assessment. If found promising in Graves’ disease
TCZ group compared with placebo and 2 such events were therapy, the likely next step will be to test them for GO
deemed serious. Overall, TCZ appears to be quite promising given the pathophysiological similarities between the two
for corticosteroid-resistant disease if the data are confirmed disorders.
in subsequent studies. One such agent is CFZ533, an antibody that has been
tested in a number of autoimmune conditions (e.g. rheu-
6.3 Tumor Necrosis Factor α Inhibitors matoid arthritis, Sjögren’s syndrome, myasthenia gravis)
and in the field of organ transplantation due to its ability
There is evidence that early in the course of GO there is to interfere with CD40-CD154 interaction. The drug is
an abundance of tumor necrosis factor α (TNFɑ). This an Fc-silenced fully human anti-human CD40 monoclonal
cytokine is tied with the Th1 driven response that dominates antibody that is thus able to inhibit the activation of CD40
the immuno-pathophysiology of the disease at that stage. pathway. This pathway is essential for further activation
Etanercept, adalimumab and infliximab are all TNFα inhib- of humoral immunity after antigen presentation by antigen
itors that work by binding the eponymous cytokine. This presenting cells (APC), therefore the hypothesis is that
class of medication had been approved for use in rheuma- CFZ533 will be able to inhibit germinal center formation,
toid arthritis, inflammatory bowel disease, and some other affinity maturation and, consequently, antigen-specific
autoimmune conditions. Available literature on the use of memory B cell generation. CFZ533 has been tested in
these agents in GO is limited, without any RCT performed Graves’ disease in an open label fashion (ClinicalTri-
or planned, to our knowledge. Ten patients with active, als.gov Identifier: NCT02713256) and 15 patients were
mild-to-moderate GO were treated with etanercept 25 mg included in the results reported on Clinicaltrials.gov. The
twice weekly for 12 weeks [79]. While on treatment, CAS primary outcome was normalization of thyroid-stimulat-
decreased from a mean of 4 to 1.6 at 12 weeks; however, 3 ing hormone (TSH) at 12 weeks—that outcome was not
of the 10 patients developed recurrence of symptoms after achieved by any of participants. However, the trial found
discontinuation of the drug. Use of adalimumab was ret- that one-third of participants did have improvement in T3
rospectively reviewed in 10 patients with GO onset within and T4 values at the same time point and there was only
9 months of the start of therapy [80]. Patients were included one case with a serious adverse event. A full publication
if they had at least 1 sign of disease activity, and most were of the trial results is expected in the near future.
treated with concurrent moderate dose IV glucocorticoids A more specific approach to reversing the immune abnor-
for the first 6 weeks of adalimumab therapy. After 12 weeks malities associated with Graves’ disease/GO is being pur-
of therapy, there were reduced inflammatory signs in 6 of sued by a group of investigators in the UK that are attempt-
10 patients, and increased inflammation in 3 of the patients. ing to harness the action of regulatory T cells (T-regs).
One patient was admitted to the hospital with sepsis and Once activated through the antigen presentation process,
diarrhea. The applicability of these finding is unclear; some these cells are able to suppress the immune response tar-
patients experienced worsening symptoms on therapy, and geting the antigen they have been exposed to, in this case
those who improved may have done so because of the con- the TSH receptor. The product being tested is ATX-GD-59
comitant GCs, adalimumab or natural history of the disease. (ClinicalTrials.gov Identifier: NCT02973802), which repre-
Finally, the only report of infliximab was a case of its use in sents a mixture of three peptides derived from the structure
a patient with DON, which resulted in a decrease in clini- of the TSH receptor. The mixture has been administered
cal activity score and improvements in vision [81]. Overall, intradermally in a manner similar to a vaccine preparation,
the use of TNFα inhibitors has resulted in a questionable but the formulation does not include the typical stimulating
benefit regarding the inflammatory status, no evidence of adjuvants that are present in vaccines and thus the hope is
benefit regarding disease severity, and further trials would that only T-regs will be activated, with the expected result
Current and Emerging Treatment Strategies for Graves’ Orbitopathy

being suppression of TSHr antibody production. The trial of full closure of the eye. Treatments include lubrication,
was completed in February, 2018, and the results are yet to topical antibiotics, as well as surgical procedures to cover
be released. To take this approach a step further, there are the area of ulceration. Orbital decompression may be nec-
investigators who are looking at a more controlled exposure essary if proptosis is the primary reason for corneal expo-
of T-regs to a specific antigen. The so-called “training” of sure. Dysthyroid optic neuropathy is defined by stretching
T-regs would occur in-vitro after the cells have been col- of the optic nerve as it becomes compressed by surrounding
lected from a patient, and they would then be exposed to the extraocular muscles. Current management consists of the
antigen that is known to be involved in the pathogenesis of use of high doses of IV glucocorticoids (500–1000 mg meth-
the disease, specifically TSH receptor in the case of Graves’ ylprednisolone daily for 3 consecutive or alternating days),
disease/GO. Re-administering the cells would be expected with a repeat course in 1 week if clinically indicated [5]. If
to lead to an immune suppression specific only to the antigen no improvement is seen with GC treatment, or there is rapid
that was targeted in the “training” phase; thus, limiting the deterioration, then decompressive surgery should be pursued
risk of infectious complications. Ongoing studies are testing immediately. Globe subluxation is caused by expansion of
various methodologies that could lead to the desired speci- the orbital content with immediate threatening impact on
ficity of the “training” phase [82]. the function of the optic nerve and integrity of the cornea.
Another specific mechanism for targeting the pathophysi- In this case the treatment is exclusively surgical, needs to be
ology of Graves’ disease and presumably GO is to “mask” instituted on an emergent basis and ranges from tarsorrhaphy
the TSH receptor from the action of thyroid-stimulating to orbital decompression.
immunoglobulin (TSI). This is being pursued with the use
of a TSH receptor antagonist, K1-70 [83]. This agent
was developed as a monoclonal antibody and is now being 8 Management of Inactive GO
tested in an open-label clinical trial (NCT02904330) that
will assess both safety and dosing regimen for a larger trial Patients with inactive GO lack the acute inflammation of
focusing on the efficacy of controlling hyperthyroidism in active disease, and therefore do not benefit from immu-
Graves’ disease and hopefully in reversing ocular changes nomodulatory therapies previously described [5]. Mild
in GO. disease can be managed with local measures, but more sig-
nificant GO often requires rehabilitative surgeries. Waiting
6.5 Revisiting Some Old Agents for at least 6 months of inactive disease is very important,
as this helps to establish a new clinical baseline and can
Utilizing some well-known drugs in a different manner reduce the number of rehabilitative surgeries needed. The
might also offer rewards in GO. A different modality of recommended surgical progression is to start with orbital
delivering glucocorticoids, liposomal encapsulated pred- decompression, followed by extraocular muscle surgery, and
nisolone, could potentially reduce the systemic impact of then followed by eyelid procedures. Discussion of specific
cumulative glucocorticoid dose and avoid adverse effects of surgical considerations and procedures is beyond the scope
chronic bolus glucocorticoid use. A Phase II clinical study of this review.
is currently recruiting GO patients (EudraCT Number:
2017-001158-33 s). Statins have been found to improve GO
outcome in a retrospective cohort and a prospective trial is
about to test that hypothesis by comparing IVGC + atorvas- 9 Summary
tatin with IVGC = placebo, in active and moderate-to-severe
GO (NCT03110848) [84]. GO is a rare yet challenging disease that significantly
decreases the quality of life of affected patients. It should
be considered together with Graves’ disease, particularly
7 Management of Sight‑Threatening as some of the therapeutic tools tested for hyperthyroidism
Disease could easily be extrapolated to the eye pathology. The initial
evaluation of all patients with GD should include an evalu-
Corneal ulceration, dysthyroid optic neuropathy, and globe ation for GO and, when present, particular attention should
subluxation are sight-threatening emergencies that require be paid to disease activity, disease severity, and the impact
immediate treatment [85]. Clinicians must evaluate for sight- on the patient’s quality of life. These elements will shape the
threatening complications at the time of the patient’s presen- therapeutic approach which should be pursued in a multi-
tation and throughout a given treatment course. Sight-threat- disciplinary manner, with the endocrinologist and the oph-
ening corneal breakdown occurs due to excessive corneal thalmologists operating ideally under a combined Thyroid
exposure due to proptosis and lid retraction, as well as lack Eye Clinic form of environment. Here we have presented a
N. Genere, M. N. Stan

review of the current treatment paradigm for GO and have 10. Kahaly GJ, et al. Psychosocial morbidity of Graves’ orbitopathy.
highlighted the avenues of therapeutic research currently Clin Endocrinol (Oxf). 2005;63(4):395–402.
11. Fayers T, Dolman PJ. Validity and reliability of the TED-QOL:
under exploration as well as those noted on the horizon. All a new three-item questionnaire to assess quality of life in thy-
patients with GO benefit from risk factor modification and roid eye disease. Br J Ophthalmol. 2011;95(12):1670–4.
normalization of thyroid function tests. Patients with active, 12. Terwee CB, et al. Development of a disease specific quality of
mild disease generally benefit from local therapies and sele- life questionnaire for patients with Graves’ ophthalmopathy: the
GO-QOL. Br J Ophthalmol. 1998;82(7):773–9.
nium, while patients with moderate-to-severe disease ben- 13. Bartley GB, et  al. The incidence of Graves’ ophthalmopa-
efit most from GC therapy. We have explored some of the thy in Olmsted County,Minnesota. Am J Ophthalmol.
second-line choices as well as the promising new agents that 1995;120(4):511–7.
aim to tackle the autoimmune mechanisms of GO, from the 14. Wiersinga WM, Bartalena L. Epidemiology and prevention of
Graves’ ophthalmopathy. Thyroid. 2002;12(10):855–60.
TSHr to the IGF-1R, from the cytokine-mechanisms like 15. Prummel MF, Wiersinga WM. Smoking and risk of Graves’
IL-6R and TNF-alpha to the Tregs and beyond. The future disease. JAMA. 1993;269(4):479–82.
will hopefully allow us to better understand the benefits of 16. Xing L, et al. Smoking was associated with poor response to
each individual approach as well as the biological “price” intravenous steroids therapy in Graves’ ophthalmopathy. Br J
Ophthalmol. 2015;99(12):1686–91.
in adverse effects associated with each therapy. If we com- 17. Eckstein A, et al. Impact of smoking on the response to treat-
bine these with a better understanding of the mechanisms ment of thyroid associated ophthalmopathy. Br J Ophthalmol.
responsible for the heterogeneity of GO, we should be able 2003;87(6):773–6.
to match a particular intervention with the GO subgroup 18. Bartalena L, et al. Cigarette smoking and treatment outcomes in
Graves ophthalmopathy. Ann Intern Med. 1998;129(8):632–5.
that is most suited for that approach, the goal of the modern 19. Pfeilschifter J, Ziegler R. Smoking and endocrine ophthalmopa-
individualized medicine. thy: impact of smoking severity and current vs lifetime cigarette
consumption. Clin Endocrinol (Oxf). 1996;45(4):477–81.
Compliance with Ethical Standards  20. Bahn Chair RS, et al. Hyperthyroidism and other causes of thy-
rotoxicosis: management guidelines of the American Thyroid
Association and American Association of Clinical Endocrinolo-
Conflict of interest  NG reports no conflicts of interest. MNS reports gists. Thyroid. 2011;21(6):593–646.
participating in Advisory Board meeting with Horizon Pharma, manu- 21. Prummel MF, et  al. Effect of abnormal thyroid function on
facturer of teprotumumab. the severity of Graves’ ophthalmopathy. Arch Intern Med.
1990;150(5):1098–101.
Funding  No funding was received for the preparation of this manuscript. 22. Ross DS, et al. 2016 American Thyroid Association Guidelines
for Diagnosis and Management of Hyperthyroidism and Other
Causes of Thyrotoxicosis. Thyroid. 2016;26(10):1343–421.
23. Bartalena L, et al. Relation between therapy for hyperthyroid-
References ism and the course of Graves’ ophthalmopathy. N Engl J Med.
1998;338(2):73–8.
1. Bahn RS. Graves’ ophthalmopathy. N Engl J Med. 24. Li HX, et  al. Relation between therapy options for Graves’
2010;362(8):726–38. disease and the course of Graves’ ophthalmopathy: a sys-
2. Eckstein AK, et  al. Euthyroid and primarily hypothyroid tematic review and meta-analysis. J Endocrinol Invest.
patients develop milder and significantly more asymmetrical 2016;39(11):1225–33.
Graves ophthalmopathy. Br J Ophthalmol. 2009;93(8):1052–6. 25. Tallstedt L, et al. Does early administration of thyroxine reduce
3. Rundle FF, Wilson CW. Development and course of exoph- the development of Graves’ ophthalmopathy after radioiodine
thalmos and ophthalmoplegia in Graves’ disease with spe- treatment? Eur J Endocrinol. 1994;130(5):494–7.
cial reference to the effect of thyroidectomy. Clin Sci. 26. Stan MN, et  al. Cohort study on radioactive iodine-induced
1945;5(3–4):177–94. hypothyroidism: implications for Graves’ ophthalmopathy
4. Bartley GB. Rundle and his curve. Arch Ophthalmol. and optimal timing for thyroid hormone assessment. Thyroid.
2011;129(3):356–8. 2013;23(5):620–5.
5. Bartalena L, et  al. The 2016 European Thyroid Associa- 27. Perros P, Crombie AL, Kendall-Taylor P. Natural history of
tion/European Group on Graves’ Orbitopathy Guidelines thyroid associated ophthalmopathy. Clin Endocrinol (Oxf).
for the Management of Graves’ Orbitopathy. Eur Thyroid J. 1995;42(1):45–50.
2016;5(1):9–26. 28. Grisolia ABD, et al. Non-surgical treatment for eyelid retraction
6. Barrio-Barrio J, et al. Graves’ ophthalmopathy: VISA versus in thyroid eye disease (TED). Br J Ophthalmol. 2018;102:158–63.
EUGOGO classification, assessment, and management. J Oph- 29. Uddin JM, Davies PD. Treatment of upper eyelid retraction asso-
thalmol. 2015;2015:249125. ciated with thyroid eye disease with subconjunctival botulinum
7. Mourits MP, et al. Clinical activity score as a guide in the man- toxin injection. Ophthalmology. 2002;109(6):1183–7.
agement of patients with Graves’ ophthalmopathy. Clin Endo- 30. Morgenstern KE, et  al. Botulinum toxin type a for dysthy-
crinol (Oxf). 1997;47(1):9–14. roid upper eyelid retraction. Ophthalmic Plast Reconstr Surg.
8. Tanda ML, et  al. Prevalence and natural history of Graves’ 2004;20(3):181–5.
orbitopathy in a large series of patients with newly diagnosed 31. Kohrle J. Selenium and the thyroid. Curr Opin Endocrinol Diabe-
graves’ hyperthyroidism seen at a single center. J Clin Endo- tes Obes. 2015;22(5):392–401.
crinol Metab. 2013;98(4):1443–9. 32. Marcocci C, et al. Selenium and the course of mild Graves’ orbit-
9. Ponto KA, et al. Public health relevance of Graves’ orbitopathy. opathy. N Engl J Med. 2011;364(20):1920–31.
J Clin Endocrinol Metab. 2013;98(1):145–52.
Current and Emerging Treatment Strategies for Graves’ Orbitopathy

33. Dehina N, et al. Lack of association between selenium status and 52. Stan MN, et  al. Randomized controlled trial of rituximab in
disease severity and activity in patients with graves’ ophthalmopa- patients with Graves’ orbitopathy. J Clin Endocrinol Metab.
thy. Eur Thyroid J. 2016;5(1):57–64. 2015;100(2):432–41.
34. Gao G, et  al. Meta-analysis of methylprednisolone pulse 53. Stan MN, Salvi M. Management of endocrine disease: rituximab
therapy for Graves’ ophthalmopathy. Clin Exp Ophthalmol. therapy for Graves’ orbitopathy—lessons from randomized con-
2014;42(8):769–77. trol trials. Eur J Endocrinol. 2017;176(2):R101–9.
35. Stiebel-Kalish H, et al. Treatment modalities for Graves’ ophthal- 54. Kahaly G, et al. Ciclosporin and prednisone v. prednisone in treat-
mopathy: systematic review and metaanalysis. J Clin Endocrinol ment of Graves’ ophthalmopathy: a controlled, randomized and
Metab. 2009;94(8):2708–16. prospective study. Eur J Clin Invest. 1986;16(5):415–22.
36. Kahaly GJ, et al. Randomized, single blind trial of intravenous 55. Prummel MF, et al. Prednisone and cyclosporine in the treat-
versus oral steroid monotherapy in Graves’ orbitopathy. J Clin ment of severe Graves’ ophthalmopathy. N Engl J Med.
Endocrinol Metab. 2005;90(9):5234–40. 1989;321(20):1353–9.
37. Marcocci C, et al. Fatal and non-fatal adverse events of gluco- 56. Ye X, et  al. Efficacy and safety of mycophenolate mofetil in
corticoid therapy for Graves’ orbitopathy: a questionnaire sur- patients with active moderate-to-severe Graves’ orbitopathy. Clin
vey among members of the European Thyroid Association. Eur J Endocrinol (Oxf). 2017;86(2):247–55.
Endocrinol. 2012;166(2):247–53. 57. Riedl M, et al. Prospective, systematically recorded mycophe-
38. Stan MN, Garrity JA, Bahn RS. The evaluation and treatment of nolate safety data in Graves’ orbitopathy. J Endocrinol Invest.
graves ophthalmopathy. Med Clin North Am. 2012;96(2):311–28. 2016;39(6):687–94.
39. Bartalena L, et al. Efficacy and safety of three different cumu- 58. Kahaly GJ, et al. Mycophenolate plus methylprednisolone versus
lative doses of intravenous methylprednisolone for moderate to methylprednisolone alone in active, moderate-to-severe Graves’
severe and active Graves’ orbitopathy. J Clin Endocrinol Metab. orbitopathy (MINGO): a randomised, observer-masked, multicen-
2012;97(12):4454–63. tre trial. Lancet Diabetes Endocrinol. 2018;6(4):287–98.
40. He Y, et al. Comparison of two different regimens of intrave- 59. Rivera-Grana E, et  al. Methotrexate as a corticosteroid-spar-
nous methylprednisolone for patients with moderate to severe ing agent for thyroid eye disease. J Clin Exp Ophthalmol.
and active Graves’ ophthalmopathy: a prospective, randomized 2015;6(2):422.
controlled trial. Endocr J. 2017;64(2):141–9. 60. Rubinov A, et al. Role of methotrexate in thyroid-related orbit-
41. Zhu W, et al. A prospective, randomized trial of intravenous opathy. Can J Ophthalmol. 2018;53(1):34–8.
glucocorticoids therapy with different protocols for patients 61. Strianese D, et al. Methotrexate for the treatment of thyroid eye
with graves’ ophthalmopathy. J Clin Endocrinol Metab. disease. J Ophthalmol. 2014;2014:128903.
2014;99(6):1999–2007. 62. Sipkova Z, et al. Early use of steroid-sparing agents in the inac-
42. Bartalena L, et al. Does early response to intravenous gluco- tivation of moderate-to-severe active thyroid eye disease: a step-
corticoids predict the final outcome in patients with moderate- down approach. Clin Endocrinol (Oxf). 2018;89(6):834–9.
to-severe and active Graves’ orbitopathy? J Endocrinol Invest. 63. Perros P, et al. Azathioprine in the treatment of thyroid-associated
2017;40(5):547–53. ophthalmopathy. Acta Endocrinol (Copenh). 1990;122(1):8–12.
43. Gorman CA, et al. A prospective, randomized, double-blind, 64. Rajendram R, et al. Combined immunosuppression and radio-
placebo-controlled study of orbital radiotherapy for Graves’ oph- therapy in thyroid eye disease (CIRTED): a multicentre, 2 × 2
thalmopathy. Ophthalmology. 2001;108(9):1523–34. factorial, double-blind, randomised controlled trial. Lancet Dia-
44. Prummel MF, et al. A randomized controlled trial of orbital radi- betes Endocrinol. 2018;6(4):299–309.
otherapy versus sham irradiation in patients with mild Graves’ 65. Baschieri L, et al. Intravenous immunoglobulin versus corti-
ophthalmopathy. J Clin Endocrinol Metab. 2004;89(1):15–20. costeroid in treatment of Graves’ ophthalmopathy. Thyroid.
45. Mourits MP, et  al. Radiotherapy for Graves’ orbit-
1997;7(4):579–85.
opathy: randomised placebo-controlled study. Lancet. 66. Kahaly G, et al. Randomized trial of intravenous immunoglobulins
2000;355(9214):1505–9. versus prednisolone in Graves’ ophthalmopathy. Clin Exp Immu-
46. Bradley EA, et al. Orbital radiation for graves ophthalmopathy: a nol. 1996;106(2):197–202.
report by the American Academy of Ophthalmology. Ophthalmol- 67. Dickinson AJ, et al. Double-blind, placebo-controlled trial of
ogy. 2008;115(2):398–409. octreotide long-acting repeatable (LAR) in thyroid-associated
47. Ng CM, et al. Combined orbital irradiation and systemic ster- ophthalmopathy. J Clin Endocrinol Metab. 2004;89(12):5910–5.
oids compared with systemic steroids alone in the management 68. Chang TC, Liao SL. Slow-release lanreotide in Graves’ ophthal-
of moderate-to-severe Graves’ ophthalmopathy: a preliminary mopathy: a double-blind randomized, placebo-controlled clinical
study. Hong Kong Med J. 2005;11(5):322–30. trial. J Endocrinol Invest. 2006;29(5):413–22.
48. Bartalena L, et al. Orbital cobalt irradiation combined with sys- 69. Stan MN, et al. Randomized, double-blind, placebo-controlled
temic corticosteroids for Graves’ ophthalmopathy: comparison trial of long-acting release octreotide for treatment of Graves’
with systemic corticosteroids alone. J Clin Endocrinol Metab. ophthalmopathy. J Clin Endocrinol Metab. 2006;91(12):4817–24.
1983;56(6):1139–44. 70. Wemeau JL, et al. Octreotide (long-acting release formulation)
49. Marcocci C, et al. Orbital radiotherapy combined with high dose treatment in patients with graves’ orbitopathy: clinical results of a
systemic glucocorticoids for Graves’ ophthalmopathy is more four-month, randomized, placebo-controlled, double-blind study.
effective than radiotherapy alone: results of a prospective rand- J Clin Endocrinol Metab. 2005;90(2):841–8.
omized study. J Endocrinol Invest. 1991;14(10):853–60. 71. Douglas RS, et al. B cells from patients with Graves’ disease
50. Oeverhaus M, et al. Combination Therapy of Intravenous Ster- aberrantly express the IGF-1 receptor: implications for disease
oids and Orbital Irradiation is More Effective Than Intravenous pathogenesis. J Immunol. 2008;181(8):5768–74.
Steroids Alone in Patients with Graves’ Orbitopathy. Horm Metab 72. Tsui S, et al. Evidence for an association between thyroid-stim-
Res. 2017;49(10):739–47. ulating hormone and insulin-like growth factor 1 receptors: a
51. Salvi M, et al. Efficacy of B-cell targeted therapy with rituxi- tale of two antigens implicated in Graves’ disease. J Immunol.
mab in patients with active moderate to severe Graves’ orbitopa- 2008;181(6):4397–405.
thy: a randomized controlled study. J Clin Endocrinol Metab. 73. Smith TJ, et al. Teprotumumab for thyroid-associated ophthal-
2015;100(2):422–31. mopathy. N Engl J Med. 2017;376(18):1748–61.
N. Genere, M. N. Stan

74. Kumar S, Bahn RS. Relative overexpression of macrophage- 80. Ayabe R, et  al. Adalimumab as steroid-sparing treatment of
derived cytokines in orbital adipose tissue from patients inflammatory-stage thyroid eye disease. Ophthalmic Plast Recon-
with graves’ ophthalmopathy. J Clin Endocrinol Metab. str Surg. 2014;30(5):415–9.
2003;88(9):4246–50. 81. Durrani OM, Reuser TQ, Murray PI. Infliximab: a novel treatment
75. Smolen JS, et al. Effect of interleukin-6 receptor inhibition with for sight-threatening thyroid associated ophthalmopathy. Orbit.
tocilizumab in patients with rheumatoid arthritis (OPTION study): 2005;24(2):117–9.
a double-blind, placebo-controlled, randomised trial. Lancet. 82. Dawson NAJ, Vent-Schmidt J, Levings MK. Engineered toler-
2008;371(9617):987–97. ance: tailoring development, function, and antigen-specificity of
76. Perez-Moreiras JV, Alvarez-Lopez A, Gomez EC. Treatment of regulatory T cells. Front Immunol. 2017;8:1460.
active corticosteroid-resistant graves’ orbitopathy. Ophthalmic 83. Sanders P, et al. Crystal structure of the TSH receptor (TSHR)
Plast Reconstr Surg. 2014;30(2):162–7. bound to a blocking-type TSHR autoantibody. J Mol Endocrinol.
77. Atienza-Mateo BCRV, Martín-Varillas J, et al. SAT0601 Anti-il6- 2011;46(2):81–99.
receptor tocilizumab in graves’ orbitopathy. multicenter study of 84. Stein JD, et al. Risk factors for developing thyroid-associated
29 patients. Ann Rheumatic Dis. 2018;77:1153–4. ophthalmopathy among individuals with Graves disease. JAMA
78. Perez-Moreiras JV, et al. Efficacy of tocilizumab in patients with Ophthalmol. 2015;133(3):290–6.
moderate to severe corticosteroid resistant Graves orbitopathy: a 85. Bahn RS, Bartley GB, Gorman CA. Emergency treatment of
randomized clinical trial. Am J Ophthalmol. 2018;195:181–90. Graves’ ophthalmopathy. Baillieres Clin Endocrinol Metab.
79. Paridaens D, et al. The effect of etanercept on Graves’ ophthal- 1992;6(1):95–105.
mopathy: a pilot study. Eye (Lond). 2005;19(12):1286–9.

You might also like