Wang-Douglas2020 ReferenceWorkEntry ThyroidEyeDisease

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 21

Thyroid Eye Disease

Yao Wang and Raymond S. Douglas

Contents
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
Thyroid Dysfunction in Graves’ Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
Thyroid Eye Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
Epidemiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
Risk Factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
Molecular Pathogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
Clinical Course of TED . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
Imaging Techniques . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13
Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14

Abstract autoimmune thyroid disease. Clinical manifes-


Thyroid eye disease (TED) can be a debilitat- tations commonly include eyelid retraction,
ing disorder that occurs in the setting of exposure keratitis, exophthalmos, and restric-
tive strabismus. Patients frequently complain
of dry eyes, photophobia, and excessive lacri-
mation. While the exact relationship between
the thyroid and orbit is not fully understood,
Y. Wang
Department of Surgery, Division of Ophthalmology, recent research has shown that orbital tissues
Cedars Sinai Medical Center, Los Angeles, CA, USA are infiltrated by immunocompetent cells
Beverly Hills Ophthalmic Plastic and Reconstructive which subsequently causes orbital extraocular
Surgery, Beverly Hills, CA, USA muscle and connective tissue expansion.
e-mail: wangx699@umn.edu Numerous cell types and molecular mediators
R. S. Douglas (*) have been found to be involved in its patho-
Department of Surgery, Division of Ophthalmology, genesis, and early trials targeting various
Cedars Sinai Medical Center, Los Angeles, CA, USA
immunomodulators as disease-modifying ther-
Beverly Hills Ophthalmic Plastic and Reconstructive apy have shown promising results.
Surgery, Beverly Hills, CA, USA
State Key Laboratory of Ophthalmology, Zhongshan
Ophthalmic Center, Sun Yat-sen University, Guangzhou,
China

© Springer Nature Switzerland AG 2020 1


D. M. Albert et al. (eds.), Albert and Jakobiec's Principles and Practice of Ophthalmology,
https://doi.org/10.1007/978-3-319-90495-5_61-1
2 Y. Wang and R. S. Douglas

Keywords chapter, we describe the thyroid dysfunction in


GD, and the pathogenesis, diagnosis, and clinical
Thyroid eye disease · Graves’
course of TED.
ophthalmopathy · Graves’ disease

Thyroid Dysfunction in Graves’


Introduction
Disease
Graves’ disease (GD) is an autoimmune condition
Thyroid hormone overproduction in GD results
characterized by dysregulated thyroid gland activ-
from the imbalanced actions of activating (most
ity. In North America, its incidence of GD is
common) and blocking antibodies directed
~0.4% [1]. An age-adjusted incidence rate is 16
against the thyrotropin receptor (TSHR) [28–30].
cases and 2.9 cases per 100,000 population per
This G-protein-coupled receptor is displayed on
year, respectively, for women and men [1]. Esti-
the surface of thyroid epithelial cells where it
mates of the female-to-male ratio have ranged
ordinarily binds to and becomes activated by
from 2.5:1 to 7:1, with a ratio of 4:1 cited in
TSH from the anterior pituitary. In GD, anti-
most studies [2–7]. Individuals with GD are also
TSHR antibodies are detectable in nearly all
more likely than the general population to develop
patients with hyperthyroidism [28, 31, 32].
other autoimmune diseases including insulin-
Clinical presentation of hyperthyroidism might
dependent diabetes mellitus [8–11], rheumatoid
include weight loss (despite normal or increased
arthritis [12–14], pernicious anemia [15], and vit-
appetite), heat intolerance, menstrual irregulari-
iligo [16, 17].
ties, infertility, palpitations, emotional lability,
The pathogenesis of GD involves stimulatory
hyperdefecation, and tremor. Increased incidence
autoantibodies directed against thyrotropin recep-
of atrial fibrillation and accelerated bone loss are
tor (TSHR), resulting in glandular enlargement
among the consequences of elevated circulating
and overproduction of thyroid hormones [18].
thyroid hormone levels, especially in older
GD consists of both systemic and anatomically
patients. The most sensitive laboratory assess-
localized immune responses. Manifestations
ment of thyrotoxicosis remains a depressed
include hyperthyroid goiter, dermopathy, and thy-
serum TSH concentration coupled with elevated
roid eye disease (TED) [19]. While TED is typi-
thyroxine and triiodothyronine levels and
cally associated with GD, it can manifest in
increased radioactive iodine (RAI) uptake in the
primary hypothyroidism, Hashimoto thyroiditis,
thyroid [33]. The presence of high titers of anti-
or euthyroidism [19]. While the clinical course
TSHR antibodies usually confirms the diagnosis
of glandular GD is usually predictable and can
of GD.
be easily treated, TED is not. Specific and safe
With regard to therapy, RAI is a definitive
therapies are limited, in large part because of poor
treatment in the United States for most patients
insight into disease pathogenesis. Moreover, the
over 21 years. Following administration, the vol-
relationship between thyroid gland dysfunction
ume of the thyroid gland is often reduced [34].
and TED remains unclear.
The most common outcome of RAI therapy is
Clinically important TED occurs in approxi-
irreversible hypothyroidism, which develops in
mately 10–45% of patients with GD, with clinical
the majority of adequately treated patients [35,
presentations ranging from mild ocular irritation
36]. Another approach involves the surgical
to severe proptosis, strabismus, and optic nerve
removal of thyroid tissue, which, in the hands of
compression [19–21]. The proximate causes of
a skilled surgeon, can be performed safely and
TED are probably unrelated to thyrotoxicosis
effectively.
[22–27]. Rather, it is driven by an underlying
autoimmune process. The complex pathogenesis
makes the management of TED difficult. In this
Thyroid Eye Disease 3

Thyroid Eye Disease studies have shown that RAI worsened TED coin-
ciding appearance of TSI [61, 62]. There is a
Epidemiology relative risk of 4.23 (95% CI 2.04–8.77) for devel-
oping TED following RAI compared to anti-thy-
The prevalence of TED is more evenly distributed roid medication [63]. The resultant damage from
among men and women than is GD [3, 37]. TED RAI could potentiate further release of thyroid
most commonly presents during the fourth and antigens, which in turn, increases production of
fifth decades of life, which suggests that as yet antibodies toward TSH-R [64]. Concomitant cor-
unidentified acquired factors might contribute to ticosteroid use (oral prednisone of 0.3–0.5 mg/kg/
susceptibility (Fig. 1). In one large series, the age day with a 3-month total course) was found to be
range was 15–86 years, with a median of 52 years; an effective prophylaxis [62]. Later studies con-
however, TED may occur at any age and, albeit firmed that a 6-week course of 0.2 mg/kg per day
extremely rarely, even in neonates [20, 38–40]. was equally sufficient [65–67].
The most common cause of bilateral exoph-
thalmos is TED, as it accounts for approximately
85% of cases [41]. TED also presents as unilateral Molecular Pathogenesis
exophthalmos, accounting for 15–28% of cases
[42, 43]. Although many patients are thought to The pathogenesis of TED results from infiltration
have unilateral disease on the basis of clinical of tissues by immunocompetent cells early in the
findings, the majority of patients actually show disease process and subsequent orbital volume
bilateral disease radiographically [44]. expansion. The infiltrate consists of predomi-
nantly CD4+ T lymphocytes exhibiting an acti-
vated memory phenotype and numerous
Risk Factors granulated mast cells in the extraocular muscle
(EOM) interstitial tissue, orbital fat, and connec-
Cigarette smoking has been considered the stron- tive tissue (Fig. 2) [30, 68, 69]. Interferon (IFN)-g,
gest risk factor for developing TED [45–49]. The tumor necrosis factor (TNF)-a, and interleukin
odds ratio relative to controls is as high as 20.2 for (IL)-1 have been detected adjacent to mononu-
active smokers and 8.9 for former smokers, clear cell infiltrates, suggesting cytokine produc-
suggesting both direct and secondary effects of tion by these cells [70]. Local production of both
smoking [50, 51]. Patients with TED who are Th1 and Th2 cytokines appears likely, and the
smokers also have more severe eye disease [48, balance between cytokine class predominance
52]. Additionally, the risk of developing TED is may change at different disease stages [71–74].
associated with the number of cigarettes smoked Th1 responses appear to predominate in patients
following the diagnosis of GD, rather than the with early disease while the Th2 pattern might
cumulative pack-years [51]. Smokers also had become more abundant later [75].
less therapeutic response to orbital radiation or The basis for many of the clinical symptoms
corticosteroids [48]. Smoking can worsen several and signs of TED can be reconciled with increased
other autoimmune diseases including rheumatoid volume of EOM and orbital fatty connective tis-
arthritis [53, 54] and Crohn’s disease [55], but the sue (Fig. 3) [20, 76]. Increased orbital volume
mechanisms remain unclear. Serum cytokine eventually displaces the globe forward, decreases
levels do not differ substantially in smokers and venous drainage, restricts extraocular movement,
nonsmokers with TED [56–59]. and can cause compressive optic neuropathy [77].
As previously mentioned, RAI is a common Some patients demonstrate predominant enlarge-
modality used to treat hyperthyroidism in GD. ment of the EOM (Fig. 4), while others exhibit
There is a transient increase of TSHR antibodies increased orbital adipose volume with little EOM
(TRAb) and thyroid stimulating immunoglobulin abnormality (Fig. 5) [78–80]. Intact muscle fibers
(TSI) following RAI treatment [60]. Numerous are widely separated by edematous connective
4 Y. Wang and R. S. Douglas

Fig. 1 Age distribution of


500 patients with
hyperthyroidism (a)
compared with that of 194
patients who underwent
orbital decompression for
severe orbitopathy (b).
(From Jacobson and
Gorman [140], 5:200–220.)

Fig. 2 Pathologic changes early in the course of thyroid lymphoid infiltrate and increased intracellular volume.
eye disease. In the acute phase, a lymphocytic inflamma- (From Jakobiec FA, Font RL: Orbit. In: Spencer WH, ed.
tory infiltrate predominates. This may be focal and Ophthalmic pathology. 3rd edn. Philadelphia: WB
involves the endomysial connective tissue. Lower (a) and Saunders; 1986.)
higher (b) magnification H & E staining. Note the dense

tissue in early disease [81]. The perimysial tissue Orbital Fibroblasts


contains excessive glycosaminoglycans (GAGs) Fibroblasts both respond to and produce numerous
composed of hyaluronan and chondroitin sulfate molecular mediators that serve to activate and mod-
[82]. It is only late in the disease that the fibers ulate the behavior of bone marrow-derived cells and
become damaged, in large part from extensive provoke their migration to sites of inflammation
fibrosis that often follows aggressive inflamma- (Fig. 7) [90, 91]. The unique phenotype of orbital
tion (Fig. 6). The GAGs also accumulates in fatty fibroblasts may underlie the isolated involvement of
connective tissue [83–85]. The fibroblast is an the orbit in TED [92]. Orbital fibroblasts exhibit a
important source of hyaluronan [86–89]. The distinct morphology in vitro, display receptors and
extraordinary water binding capacity of gangliosides, and generate macromolecular compo-
hyaluronan attracts water and accounts, at least nents of the extracellular matrix differently from
in part, for the expansion of tissues. other fibroblasts [89, 93–96].
Thyroid Eye Disease 5

Fig. 3 Enlarged
extraocular muscles. These
are the exenterated orbital
contents from a recently
diagnosed patient. Orbital
fat has been dissected away
to demonstrate the
extraocular muscles. (From
Hufnagel et al. [84],
91:1411–1419.)

Fig. 4 Axial CT image of a


patient with enlarged
nontendinous portion of
extraocular muscles. The
equator of the globe is
anterior to the lateral orbital
rim, indicating proptosis

There are two subpopulations of orbital fibro- Fibrocytes


blasts [97]. A subset of orbital fibroblasts from the Orbital fibroblasts appear to be derived to
adipose/connective tissue (~50%) express Thy-1, fibrocytes from the bone marrow [99]. These pro-
a surface glycoprotein marker [98]. In contrast, genitor cells are from monocyte and B cell line-
those associated with EOM uniformly display ages [99]. They express hematopoietic stem cell
Thy-1 [97]. Thy1-expressing orbital fibroblasts marker CD34, CD45, and alpha-smooth muscle
can differentiate into myofibroblasts [93]. Thy1- actin, collagen I and III, fibronectin, and vimentin
deficient orbital fibroblasts can differentiate into [100]. Fibrocytes comprise of only 0.5% of circu-
adipocytes [97]. The demonstration of adipogenic lating peripheral blood mononuclear cells. They
potential in Thy-1 deficient orbital fibroblasts has are more abundant in patients with GD compared
substantial implications for the pathogenesis of to controls [99]. They can traffic to sites of inflam-
TED, since expansion of orbital fat is a prominent mation and are responsible for tissue remodeling
feature of the disease. [101]. When activated, they produce cytokines
and chemokines similar to that of activated orbital
6 Y. Wang and R. S. Douglas

Fig. 5 Occasionally, patients with thyroid eye disease Increased orbital fat volume with minimal enlargement of
(TED) exhibit exophthalmos reflecting increased orbital extraocular muscles. The equator of the proptotic left globe
fat volume. (a) Euthyroid patient with TED with 4-mm is anterior to the lateral orbital rim
proptosis of the left eye and left lower eyelid retraction. (b)

Fig. 6 Pathologic changes


late in the course of thyroid
eye disease. H & E staining
demonstrating progressive
fibrosis of extraocular
muscle occurring late in the
disease. Note that the entire
muscle has been replaced
by fibrous connective tissue
with a scattering of
mononuclear inflammatory
cells

fibroblasts [102] and can differentiate into weaker but significant correlation was established
myofibroblasts and adipocytes [101]. between antibody levels and proptosis [184].
In addition to its expression on the thyroid
Thyrotropin Receptor (TSHR) epithelium, TSHR has been found to be present
A feature common to nearly all individuals with on many different cell types throughout the body.
GD is the presence of activating anti-TSHR anti- Further, TSHR mRNA has been found in orbital
bodies [103, 104]. This appears true for individ- tissues of patients with GD as well as healthy
uals with and without clinically important TED. controls. Fibroblasts from all anatomic regions
When TSHR was initially cloned and character- appear to express similar relatively low levels of
ized, several investigators attempted to insinuate TSHR [109–112]. Moreover, treatment of orbital
this receptor in the pathogenesis of TED. Several fibroblasts with extremely high concentrations of
studies have examined the relationship between TSH has failed to elicit convincing and biologi-
TSHR antibodies and TED [105–108]. Gerding et cally meaningful responses [113]. On the other
al. reported that TSI levels are positively corre- hand, although fibrocytes from both patients
lated with clinical activity score (CAS), while with TED and healthy controls, the fraction of
Thyroid Eye Disease 7

Fig. 7 Schematic of the


complex interaction
between bone marrow-
derived cells and orbital
fibroblasts. These
fibroblasts exhibit
exaggerated responses to
and production of
proinflammatory mediators.
When activated by
cytokines, they mediate
immune infiltration,
inflammation, fibrosis,
hyaluronan accumulation,
and tissue remodeling

fibrocytes expressing TSHR is increased in Insulin-Like Growth Factor-I (IGF-I)


patients with TED [102]. When orbital fibroblasts Fibroblasts from patients with GD respond to
expressing Thy1 were treated with TSH or M22 disease-specific IgGs (GD-IgG). Rotella et al.
(stimulatory TSHR antibody) or were subjected to reported that GD-IgG from several patients with
adipocyte differentiating factors, there was TED could enhance collagen synthesis in normal
enhanced adipocyte differentiation [114]. To human dermal (arm) fibroblasts [121]. Sera and
date, the exact role of this autoantigen in TED GD-IgG from these patients induced the expres-
remains uncertain. sion of IL-16 and in disease-derived fibroblasts
but not in those from control donors [122]. IL-16,
Cytokines a CD4-specific chemoattractant cytokine, has
Orbital tissue remodeling in TED appear to be due been implicated in a number of human autoim-
to cytokine-depending cytokine activation [115]. mune diseases, including rheumatoid arthritis
Evidence of numerous cytokines, including IFNγ, [123–126], inflammatory bowel disease [127,
TNFα, IL-1α, IL-1β, IL-4, IL-6, IL-10, and IL-2R, 128], and lupus erythematosus [129, 130].
have been detected in orbital fat and extraocular RANTES is a potent chemokine with pro-
muscle in patients with TED [70, 72, 116]. Fur- inflammatory properties. It has also been impli-
ther, serum IL-6 and IL-6R was found to be ele- cated in autoimmunity and can be detected in the
vated in patients with GD and even higher in thyroid gland of patients with GD [131–133].
patients with TED [57]. The GD-IgG provoking IL-16 and RANTES
Tocilizumab, a recombinant humanized mono- expression in fibroblasts is directed against the
clonal antibody directed against the IL-6 receptor, insulin-like growth factor-1 receptor (IGF-IR)
has been used to successfully treat patients with [91, 122]. IGF-IR is a membrane-spanning tyro-
rheumatologic conditions, such as rheumatoid sine kinase protein, which can be activated by
arthritis and giant cell arteritis [117]. Early studies insulin or insulin like growth factor-I (IGF-I)
have shown improvement in TED severity and [134]. A disproportionate fraction of fibroblasts
activity in patients on tocilizumab [118–120]. derived from patients with GD display IGF-1R
which appears to mediate the response [91]. This
same GD-IgG enhances the production of
hyaluronan by GD orbital fibroblasts [135].
8 Y. Wang and R. S. Douglas

Blocking fibroblast signaling through the mTOR/ p < 0.001) at 24 weeks. TED quality-of-life
FRAP/Akt/p70S6K pathway with rapamycin can score also improved in a greater proportion of
attenuate the induction of IL-16 and hyaluronan patients receiving teprotumumab compared to
production [122, 135]. Thus, strong evidence has placebo at 24 weeks (overall 15.55 vs. 5.92,
been generated for IGF-1R and activating anti- p < 0.001).
bodies directed against the receptor playing Teprotumumab was well tolerated in both the
potentially important roles in the pathogenesis of Phase 2 and Phase 3 trials [138, 139]. The most
TED. common side effects were muscle spasms, nausea,
IGF-IR and TSHR may also work together in and alopecia. Mild cases of hyperglycemia were
the pathogenesis of TED. The relationship detected in several patients with preexisting glu-
between IGF-IR and TSHR was first described cose intolerance or diabetes and did not result in
by Tramontano et al. in 1986 [136]. Since then, discontinuation of teprotumumab. There were no
Tsui and colleagues have shown that TSHR and cases of diabetic ketoacidosis. There were no sig-
IGF-IR form a protein complex that is found on nificant abnormalities in electrolytes. A single
orbital fibroblasts [137]. Antibodies directed infusion reaction occurred in one patient in
against either IGF-IR or TSHR can immunopre- Phase 3 trial that resulted in discontinuation of
cipitate both proteins [137]. Similarly, in orbital the drug [139]. Another patient developed ele-
fibroblasts, IGF-IR blocking antibodies can also vated blood pressure which was treated with med-
block TSH, TSHR-stimulating antibody, and GD- ications prior to scheduled infusions.
IgGs [137]. Therefore, interruption of IGF-IR
activity seemed to be a possible therapeutic target
for TED. Clinical Course of TED
Teprotumumab, a fully human, inhibitory
monoclonal antibody against IGF-IR, was Clinical Manifestations of TED
recently evaluated in both a Phase 2 and Phase 3 Patients with TED often complain of excessive
multicenter, placebo-controlled, double-masked lacrimation, a gritty sensation, discomfort, and
clinical trials and demonstrated efficacy in photophobia. These symptoms might prompt
patients with active, moderate-to-severe TED them to seek medical attention or might only be
[138, 139]. The studies enrolled patients 18– elicited through the physician’s careful
75 years old, who exhibited signs of TED within questioning. Similarly, the physical signs of
9 months, CAS  4 points with moderate-to- TED might prove subtle, despite substantial dis-
severe disease. Patients were randomized to comfort. The most common clinical finding is
receive either placebo or teprotumumab adminis- eyelid retraction [19] and occurs commonly in
trated intravenously once every 3 weeks for a total 37–92% of patients and gives a characteristic star-
of eight infusions. Pooled data of 171 patients in ing appearance (Figures 8 and 9) [140]. It may
the intent-to-treat analysis (84 patients receiving present as an isolated finding or in association
teprotumumab and 87 receiving placebo) demon- with exophthalmos [141]. Upper eyelid retraction
strated that a greater proportion of patients receiv- may result from increased sympathomimetic tone
ing teprotumumab had at least 2 mm reduction in but may also be the consequence of eyelid fibrosis
proptosis (65/84 [73.8%] vs. 13/87 [14.9%], [142–145]. Retraction of the lower lid usually
p < 0.001) at week 24. Additionally, patients correlates with the severity of proptosis [146].
who received teprotumumab had greater reduc- Periorbital inflammation often predominates
tion of proptosis than placebo ( 2.63 mm vs. early in the disease course. The most characteristic
0.31 mm, p < 0.001), greater percentage of signs are eyelid erythema/edema and caruncular
achieving low CAS scores (0 or 1) compared to and conjunctival injection/edema [67, 147]. Con-
placebo (52/84 [61.9%] vs. 19/87 [21.8%], junctival and caruncle signs usually suggest active
p < 0.001), and greater diplopia responder rate disease [148, 149]. Conjunctival signs include
compared to placebo (69.7% vs. 30.5%, deep temporal injection (Fig. 10). Enlarged
Thyroid Eye Disease 9

Fig. 8 Upper eyelid


retraction (Dalrymple’s
sign) and soft tissue
changes in thyroid eye
disease. The retraction can
be asymmetric. Patients
characteristically exhibit a
staring gaze

Fig. 9 (a) Left upper eyelid retraction. (b) Upper eyelid lag on downward gaze (von Graefe’s sign)

vessels may be visible over the insertion of the confound these measurements. Readings as high
medial and lateral rectus muscles. Fluctuating as 23 and 25 mm, respectively, can prove normal
upper or lower eyelid swelling indicates active in black women and men. This is due to shallower
disease, while chronic swelling in the absence of bony orbits [42, 150]. The onset of exophthalmos
erythema suggests congestive ophthalmopathy. is usually gradual and insidious, although in some
Exposure keratitis occurs in patients with TED cases it can be stormy or “malignant” (Fig. 13).
for several reasons. Proptosis and eyelid dysfunc- Most patients exhibit mild to moderate proptosis
tion can cause inadequate eyelid closure, which in [19, 150–152]. Although sometimes markedly
turn causes excessive moisture loss. Loss of Bell’s asymmetric, TED usually presents as a clearly
phenomenon from inferior rectus infiltration can bilateral process [42, 43, 153–155]. The usual
also exacerbate exposure. Additionally, lacrimal presentation includes evidence of both EOM and
gland infiltration causes diminished tear produc- fatty/connective tissue depot involvement,
tion. A careful slit-lamp examination using rose although a predominance of one of these manifes-
bengal and fluorescein stains allows identification tations can occur [78, 80]. Individuals younger
of exposure. Many patients benefit from artificial than 40 years of age are considerably more likely
tear supplementation, punctal occlusion, and to exhibit proptosis related to fat expansion in the
occlusive dressing at night. Rarely, severe expo- apparent absence of muscle infiltration [156]. On
sure keratitis can result in cornea thinning, ulcer- the other hand, older patients, especially those in
ation, and even perforation (Fig. 11). their eighth decade or beyond, can develop severe,
Exophthalmos occurs in 25–60% of patients isolated muscle enlargement associated with com-
(Fig. 12) [19]. As quantified with an exo- pressive optic neuropathy, usually as a conse-
phthalmometer, any measurement of 21 mm or quence of fusiform enlargement of the EOMs
differences greater than 2 mm between eyes sug- [157].
gests orbital disease. Racial differences can
10 Y. Wang and R. S. Douglas

Fig. 10 Deep injection of


temporal conjunctival
vessels (Goldzieher’s sign)

Fig. 11 (a) Exposure keratitis manifested by a superficial epithelial defect. (b) Less commonly, chronic exposure
keratopathy resulting in visual loss from changes including corneal vascularization and scarring

Fig. 12 (a) Marked proptosis in thyroid eye disease associated with upper and lower eyelid retraction. (b) Measurement
of exophthalmos with exophthalmometer

Clinically severe TED can present as the corresponding fields of gaze. Range of motion
dysfunctional eye motility (Fig. 14). Limited eye should be documented in the six cardinal posi-
movements are usually associated with diplopia in tions. The range of fusion can be tested with the
Thyroid Eye Disease 11

Fig. 13 (a) Patient with characteristic findings of severe, more subtle findings. (b) Patient with fulminant thyroid
acute (malignant) thyroid eye disease, including soft tissue eye disease, lid swelling, and secondary ptosis. (Courtesy
inflammation, congestion, exophthalmos, and dysthyroid of G. H. Daniels, Massachusetts General Hospital,
optic neuropathy. The majority of patients present with Boston.)

Fig. 14 (a) Limitation of upward gaze of the left eye caused by restrictive myopathy of the inferior rectus muscle. (b) Left
esotropia caused by a tethered (and restricted) left medial rectus muscle

red glass test or Maddox rod and prisms. Signifi- Elevated intraocular pressure in the primary
cant changes in the exam signal evolving disease position or on upgaze can accompany TED,
[158]. Involvement of the inferior, medial, supe- often related to either restrictive myopathy or
rior, and lateral rectus muscle is encountered in orbital congestion [167–169]. This results from
decreasing frequency [19, 159–161]. Certainly, the pull of an inelastic inferior rectus muscle on
patients with involvement of all muscles may be the eye. It can be demonstrated on attempted
encountered. Motility dysfunction from involve- upgaze [170]. Although this finding is not specific
ment of the oblique muscles is generally over- to TED, it has been observed in a large fraction of
whelmed by those of the rectus muscles [162, these patients, regardless of whether they manifest
163]. It should also be noted that most patients exophthalmos or not.
with TED, including those with no evidence of The most feared complication of TED is
ocular motility abnormalities, exhibit some dysthyroid optic neuropathy, typically caused by
degree of EOM involvement demonstrable by compression. This occurs in ~5% of individuals
radiographic techniques [76, 164–166]. with TED, and affected individuals usually do not
exhibit marked proptosis or optic nerve changes
12 Y. Wang and R. S. Douglas

Fig. 15 Optic nerve


swelling can be seen in
thyroid eye disease

under ophthalmoscopic examination [20, 171, generalized constriction represent the most com-
172]. European Group on Graves’ Orbitopathy mon visual field defects in TED [183].
(EUGOGO) found that 25% of patients with
dysthyroid optic neuropathy had CAS or three or Natural History of TED
less, suggesting that these patients may not have TED typically pursues a self-limited course,
significant visible inflammation [173]. Risk fac- becoming quiescent within 3–5 years of its onset
tors for optic neuropathy include older age, [20, 184, 185]. In a landmark study, Rundle and
smoking, male gender, and significant strabismus Wilson followed the natural history of 226
with mild proptosis [174]. Because of its often patients with TED and described orbital changes
insidious onset and subtlety, visual loss may pro- in the three phases of disease [184, 185]. The
gress undetected or its cause may be mis- active inflammatory phase is associated with
diagnosed [175, 176]. Typically, acuity and color orbital and periorbital signs including proptosis
perception are slightly decreased with a question- and eyelid retraction. Subsequently, the static
able afferent pupillary defect and vague visual- phase heralds little clinical improvement despite
field defects, while the optic disk appears entirely reduced inflammation. Finally, gradual improve-
normal to clinical examination [177]. Less com- ment in lid retraction and ocular motility may
monly, optic nerve edema and peripapillary hem- occur in the chronic quiescent phase. Upper eyelid
orrhages occur (Fig. 15). These represent ominous retraction improves in a majority of patients. Ocu-
findings necessitating aggressive treatment. lar motility follows a more variable course, and
Direct orbital manometry has elegantly demon- unless vision is threatened, patients should be
strated an increase in orbital pressure in TED followed regularly to document stability prior to
[178]. Increased pressure results from reduced therapeutic intervention [158, 186]. Proptosis
orbital compliance and can compromise the optic usually improves only slightly, even after long
nerve [179–181]. Patients developing optic neu- periods of time, and the increased orbital content
ropathy may develop late nerve atrophy and irre- often proves irreversible [21]. Occasional cases of
versible visual loss. Suspected optic neuropathy spontaneously improved proptosis have been
necessitates careful assessment of visual acuity, reported [187].
color vision, visual fields, afferent pupillary Clinical manifestations during the chronic
responses, and funduscopy [182]. Arcuate or alti- phase are typically stable. However, a small per-
tudinal defects, paracentral scotomas, and centage of patients do experience reactivation, in
which patients reenter the active inflammatory
Thyroid Eye Disease 13

phase with fluctuating and often worsening clini- Imaging Techniques


cal signs and symptoms [188–191]. In a retrospec-
tive study, 5% of patients developed late Computed tomography (CT) without contrast
reactivation. At the time of reactivation, there remains the most widely utilized imaging tool in
was an average of 12 years from initial TED the diagnosis of TED [195]. Enlargement of the
presentation. Further, all the patients were euthy- EOM bellies, with tendon sparing, is the classic
roid at that time [188]. The most common wors- finding but has proven far from universal [76,
ening symptom was proptosis. Recurrent active 196–198]. Both direct axial and coronal cuts in
phase lasted for mean of 14 months. Potential risk addition to small (2–3 mm) cuts through the
factors for reactivation included history of orbital apex are utilized for evaluation. CT is
smoking during initial TED presentation, peri- particularly helpful in assessing the relationship
ocular surgery, fluctuating thyroid levels, preg- between the EOM and optic nerve at the orbital
nancy, and significant life stressors [191]. apex and can aid in surgical planning [176, 199].
B-mode ultrasonography represents a useful
Clinical Classification System imaging technique for the diagnosis of TED
Useful classification of TED has proven difficult, [166, 200]. Enlarged EOM can be identified but
a consequence of the complex and variable course the approach yields images confined to the ante-
of the disease. Lack of consensus regarding its rior one-third of the orbit with a standard 10 MHz
pathogenesis and descriptors has severely ham- probe. Increased EOM reflectivity, a potential
pered progress in evaluating therapeutic interven- marker for TED activity, can be measured by A-
tion. NOSPECS, developed by Werner et al. and mode ultrasound. This technique, however, is
adopted by the American Thyroid Association extremely operator-dependent [201].
categorizes the disease based upon clinical pre- A number of small studies have explored the
sentation [192, 193]. This classification has been potential utility of magnetic resonance imaging
criticized because it relies on subjective evalua- (MRI) in the diagnosis of active TED [202–204].
tion, fails to take into account the severity of The ability of MRI to identify the typical fusiform
manifestations, and is relatively insensitive to rectus muscle enlargement and orbital fat expan-
subtle changes. Thus, it has been abandoned. sion has been established (Fig. 16) [164, 165]. Its
Other systems have been proposed, but none is utility in the assessment of disease activity has
well-standardized [148, 149]. The most widely been explored by assessing water content in mus-
utilized of these is the CAS system, first described cles, which may correlate with active inflamma-
by Mourits and colleagues [148, 149]. It attempts tion [205–208]. Strategies for enhancing its
to identify patients with active disease who are sensitivity and specificity as an effective tool
likely to respond to medical therapy. Each symp- include fat suppression, short-term inversion
tom or sign (retrobulbar pain, eyelid erythema, recovery, and T2 relaxation time. None of these,
conjunctival injection, chemosis, swelling of the however, has proven consistently valuable [209,
caruncle, eyelid edema or fullness) is assigned 210].
equal weight, and the score is a simple summa-
tion. Unfortunately, the CAS score is limited to
the subjectivity of both patient and practitioner. In Conclusion
addition, the manner in which the score is gener-
ated fails to account for active improvement or TED remains a challenging disease to treat, in part
worsening of the disease. In practice, the charac- due to its complex pathophysiology. Historically,
terization of TED remains imperfect and based on medical therapies have not been able to modify
vague and largely subjective clinical parameters the disease course and primarily aimed at symp-
[67, 158, 194]. tomatic relief only. Recently on January 21, 2020,
the US Food and Drug Administration approved
teprotumumab to treat TED. The clinical usage of
14 Y. Wang and R. S. Douglas

Fig. 16 Magnetic resonance imaging of a patient with inferior rectus muscle (white arrow). (b) Coronal image
thyroid eye disease. (a) Parasagittal image demonstrates demonstrates enlarged right inferior and lateral rectus mus-
enlargement of the nontendinous portion of the right cles (black arrows)

teprotumumab likely extends beyond the drug’s 8. Bilimoria KY, Pescovitz OH, DiMeglio LA. Autoim-
use in the clinical trials and can be used as first- mune thyroid dysfunction in children with type 1
diabetes mellitus: screening guidelines based on a
line therapy in patients with clinically significant retrospective analysis. J Pediatr Endocrinol Metab.
TED. 2003;16(8):1111–1117.
9. Burek CL, Rose NR, Guire KE, Hoffman WH. Thy-
roid autoantibodies in black and in white children and
adolescents with type 1 diabetes mellitus and their first
References degree relatives. Autoimmunity. 1990;7(2–3):157–
167.
1. Bartley GB, Fatourechi V, Kadrmas EF, et al. The 10. Chikuba N, Akazawa S, Yoshihiko Y, et al. Type 1
incidence of graves’ ophthalmopathy in Olmsted (insulin-dependent) diabetes mellitus with coexisting
County, Minnesota. Am J Ophthalmol. 1995;120 autoimmune thyroid disease in Japan. Intern Med.
(4):511–517. 2008;31(9):1076–1080.
2. Gorman CA. Temporal relationship between onset of 11. Kalmann R, Mourits MP. Diabetes mellitus: a risk
“graves” ophthalmopathy and diagnosis of thyrotox- factor in patients with graves’ orbitopathy. Br J
icosis. Mayo Clin Proc. 1983;58(8):515–519. Ophthalmol. 1999;83(4):463–465.
3. Wiersinga WM, Smit T, Van Der Gaag R, Mourits M, 12. Kirkegaard C, Bech K, Bliddal H, Danneskiold-Saoe
Koornneef L. Clinical presentation of graves’ B, Feldt-Rasmussen U. Thyroid stimulating anti-
ophthalmopathy. Ophthalmic Res. 1989;21(2):73–82. bodies in rheumatoid arthritis: an in vitro phenome-
4. Wiersinga WM, Smit T, van der Gaag R, Koornneef non. J Endocrinol Investig. 1987;10(5):495–498.
L. Temporal relationship between onset of graves’ 13. Ruggeri RM, Galletti M, Mandolfino MG, et al. Thy-
ophthalmopathy and onset of thyroidal graves’ dis- roid hormone autoantibodies in primary Sjögren syn-
ease. J Endocrinol Investig. 1988;11(8):615–619. drome and rheumatoid arthritis are more prevalent
5. Perros P, Crombie AL, Matthews JNS, Kendall-Tay- than in autoimmune thyroid disease, becoming pro-
lor P. Age and gender influence the severity of thy- gressively more frequent in these diseases. J Endo-
roid-associated ophthalmopathy: a study of 101 crinol Investig. 2002;25(5):447–454.
patients attending a combined thyroid-eye clinic. 14. Sanders PA, Grennan DM, Dyer PA, de Lange GG,
Clin Endocrinol. 1993;38(4):367–372. Harris R. Immunogenetic studies in families with
6. Perros P, Crombie AL, Kendall-Taylor P. Natural rheumatoid arthritis and autoimmune thyroid disease.
history of thyroid associated ophthalmopathy. Clin J Med Genet. 2008;22(6):451–456.
Endocrinol. 1995;42(1):45–50. 15. Perros P, Singh RK, Ludlam CA, Frier BM. Preva-
7. Akamizu T, Nakamura Y, Tamaoki A, Inaba Y, Amino lence of pernicious anaemia in patients with type 1
N, Seino Y. Prevalence and Clinico-epidemiology of diabetes mellitus and autoimmune thyroid disease.
familial graves’ disease in Japan based on Nationwide Diabet Med. 2000;17(10):749–751.
epidemiologic survey in 2001. Endocr J. 2003;50 16. Hegedus L, Heidenheim M, Gervil M, Hjalgrim H,
(4):429–43. Hoier-Madsen M. High frequency of thyroid
Thyroid Eye Disease 15

dysfunction in patients with vitiligo. Acta Derm 31. Cho BY. Clinical applications of TSH receptor anti-
Venereol. 1994;74(2):120–123. bodies in thyroid diseases. J Korean Med Sci. 2002;17
17. Kemp EH, Waterman EA, Gawkrodger DJ, Watson (3):293–301.
PF, Weetman AP. Autoantibodies to tyrosinase- 32. Smith BR, McLachlan SM, Furmaniak J. Autoanti-
related protein-1 detected in the sera of vitiligo bodies to the thyrotropin receptor. Endocr Rev. 1988.
patients using a quantitative radiobinding assay. Br J 33. Daniels GH. Hyperthyroidism: multiple possibilities
Dermatol. 1998;139(5):798–805. in the female patient. Int J Fertil Womens Med.
18. L.E. Braverman DC. Werner and Ingbar’s the thyroid: 1999;44(1):6–11.
a fundamental and clinical text. Ann Intern Med. 34. Bartalena L, Pinchera A, Marcocci C. Management of
1996;117(6):541–2. graves’ ophthalmopathy: reality and perspectives.
19. Bartley GB, Fatourechi V, Kadrmas EF, et al. Clinical Endocr Rev. 2000;21(2):168–199.
features of graves’ Ophthalmopathy in an incidence 35. Chiovato L, Fiore E, Vitti P, et al. Outcome of thyroid
cohort. Am J Ophthalmol. 1996;121(3):284–90. function in graves’ patients treated with radioiodine:
20. Bartley GB. The epidemiologic characteristics and role of thyroid-stimulating and thyrotropin-blocking
clinical course of ophthalmopathy associated with antibodies and of radioiodine-induced thyroid dam-
autoimmune thyroid disease in Olmsted County, Min- age. J Clin Endocrinol Metab. 1998;83(1):40–46.
nesota. Trans Am Ophthalmol Soc. 1994;92:477–588. 36. Berg G, Michanek A, Holmberg E, Nyström E. Clin-
21. Bartley GB, Fatourechi V, Kadrmas EF, et al. Long- ical outcome of radioiodine treatment of hyperthy-
term follow-up of graves ophthalmopathy in an inci- roidism: a follow-up study. J Intern Med. 1996;239
dence cohort. Ophthalmology. 1996;103(6):958–962. (2):165–171.
22. Khoo DH, Eng PH, Ho SC, et al. Graves’ 37. Kendler DL, Lippa J, Rootman J. The initial clinical
Ophthalmopathy in the absence of elevated free thy- characteristics of graves’ Orbitopathy vary with age
roxine and triiodothyronine levels: prevalence, natu- and sex. Arch Ophthalmol. 1993;111(2):197–201.
ral history, and thyrotropin receptor antibody levels. 38. Dallow R, Pratt S. Hereditary aspects of graves’ dis-
Thyroid. 2000;10(12):1093–1100. ease in infancy and childhood. In: Albert D, Jakobiec
23. Benker G, Reinwein D, Kahaly G, et al. Is there a F, editors. Principles and practice of ophthalmology.
methimazole dose effect on remission rate in graves’ Philadelphia: WB Saunders; 1994. p. 1881–90.
disease? Results from a long-term prospective study. 39. Hollingworth DR, Mabry CC, Eckerd JM. Hereditary
Clin Endocrinol. 1998;49(4):451–457. aspects of graves’ disease in infancy and childhood. J
24. Bromberg N, Romaldini JH, Werner RS, Sgarbi JA, Pediatr. 1972;81(3):446–459.
Werner MC. The evolution of graves’ 40. Bartley GB, Fatourechi V, Kadrmas EF, et al. The
ophthalmopathy during treatment with antithyroid chronology of graves’ ophthalmopathy in an incidence
drug alone and combined with triiodothyronine. J cohort. Am J Ophthalmol. 1996;121(4):426–434.
Endocrinol Investig. 1992;15(3):191–195. 41. Rootman J. Inflammatory diseases of the orbit. J Fr
25. García-Mayor RVG, Páramo C, Cano RL, Méndez Ophtalmol. 2001;24:155–61.
LFP, Galofré JC, Andrade A. Antithyroid drug and 42. Grove AS. Evaluation of exophthalmos. N Engl J
Graves’ hyperthyroidism. Significance of treatment Med. 1975;292(19):1005–1013.
duration and TRAb determination on lasting remis- 43. Lawton NF. Exclusion of dysthyroid eye disease as a
sion. J Endocrinol Investig Off J Ital Scociety Endo- cause of unilateral proptosis. Trans Ophthalmol Soc
crinol. 1992;15(11):815–820. U K. 1979;99(2):226–228.
26. Kashiwai T, Tada H, Asahi K, et al. Significance of 44. Villadolid MC, Yokoyama N, Izumi M, et al.
thyroid stimulating antibody and long term follow up Untreated graves’ disease patients without clinical
in patients with Euthyroid graves’ disease. Endocr J. ophthalmopathy demonstrate a high frequency of
1995;42(3):405–412. extraocular muscle (EOM) enlargement by magnetic
27. Marcocci C, Bartalena L, Bogazzi F, Bruno-Bossio G, resonance. J Clin Endocrinol Metab. 1995;80
Pinchera A. Relationship between graves’ (9):2830–2833.
Ophthalmopathy and type of treatment of graves’ 45. Mack WP, Stasior GO, Cao HJ, Stasior OG, Smith TJ.
hyperthyroidism. Thyroid. 1992;2(2):171–178. The effect of cigarette smoke constituents on the
28. Orgiazzi J. Anti-TSH receptor antibodies in clinical expression of HLA-DR in orbital fibroblasts derived
practice. Endocrinol Metab Clin N Am. 2000;29 from patients with graves ophthalmopathy. Ophthal
(2):339–355, vii. Plast Reconstr Surg. 1999;15(4):260–271.
29. Prabhakar BS, Bahn RS, Smith TJ. Current perspec- 46. Prummel MF, Wiersinga WM. Smoking and risk of
tive on the pathogenesis of graves’ disease and graves’ disease. JAMA. 1993;269(4):479–82.
Ophthalmopathy. Endocr Rev. 2003;24(6):802–35. 47. Bartalena L, Martino E, Marcocci C, et al. More on
30. Weetman AP, Cohen S, Gatter KC, Fells P, Shine B. smoking habits and graves’ ophthalmopathy. J Endo-
Immunohistochemical analysis of the retrobulbar tis- crinol Investig. 1989;12(10):733–7.
sues in graves’ ophthalmopathy. Clin Exp Immunol. 48. Bartalena L, Marcocci C, Tanda ML, et al. Cigarette
1989;75(2):222–7. smoking and treatment outcomes in graves
ophthalmopathy. Ann Intern Med. 1998;129(8):632–5.
16 Y. Wang and R. S. Douglas

49. Bartalena L, Bogazzi F, Tanda ML, Manetti L, systematic review. Clin Endocrinol. 2008;69
Dell’Unto E, Martino E. Cigarette smoking and the (6):943–50.
thyroid. Eur J Endocrinol. 1995;133(5):507–512. 64. Ponto KA, Zang S, Kahaly GJ. The tale of radioiodine
50. Hägg E, Asplund K. Is endocrine ophthalmopathy and graves’ orbitopathy. Thyroid. 2010;20(7):785–93.
related to smoking? Br Med J (Clin Res Ed). 65. Lai A, Sassi L, Compri E, et al. Lower dose predni-
1987;295(6599):634–63. sone prevents radioiodine-associated exacerbation of
51. Pfeilschifter J, Ziegler R. Smoking and endocrine initially mild or absent graves’ orbitopathy: a retro-
ophthalmopathy: impact of smoking severity and cur- spective cohort study. J Clin Endocrinol Metab.
rent vs lifetime cigarette consumption. Clin Endo- 2010;95(3):1333–7.
crinol. 1996;45(4):477–81. 66. Shiber S, Stiebel-Kalish H, Shimon I, Grossman A,
52. Eckstein A, Quadbeck B, Mueller G, et al. Impact of Robenshtok E. Glucocorticoid regimens for preven-
smoking on the response to treatment of thyroid asso- tion of graves’ ophthalmopathy progression follow-
ciated ophthalmopathy. Br J Ophthalmol. 2003;87 ing radioiodine treatment: systematic review and
(6):773–776. meta-analysis. Thyroid. 2014;24(10):1515–23.
53. Harrison BJ, Silman AJ. Does smoking influence 67. Prummel MF, Bakker A, Wiersinga WM, et al. Multi-
disease outcome in patients with rheumatoid arthritis? center study on the characteristics and treatment strat-
J Rheumatol. 2000;27(3):569–570. egies of patients with graves’ orbitopathy: the first
54. Silman AJ, Newman J, MacGregor AJ. Cigarette European group on graves’ Orbitopathy experience.
smoking increases the risk of rheumatoid arthritis: Eur J Endocrinol. 2003;148(5):491–5.
results from a nationwide study of disease-discordant 68. Wegelius O, Asboe-Hansen G, Lamberg BA.
twins. Arthritis Rheum. 1996;39(5):732–735. Retrobulbar connective tissue changes in malignant
55. Calkins BM. A meta-analysis of the role of smoking exophthalmos. Acta Endocrinol (Copenh). 1957;25
in inflammatory bowel disease. Dig Dis Sci. 1989;34 (4):452–456.
(12):1841–1854. 69. Heufelder AE, Bahn RS. Elevated expression in situ
56. Wakelkamp IMMJ, Gerding MN, Van Der Meer of selectin and immunoglobulin superfamily type
JWC, Prummel MF, Wiersinga WM. Both Th1- and adhesion molecules in retroocular connective tissues
Th2-derived cytokines in serum are elevated in from patients with Graves’ ophthalmopathy. Clin Exp
graves’ ophthalmopathy. Clin Exp Immunol. Immunol. 1993;91(3):381–389.
2000;121(3):453–457. 70. Heufelder AE, Bahn RS. Detection and localization of
57. Salvi M, Girasole G, Pedrazzoni M, et al. Increased cytokine immunoreactivity in retro-ocular connective
serum concentrations of interleukin-6 (IL-6) and sol- tissue in Graves’ ophthalmopathy. Eur J Clin Invest.
uble IL-6 receptor in patients with Graves’ disease. J 1993;23(1):10–17.
Clin Endocrinol Metab. 1996;81(8):2976–2979. 71. de Carli M, D’Elios MM, Mariotti S, et al. Cytolytic T
58. Bartalena L, Manetti L, Tanda ML, et al. Soluble cells with Th1-like cytokine profile predominate in
interleukin-1 receptor antagonist concentration in retroorbital lymphocytic infiltrates of Graves’
patients with graves’ ophthalmopathy is neither ophthalmopathy. J Clin Endocrinol Metab. 1993;77
related to cigarette smoking nor predictive of subse- (5):1120–1124.
quent response to glucocorticoids. Clin Endocrinol. 72. Hiromatsu Y, Kaku H, Miyake I, Murayama S,
2000;52(5):647–651. Soejima E. Role of cytokines in the pathogenesis of
59. Wakelkamp IM, Prummel MF, Wiersinga WM. thyroid-associated ophthalmopathy. Thyroid.
Serum IL-18 levels are not increased in patients with 2002;12(3):217–221.
untreated Graves’ ophthalmopathy. Horm Metab Res. 73. Hiromatsu Y, Yang D, Bednarczuk T, Miyake I, Non-
2004;36(1):44–47. aka K, Inoue Y. Cytokine profiles in eye muscle tissue
60. Atkinson S, McGregor AM, Kendall-Taylor P, Peter- and orbital fat tissue from patients with thyroid-asso-
son MM, Smith BR. Effect of radioiodine on stimu- ciated ophthalmopathy. J Clin Endocrinol Metab.
latory activity of graves’ immunoglobulins. Clin 2000;85(3):1194–1199.
Endocrinol. 1982;16(6):537–43. 74. McLachlan SM. Graves’ disease: the T(H)1/T(H)2 par-
61. Kriss JP, Pleshakov V, Rosenblum AL, Holderness M, adigm versus the “hygiene” hypothesis and defective
Sharp G, Utiger R. Studies on the pathogenesis of the immune regulation. Thyroid. 2003;13(2):127–128.
ophthalmopathy of Graves’ disease. J Clin Endo- 75. Aniszewski JP, Valyasevi RW, Bahn RS. Relationship
crinol Metab. 1967;27(4):582–593. between disease duration and predominant orbital T
62. Bartalena L, Marcocci C, Bogazzi F, et al. Relation cell subset in Graves’ ophthalmopathy. J Clin Endo-
between therapy for hyperthyroidism and the course crinol Metab. 2000;85(2):776–780.
of graves’ ophthalmopathy. N Engl J Med. 1998;338 76. Orbes G, Gorman CA, Brennan MD, Gehring DG,
(2):73–8. Ilstrup DM, Earnest Ft. Ophthalmopathy of Graves’
63. Acharya SH, Avenell A, Philip S, Burr J, Bevan JS, disease: computerized volume measurements of the
Abraham P. Radioiodine therapy (RAI) for graves’ orbital fat and muscle. Am J Neuroradiol. 1986;7
disease (GD) and the effect on ophthalmopathy: a (4):651–656.
Thyroid Eye Disease 17

77. Hudson HL, Levin L, Feldon SE. Graves exophthal- chemoattractant expression in fibroblasts from
mos unrelated to extraocular muscle enlargement. patients with graves’ disease is mediated through the
Superior rectus muscle inflammation may induce insulin-like growth factor I receptor pathway. J
venous obstruction. Ophthalmology. 1991;98 Immunol. 2003;170(12):6348–54.
(10):1495–1499. 92. Sisson JC, Spaugh BI, Vanderburg JA. Functional
78. Peyster RG, Ginsberg F, Silber JH, Adler LP. Exoph- aspects of fibroblasts derived from the retrobulbar
thalmos caused by excessive fat: CT volumetric anal- tissue of man. Exp Eye Res. 1970;10(2):201–206.
ysis and differential diagnosis. AJR Am J Roentgenol. 93. Koumas L, Smith TJ, Feldon S, Blumberg N, Phipps
1986;146(3):459–464. RP. Thy-1 expression in human fibroblast subsets
79. Kazim M, Goldberg RA, Smith TJ. Insights into the defines myofibroblastic or lipofibroblastic pheno-
pathogenesis of thyroid-associated orbitopathy: types. Am J Pathol. 2003;163(4):1291–300.
evolving rationale for therapy. Arch Ophthalmol. 94. Smith TJ, Sempowski GD, Berenson CS, Cao HJ,
2002;120(3):380–386. Wang HS, Phipps RP. Human thyroid fibroblasts
80. Nunery WR, Martin RT, Heinz GW, Gavin TJ. The exhibit a distinctive phenotype in culture: character-
association of cigarette smoking with clinical sub- istic ganglioside profile and functional CD40 expres-
types of ophthalmic Graves’ disease. Ophthalmic sion. Endocrinology. 1997;138(12):5576–5588.
Plast Reconstr Surg. 1993;9(2):77–82. 95. Smith TJ, Sciaky D, Phipps RP, Jennings TA. CD40
81. Tallstedt L, Norberg R. Immunohistochemical expression in human thyroid tissue: evidence for
staining of normal and Graves’ extraocular muscle. involvement of multiple cell types in autoimmune
Invest Ophthalmol Vis Sci. 1988;29(2):175–184. and neoplastic diseases. Thyroid. 1999;9(8):749–755.
82. Kahaly G, Forster G, Hansen C. Glycosaminoglycans 96. Smith TJ, Wang HS, Evans CH. Leukoregulin is a
in thyroid eye disease. Thyroid. 1998;8(5):429–432. potent inducer of hyaluronan synthesis in cultured
83. Pappa A, Jackson P, Stone J, et al. An ultrastructural human orbital fibroblasts. Am J Physiol. 1995;268(2
and systemic analysis of glycosaminoglycans in thy- Pt 1):C382–388.
roid-associated ophthalmopathy. Eye. 1998;12(Pt 97. Koumas L, Smith TJ, Phipps RP. Fibroblast subsets in
2):237–44. the human orbit: Thy-1+ and Thy-1- subpopulations
84. Hufnagel TJ, Hickey WF, Cobbs WH, Jakobiec FA, exhibit distinct phenotypes. Eur J Immunol. 2002;32
Iwamoto T, Eagle RC. Immunohistochemical and (2):477–85.
ultrastructural studies on the exenterated orbital tis- 98. Smith TJ, Sempowski GD, Wang HS, Del Vecchio PJ,
sues of a patient with Graves’ disease. Ophthalmol- Lippe SD, Phipps RP. Evidence for cellular heteroge-
ogy. 1984;91(11):1411–1419. neity in primary cultures of human orbital fibroblasts.
85. Kaback LA, Smith TJ. Expression of hyaluronan syn- J Clin Endocrinol Metab. 1995;80(9):2620–2625.
thase messenger ribonucleic acids and their induction 99. Douglas RS, Afifiyan NF, Hwang CJ, et al. Increased
by interleukin-1beta in human orbital fibroblasts: generation of fibrocytes in thyroid-associated
potential insight into the molecular pathogenesis of ophthalmopathy. J Clin Endocrinol Metab. 2010;95
thyroid-associated ophthalmopathy. J Clin Endo- (1):430–8.
crinol Metab. 1999;84(11):4079–4084. 100. Bucala R, Spiegel LA, Chesney J, Hogan M, Cerami
86. Smith TJ. Orbital fibroblasts exhibit a novel pattern of A. Circulating fibrocytes define a new leukocyte sub-
responses to proinflammatory cytokines: potential population that mediates tissue repair. Mol Med.
basis for the pathogenesis of thyroid-associated 1994;1(1):71–81.
ophthalmopathy. Thyroid. 2002;12(3):197–203. 101. Smith TJ. Potential role for bone marrow-derived
87. Smith TJ. Unique properties of orbital connective fibrocytes in the orbital fibroblast heterogeneity asso-
tissue underlie its involvement in Graves’ disease. ciated with thyroid-associated ophthalmopathy. Clin
Minerva Endocrinol. 2003;28(3):213–222. Exp Immunol. 2010;162(1):24–31.
88. Smith TJ, Bahn RS, Gorman CA. Connective tissue, 102. Gillespie EF, Papageorgiou KI, Fernando R, et al.
glycosaminoglycans, and diseases of the thyroid. Increased expression of TSH receptor by fibrocytes
Endocr Rev. 1989;10(3):366–391. in thyroid-associated ophthalmopathy leads to che-
89. Smith TJ, Bahn RS, Gorman CA, Cheavens M. Stim- mokine production. J Clin Endocrinol Metab.
ulation of glycosaminoglycan accumulation by inter- 2012;97(5):E740–746.
feron gamma in cultured human retroocular 103. Akamizu T, Mori T, Nakao K. Pathogenesis of Graves’
fibroblasts. J Clin Endocrinol Metab. 1991;72 disease: molecular analysis of anti-thyrotropin receptor
(5):1169–1171. antibodies. Endocr J. 1997;44(5):633–646.
90. Smith TJ, Wang HS, Hogg MG, Henrikson RC, Keese 104. Davies TF, Roti E, Braverman LE, DeGroot LJ. Thy-
CR, Giaever I. Prostaglandin E2 elicits a morpholog- roid controversy–stimulating antibodies. J Clin Endo-
ical change in cultured orbital fibroblasts from crinol Metab. 1998;83(11):3777–3785.
patients with Graves ophthalmopathy. Proc Natl 105. Goh SY, Ho SC, Seah LL, Fong KS, Khoo DH.
Acad Sci U S A. 1994;91(11):5094–5098. Thyroid autoantibody profiles in ophthalmic domi-
91. Pritchard J, Han R, Horst N, Cruikshank WW, Smith nant and thyroid dominant Graves’ disease differ
TJ. Immunoglobulin activation of T cell
18 Y. Wang and R. S. Douglas

and suggest ophthalmopathy is a multiantigenic dis- orbitopathy. Ophthalmic Plast Reconstr Surg.
ease. Clin Endocrinol (Oxf). 2004;60(5):600–607. 2014;30(2):162–167.
106. Kim WB, Chung HK, Park YJ, Park DJ, Lee HK, Cho 119. Atienza-Mateo B, Calvo-Río V, Martín-Varillas JL, et
BY. Clinical significance of classification of Graves’ al. SAT0601 Anti-il6-receptor tocilizumab in graves’
disease according to the characteristics of TSH receptor orbitopathy. multicenter study of 29 patients. 2019.
antibodies. Korean J Intern Med. 2001;16(3):187–200. 120. Perez-Moreiras JV, Gomez-Reino JJ, Maneiro JR, et
107. Kosugi S, Inoue D, Sugawa H, Enomoto T, Mori T, al. Efficacy of Tocilizumab in Patients With Moder-
Imura H. Similarity and dissimilarity between clinical ate-to-Severe Corticosteroid-Resistant Graves
and laboratory findings, especially anti-thyrotropin Orbitopathy: A Randomized Clinical Trial. Am J
receptor antibody in ophthalmic Graves’ disease with- Ophthalmol. 2018;195:181–190.
out persistent hyperthyroidism and hyperthyroid 121. Rotella CM, Zonefrati R, Toccafondi R, Valente WA,
Graves’ disease. Endocrinol Jpn. 1990;37(3):343–354. Kohn LD. Ability of monoclonal antibodies to the
108. Macchia E, Concetti R, Borgoni F, Cetani F, Fenzi thyrotropin receptor to increase collagen synthesis in
GF, Pinchera A. Assays of TSH-receptor antibodies in human fibroblasts: an assay which appears to measure
576 patients with various thyroid disorders: their inci- exophthalmogenic immunoglobulins in Graves’ sera.
dence, significance and clinical usefulness. Autoim- J Clin Endocrinol Metab. 1986;62(2):357–367.
munity. 1989;3(2):103–112. 122. Pritchard J, Horst N, Cruikshank W, Smith TJ. Igs
109. Bell A, Gagnon A, Grunder L, Parikh SJ, Smith TJ, from patients with Graves’ disease induce the expres-
Sorisky A. Functional TSH receptor in human sion of T cell chemoattractants in their fibroblasts. J
abdominal preadipocytes and orbital fibroblasts. Am Immunol. 2002;168(2):942–950.
J Physiol Cell Physiol. 2000;279(2):C335–40. 123. Weis-Klemm M, Alexander D, Pap T, et al. Synovial
110. Heufelder AE. Involvement of the orbital fibroblast fibroblasts from rheumatoid arthritis patients differ in
and TSH receptor in the pathogenesis of Graves’ their regulation of IL-16 gene activity in comparison
ophthalmopathy. Thyroid. 1995;5(4):331–340. to osteoarthritis fibroblasts. Cell Physiol Biochem.
111. Heufelder A, Bahn R. Evidence for the presence of a 2004;14(4–6):293–300.
functional TSH-receptor in retroocular fibroblasts 124. Lard LR, Roep BO, Toes RE, Huizinga TW.
from patients with graves’ ophthalmopathy. Exp Enhanced concentrations of interleukin 16 are associ-
Clin Endocrinol. 1992;100:62–7. ated with joint destruction in patients with rheumatoid
112. Heufelder AE, Dutton CM, Sarkar G, Donovan KA, arthritis. J Rheumatol. 2004;31(1):35–39.
Bahn RS. Detection of TSH receptor RNA in cultured 125. Blaschke S, Schulz H, Schwarz G, Blaschke V,
fibroblasts from patients with Graves’ Muller GA, Reuss-Borst M. Interleukin 16 expression
ophthalmopathy and pretibial dermopathy. Thyroid. in relation to disease activity in rheumatoid arthritis. J
1993;3(4):297–300. Rheumatol. 2001;28(1):12–21.
113. Bahn RS, Dutton CM, Natt N, Joba W, Spitzweg C, 126. Kageyama Y, Ozeki T, Suzuki M, et al. Interleukin-16
Heufelder AE. Thyrotropin receptor expression in in synovial fluids from cases of various types of
Graves’ orbital adipose/connective tissues: potential arthritis. Joint Bone Spine. 2000;67(3):188–193.
autoantigen in Graves’ ophthalmopathy. J Clin Endo- 127. Middel P, Reich K, Polzien F, et al. Interleukin 16
crinol Metab. 1998;83(3):998–1002. expression and phenotype of interleukin 16 producing
114. Kumar S, Nadeem S, Stan MN, Coenen M, Bahn cells in Crohn’s disease. Gut. 2001;49(6):795–803.
RS. A stimulatory TSH receptor antibody enhances 128. Seegert D, Rosenstiel P, Pfahler H, Pfefferkorn P,
adipogenesis via phosphoinositide 3-kinase activa- Nikolaus S, Schreiber S. Increased expression of IL-
tion in orbital preadipocytes from patients with 16 in inflammatory bowel disease. Gut. 2001;48
Graves’ ophthalmopathy. J Mol Endocrinol. (3):326–332.
2011;46(3):155–163. 129. Lard LR, Roep BO, Verburgh CA, Zwinderman AH,
115. Smith TJ. Insights into the role of fibroblasts in human Huizinga TW. Elevated IL-16 levels in patients with
autoimmune diseases. Clin Exp Immunol. 2005;141 systemic lupus erythematosus are associated with dis-
(3):388–97. ease severity but not with genetic susceptibility to
116. Prummel MF, Wiersinga WM, Van der Gaag R, lupus. Lupus. 2002;11(3):181–185.
Mourits MP, Koornneef L. Soluble IL-2 receptor 130. Lee S, Kaneko H, Sekigawa I, Tokano Y, Takasaki Y,
levels in patients with Graves’ ophthalmopathy. Clin Hashimoto H. Circulating interleukin-16 in systemic
Exp Immunol. 1992;88(3):405–409. lupus erythematosus. Br J Rheumatol. 1998;37
117. Smolen JS, Beaulieu A, Rubbert-Roth A, et al. Effect (12):1334–1337.
of interleukin-6 receptor inhibition with tocilizumab 131. Gianoukakis AG, Douglas RS, King CS, Cruikshank
in patients with rheumatoid arthritis (OPTION study): WW, Smith TJ. Immunoglobulin G from patients with
a double-blind, placebo-controlled, randomised trial. Graves’ disease induces interleukin-16 and RANTES
Lancet. 2008;371(9617):987–997. expression in cultured human thyrocytes: a putative
118. Perez-Moreiras JV, Alvarez-Lopez A, Gomez EC. mechanism for T-cell infiltration of the thyroid in
Treatment of active corticosteroid-resistant graves’ autoimmune disease. Endocrinology. 2006;147
(4):1941–1949.
Thyroid Eye Disease 19

132. Kemp EH, Metcalfe RA, Smith KA, Woodroofe MN, 146. van den Bosch WA, Tjon-Fo-Sang MJ, Lemij HG.
Watson PF, Weetman AP. Detection and localization Eyeball position in Graves orbitopathy and its signif-
of chemokine gene expression in autoimmune thyroid icance for eyelid surgery. Ophthalmic Plast Reconstr
disease. Clin Endocrinol (Oxf). 2003;59(2):207–213. Surg. 1998;14(5):328–335.
133. Gianoukakis AG, Martino LJ, Horst N, Cruikshank 147. Prummel MF. Pathogenetic and clinical aspects of
WW, Smith TJ. Cytokine-induced lymphocyte endocrine ophthalmopathy. Exp Clin Endocrinol Dia-
chemoattraction from cultured human thyrocytes: evi- betes. 1999;107 Suppl 3:S75–78.
dence for interleukin-16 and regulated upon activa- 148. Mourits MP, Koornneef L, Wiersinga WM, Prummel
tion, normal T cell expressed, and secreted MF, Berghout A, van der Gaag R. Clinical criteria for
expression. Endocrinology. 2003;144(7):2856–2864. the assessment of disease activity in Graves’
134. Lawrence MC, McKern NM, Ward CW. Insulin ophthalmopathy: a novel approach. Br J Ophthalmol.
receptor structure and its implications for the IGF-1 1989;73(8):639–644.
receptor. Curr Opin Struct Biol. 2007;17(6):699–705. 149. Mourits MP, Prummel MF, Wiersinga WM, Koornneef
135. Smith TJ, Hoa N. Immunoglobulins from patients L. Clinical activity score as a guide in the management
with Graves’ disease induce hyaluronan synthesis in of patients with Graves’ ophthalmopathy. Clin Endo-
their orbital fibroblasts through the self-antigen, insu- crinol (Oxf). 1997;47(1):9–14.
lin-like growth factor-I receptor. J Clin Endocrinol 150. Frueh BR, Musch DC, Garber FW. Exo-
Metab. 2004;89(10):5076–5080. phthalmometer readings in patients with Graves’ eye
136. Tramontano D, Cushing GW, Moses AC, Ingbar SH. disease. Ophthalmic Surg. 1986;17(1):37–40.
Insulin-like growth factor-i stimulates the growth of 151. Yamamoto K, Saito K, Takai T, Yoshida S. Diagnosis
rat thyroid cells in culture and synergizes the stimu- of exophthalmos using orbital ultrasonography and
lation of dna synthesis induced by tsh and graves’- treatment of malignant exophthalmos with steroid
IgG. Endocrinology. 1986;119(2):940–2. therapy, orbital radiation therapy, and plasmapheresis.
137. Tsui S, Naik V, Hoa N, et al. Evidence for an associ- Prog Clin Biol Res. 1983;116:189–205.
ation between thyroid-stimulating hormone and insu- 152. Yamamoto K, Itoh K, Yoshida S, et al. A quantitative
lin-like growth factor 1 receptors: a tale of two analysis of orbital soft tissue in Graves’ disease based
antigens implicated in graves’ disease. J Immunol. on B-mode ultrasonography. Endocrinol Jpn. 1979;26
2008;181(6):4397–405. (2):255–261.
138. Smith TJ, Kahaly GJ, Ezra DG, et al. Teprotumumab 153. Grove AS, Jr. Orbital disease: examination and
for thyroid-associated Ophthalmopathy. N Engl J diagnostic evaluation. Ophthalmology. 1979;86
Med. 2017;376(18):1748–61. (5):854–863.
139. Douglas RS, Kahaly GJ, Patel A, et al. 154. Hallin ES, Feldon SE. Graves’ ophthalmopathy: I.
Teprotumumab for the Treatment of Active Thyroid Simple CT estimates of extraocular muscle volume.
Eye Disease. N Engl J Med. 2020;382(4):341–352. Br J Ophthalmol. 1988;72(9):674–677.
140. Jacobson DH, Gorman CA. Endocrine 155. Kamminga N, Jansonius NM, Pott JW, Links TP.
ophthalmopathy: current ideas concerning etiology, Unilateral proptosis: the role of medical history. Br J
pathogenesis, and treatment. Endocr Rev. 1984;5 Ophthalmol. 2003;87(3):370–371.
(2):200–220. 156. Anderson RL, Tweeten JP, Patrinely JR, Garland PE,
141. Bartley GB. The differential diagnosis and classifica- Thiese SM. Dysthyroid optic neuropathy without
tion of eyelid retraction. Ophthalmology. 1996;103 extraocular muscle involvement. Ophthalmic Surg.
(1):168–176. 1989;20(8):568–574.
142. Cockerham KP, Hidayat AA, Brown HG, Cockerham 157. Forbes G, Gorman CA, Gehring D, Baker HL, Jr.
GC, Graner SR. Clinicopathologic evaluation of the Computer analysis of orbital fat and muscle volumes
Mueller muscle in thyroid-associated orbitopathy. in Graves ophthalmopathy. AJNR Am J Neuroradiol.
Ophthalmic Plast Reconstr Surg. 2002;18(1):11–17. 1983;4(3):737–740.
143. Cruz AA, Akaishi PM, Coelho RP. Quantitative com- 158. Gorman CA. The measurement of change in Graves’
parison between upper eyelid retraction induced vol- ophthalmopathy. Thyroid. 1998;8(6):539–543.
untarily and by Graves orbitopathy. Ophthalmic Plast 159. Fells P, Kousoulides L, Pappa A, Munro P, Lawson J.
Reconstr Surg. 2003;19(3):212–215. Extraocular muscle problems in thyroid eye disease.
144. Esmaeli-Gutstein B, Hewlett BR, Pashby RC, Eye (Lond). 1994;8 ( Pt 5):497–505.
Oestreicher J, Harvey JT. Distribution of adrenergic 160. Flanders M, Hastings M. Diagnosis and surgical man-
receptor subtypes in the retractor muscles of the upper agement of strabismus associated with thyroid-related
eyelid. Ophthalmic Plast Reconstr Surg. 1999;15 orbitopathy. J Pediatr Ophthalmol Strabismus.
(2):92–99. 1997;34(6):333–340.
145. Hamada N, Okamoto Y, Yoshida H, Tsumura K, 161. Murakami Y, Kanamoto T, Tuboi T, Maeda T, Inoue
Nakamura Y, Noh JY. Sympathetic overactivity in Y. Evaluation of extraocular muscle enlargement in
the development of eyelid retraction in a patient dysthyroid ophthalmopathy. Jpn J Ophthalmol.
with euthyroid Graves’ disease evaluated by accom- 2001;45(6):622–627.
modation. Endocr J. 2000;47(5):623–628.
20 Y. Wang and R. S. Douglas

162. Knapp P. Special types of muscle anomalies associ- involvement. J Clin Endocrinol Metab. 1997;82
ated with Graves’ disease. Ophthalmology. 1979;86 (4):1027–1030.
(12):2081–2084. 178. Riemann CD, Foster JA, Kosmorsky GS. Direct
163. Wu HJ, Tsai RK. Thyroid-associated orbitopathy with orbital manometry in patients with thyroid-associated
superior oblique muscle involvement: a case report. orbitopathy. Ophthalmology. 1999;106(7):1296–
Kaohsiung J Med Sci. 2004;20(2):86–89. 1302.
164. Aydin K, Guven K, Sencer S, Cikim A, Gul N, 179. Cockerham KP, Pal C, Jani B, Wolter A, Kennerdell
Minareci O. A new MRI method for the quantitative JS. The prevalence and implications of ocular hyper-
evaluation of extraocular muscle size in thyroid tension and glaucoma in thyroid-associated
ophthalmopathy. Neuroradiology. 2003;45(3):184– orbitopathy. Ophthalmology. 1997;104(6):914–917.
187. 180. Dev S, Damji KF, DeBacker CM, Cox TA, Dutton JJ,
165. Firbank MJ, Coulthard A. Evaluation of a technique Allingham RR. Decrease in intraocular pressure after
for estimation of extraocular muscle volume using 2D orbital decompression for thyroid orbitopathy. Can J
MRI. Br J Radiol. 2000;73(876):1282–1289. Ophthalmol. 1998;33(6):314–319.
166. Given-Wilson R, Pope RM, Michell MJ, Cannon R, 181. Rootman J. Retrobulbar pressures in dysthyroid optic
McGregor AM. The use of real-time orbital ultra- neuropathy. Br J Ophthalmol. 1996;80(12):1034.
sound in Graves’ ophthalmopathy: a comparison 182. Suttorp-Schulten MS, Tijssen R, Mourits MP,
with computed tomography. Br J Radiol. 1989;62 Apkarian P. Contrast sensitivity function in Graves’
(740):705–709. ophthalmopathy and dysthyroid optic neuropathy. Br
167. Spierer A, Eisenstein Z. The role of increased intra- J Ophthalmol. 1993;77(11):709–712.
ocular pressure on upgaze in the assessment of Graves 183. Gasser P, Flammer J. Optic neuropathy of Graves’
ophthalmopathy. Ophthalmology. 1991;98 disease. A report of a perimetric follow-up.
(10):1491–1494. Ophthalmologica. 1986;192(1):22–27.
168. Gamblin GT, Harper DG, Galentine P, Buck DR, 184. Rundle F, Wilson C. Ophthalmoplegia in Grave’s
Chernow B, Eil C. Prevalence of increased intraocular disease. Clin Sci. 1944;5:17–29.
pressure in Graves’ disease–evidence of frequent sub- 185. Rundle F, Wilson C. Bulging of the eyelids with
clinical ophthalmopathy. N Engl J Med. 1983;308 exophthalmos. Clin Sci. 1944;5:31–45.
(8):420–424. 186. Mills MD, Coats DK, Donahue SP, Wheeler DT,
169. Fishman DR, Benes SC. Upgaze intraocular pressure American Academy of O. Strabismus surgery for
changes and strabismus in Graves’ ophthalmopathy. J adults: a report by the American Academy of Oph-
Clin Neuroophthalmol. 1991;11(3):162–165. thalmology. Ophthalmology. 2004;111(6):1255–
170. Simonsz HJ, Kommerell G. Increased muscle ten- 1262.
sion and reduced elasticity of affected muscles in 187. Streeten DH, Anderson GH, Jr., Reed GF, Woo P.
recent-onset Graves’ disease caused primarily by Prevalence, natural history and surgical treatment of
active muscle contraction. Doc Ophthalmol. exophthalmos. Clin Endocrinol (Oxf). 1987;27
1989;72(3–4):215–224. (1):125–133.
171. Trobe JD. Optic nerve involvement in dysthyroidism. 188. Selva D, Chen C, King G. Late reactivation of thyroid
Ophthalmology. 1981;88(6):488–492. orbitopathy. Clin Exp Ophthalmol. 2004;32(1):46–50.
172. Giaconi JA, Kazim M, Rho T, Pfaff C. CT scan 189. Kalmann R, Mourits MP. Late recurrence of unilateral
evidence of dysthyroid optic neuropathy. Ophthalmic graves orbitopathy on the contralateral side. Am J
Plast Reconstr Surg. 2002;18(3):177–182. Ophthalmol. 2002;133(5):727–729.
173. McKeag D, Lane C, Lazarus JH, et al. Clinical fea- 190. Chou PI, Feldon SE. Late onset dysthyroid optic
tures of dysthyroid optic neuropathy: a European neuropathy. Thyroid. 1994;4(2):213–216.
Group on Graves’ Orbitopathy (EUGOGO) survey. 191. Patel P, Khandji J, Kazim M. Recurrent Thyroid Eye
Br J Ophthalmol. 2007;91(4):455–458. Disease. Ophthalmic Plast Reconstr Surg. 2015;31
174. Kennerdell JS, Rosenbaum AE, El-Hoshy MH. Api- (6):445–448.
cal optic nerve compression of dysthyroid optic neu- 192. Werner SC. Modification of the classification of the
ropathy on computed tomography. Arch Ophthalmol. eye changes of Graves’ disease. Am J Ophthalmol.
1981;99(5):807–809. 1977;83(5):725–727.
175. Bartalena L, Marcocci C, Pinchera A. Treating severe 193. Werner SC. Classification of the eye changes of
Graves’ ophthalmopathy. Baillieres Clin Endocrinol Grave’s disease. J Clin Endocrinol Metab. 1969;29
Metab. 1997;11(3):521–536. (7):982–984.
176. Barrett L, Glatt HJ, Burde RM, Gado MH. Optic 194. Dickinson AJ, Perros P. Controversies in the clinical
nerve dysfunction in thyroid eye disease: CT. Radiol- evaluation of active thyroid-associated orbitopathy:
ogy. 1988;167(2):503–507. use of a detailed protocol with comparative photo-
177. Salvi M, Spaggiari E, Neri F, et al. The study of visual graphs for objective assessment. Clin Endocrinol
evoked potentials in patients with thyroid-associated (Oxf). 2001;55(3):283–303.
ophthalmopathy identifies asymptomatic optic nerve 195. Muller-Forell W, Pitz S, Mann W, Kahaly GJ. Neuro-
radiological diagnosis in thyroid-associated
Thyroid Eye Disease 21

orbitopathy. Exp Clin Endocrinol Diabetes. 1999;107 ophthalmopathy: its predictive value for therapeutic
Suppl 5:S177–183. outcome of immunosuppressive therapy. Thyroid.
196. Enzmann D, Marshal WH, Jr., Rosenthal AR, Kriss 1992;2(4):299–305.
JP. Computed tomography in Graves’ 205. Ott M, Breiter N, Albrecht CF, Pradier O, Hess CF,
ophthalmopathy. Radiology. 1976;118(3):615–620. Schmidberger H. Can contrast enhanced MRI predict
197. Rothfus WE, Curtin HD. Extraocular muscle enlarge- the response of Graves’ ophthalmopathy to orbital
ment: a CT review. Radiology. 1984;151(3):677–681. radiotherapy? Br J Radiol. 2002;75(894):514–517.
198. Ben Simon GJ, Syed HM, Douglas R, McCann JD, 206. Cakirer S, Cakirer D, Basak M, Durmaz S, Altuntas Y,
Goldberg RA. Extraocular muscle enlargement with Yigit U. Evaluation of extraocular muscles in the
tendon involvement in thyroid-associated edematous phase of Graves ophthalmopathy on con-
orbitopathy. Am J Ophthalmol. 2004;137(6):1145– trast-enhanced fat-suppressed magnetic resonance
1147. imaging. J Comput Assist Tomogr. 2004;28(1):80–
199. Glatt HJ. Optic nerve dysfunction in thyroid eye dis- 86.
ease: a clinician’s perspective. Radiology. 1996;200 207. Utech CI, Khatibnia U, Winter PF, Wulle KG. MR T2
(1):26–27. relaxation time for the assessment of retrobulbar
200. Ramsay I, Meire H. Ultrasonics in the diagnosis of inflammation in Graves’ ophthalmopathy. Thyroid.
thyroid disease. Clin Radiol. 1975;26(2):191–197. 1995;5(3):185–193.
201. Gerding MN, Prummel MF, Wiersinga WM. Assess- 208. Hoh HB, Laitt RD, Wakeley C, et al. The STIR
ment of disease activity in Graves’ ophthalmopathy sequence MRI in the assessment of extraocular mus-
by orbital ultrasonography and clinical parameters. cles in thyroid eye disease. Eye (Lond). 1994;8 ( Pt
Clin Endocrinol (Oxf). 2000;52(5):641–646. 5):506–510.
202. Yokoyama N, Nagataki S, Uetani M, Ashizawa K, 209. Laitt RD, Hoh B, Wakeley C, et al. The value of the
Eguchi K. Role of magnetic resonance imaging in the short tau inversion recovery sequence in magnetic
assessment of disease activity in thyroid-associated resonance imaging of thyroid eye disease. Br J
ophthalmopathy. Thyroid. 2002;12(3):223–227. Radiol. 1994;67(795):244–247.
203. Mayer E, Herdman G, Burnett C, Kabala J, Goddard 210. Ohnishi T, Noguchi S, Murakami N, et al. Extraocular
P, Potts MJ. Serial STIR magnetic resonance imaging muscles in Graves ophthalmopathy: usefulness of T2
correlates with clinical score of activity in thyroid relaxation time measurements. Radiology. 1994;190
disease. Eye (Lond). 2001;15(Pt 3):313–318. (3):857–862.
204. Hiromatsu Y, Kojima K, Ishisaka N, et al. Role of
magnetic resonance imaging in thyroid-associated

You might also like