You are on page 1of 8

Review 789

Future Prospects for the Treatment of Graves’


Hyperthyroidism and Eye Disease

Authors S. Neumann1, R. F. Place2, C. C. Krieger1, M. C. Gershengorn1


1
Affiliations  Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases,
National Institutes of Health, Bethesda, USA
2
 Nova Therapeutics LLC, E Foothill Blvd, Pasadena, USA

Downloaded by: Collections and Technical Services Department. Copyrighted material.


Key words Abstract ease. Herein, we review some aspects of what is

▶ Graves’ hyperthyroidism
▼ known about the pathogenesis of these 2 major

▶ Graves’ orbitopathy
Although there are adequate therapies for Graves’ components of Graves’ disease, specifically the

▶ TSH receptor (TSHR)
hyperthyroidism, mild to moderate Graves’ orbit- apparent important roles of the TSH and IGF-1

▶ IGF-1 receptor (IGF-1R)
opathy (GO) is usually treated symptomatically receptors, and thereafter describe future thera-
whereas definitive therapy is reserved for severe, peutic approaches directed at these receptors.
vision-threatening GO. Importantly, none of the We propose that targeting these receptors will
treatment regimens for Graves’ disease used yield effective and better tolerated treatments for
today are directed at the pathogenesis of the dis- Graves’ disease, especially for GO.

TSHR and IGF-1R Appear to be Disease with GD [5]. GO involves inflammation of orbital
Targets in Graves’ Disease tissue and extensive remodeling of orbital con-
▼ nective tissues including enlargement of extra-
The thyrotropin [thyroid-stimulating hormone ocular muscles, increased numbers of adipose
(TSH)] receptor (TSHR) is a major regulator of cells, and excessive deposition of hyaluronan
thyroid function by controlling the size and num- [hyaluronic acid (HA)] [6]. The underlying mech-
received 19.03.2015 ber of thyrocytes, and the synthesis and secretion anisms of the pathogenesis of GO remain unclear.
accepted 26.06.2015 of thyroid hormones [1]. In recent years, it has TSHR expression in orbital fat tissue and extra-
become evident that TSHR is also expressed in a ocular muscles [7] suggests that TSHR is one of
Bibliography variety of extrathyroidal tissues including, for the primary targets in orbital tissues in patients
DOI http://dx.doi.org/ example, orbital preadipocytes/fibroblasts, bone, with GO. A low TSHR abundance has been
10.1055/s-0035-1555901 observed in cultured orbital fibroblasts and in
white and brown adipose tissue, peripheral
Published online:
blood cells, anterior pituitary, and hypothalamus normal adipose tissue; however, TSHR expres-
July 21, 2015
Horm Metab Res 2015; [2]. Although of high interest and being studied sion is elevated in differentiated human orbital
47: 789–796 intensively, the functional role and physiological fibroblasts in vitro [8, 9]. Lastly, there is a direct
© Georg Thieme Verlag KG importance of the TSHR in these extrathyroidal correlation between the levels of TSAbs and the
Stuttgart · New York tissues are not yet well understood. severity of GO, as well as a prognostic value for
ISSN 0018-5043 TSHR plays a major role in several thyroid pathol- TSAbs in GO outcome [10].
ogies including hyperthyroidism, hypothyroid- The insulin-like growth factor 1 (IGF-1) receptor
Correspondence (IGF-1R) has been suggested to be another impor-
ism, and thyroid tumors [3]. TSHR is the main
S. Neumann
target of thyroid-stimulating auto-antibodies tant target in GO and IGF-1/IGF-1R signaling
National Institute of Diabetes
and Digestive and Kidney (TSAbs) in patients with Graves’ disease (GD). GD appears to participate in the pathogenesis of auto-
Diseases is an autoimmune disease with a prevalence of immune diseases in general. Although the role(s)
National Institutes of Health about 1 % of the U.S. population that is caused by of IGF-1R signaling is not completely understood,
Laboratory of Endocrinology persistent, unregulated TSAb stimulation of it is known that IGF-1 influences several aspects
and Receptor Biology TSHRs on thyroid cells causing overproduction of of immunity by, for example, affecting thymic B
50 South Drive
thyroid hormones leading to hyperthyroidism [3]. and T cells development [11]. IGF-1R has been
Bethesda
GD is the most common form of hyperthyroidism found to be overexpressed on orbital fibroblasts
MD 20892-8029
USA in iodine-replete areas [4]. Graves’ orbitopathy of patients with GO compared with those from
Tel.:  + 1/301/451 6307 (GO), also known as Graves’ ophthalmopathy or non-Graves’ controls [12]. A functional relation-
Fax:  + 1/301/480 4214 thyroid eye disease, is an extrathyroidal manifes- ship between TSH- and IGF-1 mediated pathways
susannen@intra.niddk.nih.gov tation of GD with a prevalence of 25 % in patients was first described by Tramontano et al. in rat

Neumann S et al. Future Treatment of Graves’ Disease  …  Horm Metab Res 2015; 47: 789–796
790 Review

thyroid cells [13], and co-localization of TSHR and IGF-1R has Management of GO is still a major challenge, and the existing
been demonstrated [14]. IGF-1R and TSHR can be co-immuno- therapies are not uniformly effective and have substantial side
precipitated from human thyrocytes and orbital fibroblasts. effects as well. Treatment strategies for patients with severe eye
Selective activation of both receptors by their cognate ligands disease include oral or intravenous corticosteroids, orbital radi-
TSH and IGF-1 has been shown to stimulate HA secretion by otherapy or orbital decompression surgery [5, 26]. Rituximab
Graves’ orbital fibroblasts (GOFs), which are fibroblast-like cells (RTX), a humanized chimeric anti-CD20 monoclonal antibody
derived from tissue excised in decompression surgery for GO that depletes both B lymphocytes in the intermediate stages of
[15, 16]. Recently, our group provided evidence for bi-directional maturation and short-lived plasma cells, has been reported to be
crosstalk between TSHR and IGF-1R that leads to increased effective for the treatment of active moderate to severe GO
TSAb-induced HA production in GOFs [17]. Our current knowl- [27, 28]. However, very recent randomized controlled studies
edge of the pathogenesis of GO supports the idea that TSHR and have led to inconsistent results. Salvi et al. [29] report a better
IGF-1R on orbital fibroblasts are primarily involved [10, 17, 18]. therapeutic outcome of RTX in active moderate to severe GO
when compared with intravenous administration of methyl-
prednisolone and suggest a disease-modifying effect of the drug.
Current Treatments for Graves’ Hyperthyroidism and GO In contrast, the randomized controlled trial by the group of Stan
▼ et al. [30] did not show a benefit of RTX over placebo. More stud-
Untreated Graves’ hyperthyroidism results in increased morbid- ies will be necessary to draw a definitive conclusion.
ity and mortality mainly due to cardiovascular and skeletal Advantages and disadvantages of current treatment strategies as

Downloaded by: Collections and Technical Services Department. Copyrighted material.


(osteoporosis) complications [19]. Moreover, GD has a negative well as causes for the incomplete effectiveness of available thera-
long-term influence on the quality of life, due to either the dis- pies are reviewed by Bartalena [5]. Variable success of treatment of
ease process itself or its treatment [20]. Treatment options for GO is caused in part due to incomplete understanding of the
GD are thyroidectomy and radioactive iodine (RAI) therapy that pathogenesis of GO and persisting difficulties in diagnosis and
aim at preventing recurrence of hyperthyroidism but often induce early assessment of GO activity and severity in clinical practice [5].
permanent hypothyroidism requiring thyroid hormone replace- Current treatment options for GD and GO are also less than opti-
ment, or anti-thyroid medication (methimazole, carbimazole, mal because they target the signs and symptoms of these dis-
­propylthiouracil) to restore the euthyroid state while awaiting eases rather than the pathogenic mechanisms. None of the
resolution of the autoimmune process and disease remission. currently approved therapies target TSHR or IGF-1R directly. In
None of these therapies is optimal. Thyroid surgery and RAI ther- an ideal case the same drug would treat GD hyperthyroidism and
apy may have acute morbidity and usually require lifelong thyroid GO due to the close link between these diseases [31]. Future treat-
hormone replacement. The antithyroid drugs methimazole and ment strategies might include TSHR-blocking antibodies or small
propylthiouracil have potential side effects including agranulocy- molecule antagonists, which directly act at the TSHR. The involve-
tosis and hepatotoxicity. Side effects of methimazole are dose- ment of the IGF-1R in GO suggests targeting the IGF-1R as a com-
related, whereas those of propylthiouracil are less clearly related to plementary therapy for GO [18, 32]. In summary, the development
dose [21]. Long-term low-dose treatment with methimazole has of treatment strategies that target the TSHR or IGF-1R alone or in
been proven safe and successful in sustained control of hyperthy- combination is desirable. Such potential new therapies are sum-
roidism and is in particular a recommended treatment option for marized in ●  ▶  Table 1 and will be described in more detail below.
patients with Graves’ orbitopathy [22–24]. However, the relapse
rate upon discontinuation of antithyroid drugs is high [25].

Table 1  Potential novel therapies for Graves’ hyperthyroidism and Graves’ orbitopathy (GO) that directly target TSHR or IGF-1R.

Potential Treatment Drug Type Mechanism of Action Pros Cons


Antibody 5C9 Monoclonal TSHR blocking activity Potential for treatment of hyperthyroidism Parenteral administration
Antibody K1–70 antibody and GO Difficult to produce in large
Inverse agonist activity quantities
High affinity Intensive quality controls
Long half-life (weeks) are required
NCGC00229600 (ANTAG2) Small molecule TSHR blocking activity Potential for treatment of hyperthyroidism Short half-life
NCGC00242364 (ANTAG3) and GO
Inverse agonist activity
Oral administration
Relative ease of production in large quantities
Teprotumumab (RV001, Monoclonal IGF-1R blocking Phase 2 clinical trial in patients with GO Parenteral administration
R1507) antibody activity Long half-life (weeks) Difficult to produce in large
quantities
Intensive quality controls
are required
Nonspecific for GO
Linsitinib (OSI-906) Small molecule IGF-1R blocking Phase 3 clinical trial for adrenocortical Short half-life
activity carcinoma Nonspecific for GO
Phase 2 clinical trial for lung, ovarian, and Unknown impact on GO
prostate cancer
Oral administration
Relative ease of production in large quantities

Neumann S et al. Future Treatment of Graves’ Disease  …  Horm Metab Res 2015; 47: 789–796
Review 791

TSHR Antagonists Small Molecule Antagonists for TSHR


▼ ▼
TSHR is a member of a small subfamily of related glycoprotein hor- Recently developed small molecule “drug-like” antagonists
mone receptors (GPHRs) that includes receptors for LH/chorionic (SMANTAGs) for TSHR are also promising drug candidates for
gonadotropin (CG) and FSH, within class A of the large family of G the treatment of Graves’ hyperthyroidism and GO [9, 41–43]. All
protein-coupled (7 transmembrane-spanning) receptors (GPCRs). SMANTAGs that have been reported to date bind to an allosteric
GPHRs are different than other members of class A GPCRs by site within the transmembrane domain of TSHR rather than the
­having a very large amino-terminal extracellular domains. TSH orthosteric site in the extracellular domain where TSH and
and TSAbs bind primarily to TSHR via interactions with regions TSAbs bind (see below). We reported the first well-characterized
within the extracellular domain [33]. The receptor signaling is neutral SMANTAG (NIDDK-CEB-52) that selectively antagonizes
induced by ligand binding and transduced by the intracellular TSHR in 2008 [44]. NIDDK-CEB-52 was shown to inhibit activa-
­surface of the transmembrane helices and the carboxyl-terminal tion of TSHR by TSH and TSAbs in a model cell system and inhib-
tail of TSHR interacting with G-proteins and arrestins [33, 34]. ited upregulation of thyroperoxidase (TPO) expression by these
Antagonists that inhibit TSHR signaling directly by binding to agonists in primary cultures of human thyrocytes. These find-
the TSHR are promising new drug treatments. With regard to ings represented proof of principle that SMANTAGs could serve
clinical usefulness, neutral antagonists (ligands that inhibit as drugs to treat patients with Graves’ hyperthyroidism. Subse-
receptor activation by agonists) could be used as leads to develop quently, we developed a series of TSHR antagonists by chemical
drugs to antagonize TSAb activation of TSHR in patients with modification of the scaffold of the TSHR agonist NCGC00161870

Downloaded by: Collections and Technical Services Department. Copyrighted material.


Graves’ hyperthyroidism and GO. Inverse agonists (ligands that [45]. Interestingly, all antagonists based on the NCGC00161870
inhibit receptor activation by agonists and additionally inhibit scaffold except one were inverse agonists. Only NCGC00242595
agonist-independent, also termed basal or constitutive receptor was a neutral antagonist at TSHR [46]. A neutral antagonist is a
signaling) could be used as leads to develop drugs to inhibit con- good tool to be able to distinguish between effects of TSH- or
stitutive TSHR signaling in patients with residual thyroid cancer TSAb-stimulated and basal TSHR activation, and therefore it is
and thereby treat them more effectively than by TSH suppres- useful as a probe of TSHR function in addition to its clinical
sion alone. Furthermore, patients with non-autoimmune hyper- potential.
thyroidism caused by constitutively activating germline
mutations [35] could benefit from an inverse agonist. Two types
of molecules that are TSHR antagonists have been developed in Therapeutic Potential of SMANTAGs in the Treatment
parallel, TSHR-blocking antibodies [36, 37] and small molecule of Hyperthyroidism
“drug-like” compounds [38] ( ● ▶  Table 1). ▼
NCGC00161856 was the first inverse agonist of TSHR to be
described [47]. It inhibited basal and TSH-stimulated signaling
Blocking Monoclonal TSHR Autoantibodies of TSHR and basal signaling by constitutively active mutant
▼ TSHRs in a model cell system. Treatment with NCGC00161856
The first human monoclonal blocking antibody was produced also lowered basal expression levels of 4 thyroid-specific genes
from B cells of a patient with Graves’ hyperthyroidism. This anti- – thyroglobulin, TPO, sodium iodide symporter (NIS), and TSHR
body, 5C9, has high affinity for the TSHR and blocks TSAb- and in primary cultures of normal human thyrocytes. These data
TSH-induced activation by inhibition of binding of TSAbs or TSH demonstrated that a SMANTAG could act as an inverse agonist in
to the extracellular domain and inhibits basal activity of TSHR as human thyroid cells.
well [39]. Another blocking TSHR antibody, K1–70, was obtained Further chemical modification led to the promising new ligand
from a patient with autoimmune thyroid eye disease who was ANTAG2 (NCGC00229600, ●  ▶  Fig. 1), which is also an allosteric
clinically hypothyroid [40]. The patient’s serum showed both inverse agonist that inhibited basal and TSH-stimulated signal-
stimulating and blocking activity at the TSHR and this observa- ing [41]. Our group demonstrated first that small molecule TSHR
tion provided the first evidence that stimulating and blocking agonists and antagonists bind in a pocket within the 7 trans-
TSHR autoantibodies can be produced in a patient at the same membrane helical bundle of TSHR [45, 48]. To have potential as
time. The functional characterization of the blocking antibodies therapy for Graves’ hyperthyroidism and GO, it would be impor-
5C9 and K1–70, their similarities and differences as well as the tant that an antagonist inhibits TSHR activation by the majority
potential binding sites at the ectodomain of the TSHR have been
reviewed recently [36]. From a therapeutic point of view, the
advantage of these “natural” inhibitors is the high affinity to the N
O
TSHR and their long half-life of several weeks. However, the O O
need for parenteral administration and the difficulty of genera- N
NHAc
N
tion, purification and quality control of these antibodies are lim-
N O N O
iting factors to their potential use in the clinic. Although TSHR H H
activation by the majority of tested TSAbs could be blocked by OMe OMe

5C9 and K1–70, it has been demonstrated that there are differ- ANTAG2 (NCGC00229600) ANTAG3 (NCGC00242364)
ences in the way 5C9 and K1–70 bind to the TSHR [37], which
might have an impact on the general applicability of blocking Fig. 1  Structures of TSHR small molecule antagonists. ANTAG2:
TSHR antibodies. These antibodies have not yet been studied for NCGC00229600, 2-[3-[(2,6-dimethylphenoxy)methyl]-4-methoxyphenyl]-
pharmacodynamics and potential side effects commonly associ- 3-(pyridin-3-ylmethyl)-1,2-dihydroquinazolin-4-one. ANTAG3:
NCGC00242364, N-[4-[[5-[3-(furan-2-ylmethyl)-4-oxo-1,2-dihydroquina-
ated with monoclonal antibody-based therapies including the risk
zolin-2-yl]-2-methoxyphenyl]methoxy]-3,5-dimethylphenyl]acetamide.
of mild immune reactions (i. e., allergic-like responses) in humans.

Neumann S et al. Future Treatment of Graves’ Disease  …  Horm Metab Res 2015; 47: 789–796
792 Review

of TSAbs. Because TSAbs bind to the extracellular domain of tive antagonist is so far the only small molecule candidate drug
TSHR, whereas SMANTAGs bind within the transmembrane to have been shown to effectively inhibit TSHR-mediated
domain, the likelihood is high that signal transduction initiated responses in vivo [50]. In a mouse model of thyroid gland stimu-
by the majority of TSAbs can be blocked by SMANTAGs. In pri- lation by endogenous TSH, ANTAG3 inhibited the elevation in
mary cultures of human thyrocytes, the inverse agonist ANTAG2 serum free T4. Furthermore, ANTAG3 inhibited the increases in
inhibited TSAb-induced increase in TPO gene expression by TPO and NIS mRNA expression caused by continuous adminis-
average 65 % by all 30 GD sera tested [41]. These data suggested tration of TRH. In a second mouse model, which is more relevant
that an antagonist like ANTAG2 would likely be an effective to GD, endogenous TSH effects were inhibited by administering
inhibitor of TSAbs from most, if not all, patients with GD and T3 [51] and thyroid glands were stimulated by administering the
could be used to treat the hyperthyroidism of GD. human monoclonal TSAb M22 [52] ( ● ▶  Fig. 2a, reproduced from

Independently, a TSHR antagonist developed at Merck Sharp & [50]). ANTAG3 inhibited the increase in free T4 and the eleva-
Dohme Corporation (Org 274179–0) was characterized [9, 43]. tions of TPO and NIS mRNAs caused by M22 ( ● ▶  Fig. 2b, c, repro-

In the first report [43], the authors showed that Org 274179–0 duced from [50]). Although acute administration of M22
was a TSHR inverse agonist that inhibited the basal, TSH- and antibody is not a model for the chronic presence of TSAbs found
TSAb-stimulated signaling in a model system expressing TSHRs, in GD, these data indicate that ANTAG3 can inhibit thyroid acti-
basal signaling by constitutively active mutant TSHRs in a model vation by a stimulating antibody in vivo. These data suggest that
system, and TSH- and TSAb-stimulated signaling in a rat thyroid a TSHR antagonist like ANTAG3 could be used to treat Graves’
cell line (FRTL-5). Org 274179–0 has the advantage of higher hyperthyroidism. These findings also suggest that SMANTAGs

Downloaded by: Collections and Technical Services Department. Copyrighted material.


nanomolar potency than the reported TSHR antagonists from would be able to inhibit TSHR signaling in vivo in extrathyroidal
our group [43]. This ligand changes neither 125I-TSH binding nor tissues.
the dissociation of 125I -TSH from the TSHR, which suggests that
Org274179–0, like ANTAG2, is an allosteric antagonist and most
likely interacts with the transmembrane domain of the TSHR. Potential Role for SMANTAGs in the Treatment of GO
However, Org274179–0 has a significant disadvantage in that it ▼
is a partial agonist at the LH receptor and also exhibits equipo- Orbital fibroblasts appear to be a primary target of the autoim-
tent antagonistic activity at the FSHR [43]. This is not completely mune attack in GO and functional TSHR has been found on GOFs
unexpected since the receptors for LH and FSH have their high- (see above). Recently, it has been shown that fibrocytes in the
est homology to TSHR within the 7 transmembrane domain [49] circulation expressing antigen CD34, which is found on hemat-
in which SMANTAGs bind. The TSHR antagonist ANTAG2 is also opoietic precursor cells, also express TSHR. TSHR-expressing
a nonselective antagonist for TSHR. ANTAG2 inhibits LH- and fibrocytes have been reported to be found at high levels in GO
FSH receptor activation as well, although with lower efficacy and patients and have been proposed to infiltrate orbital connective
potency (unpublished data). Chemical modification of ANTAG2 tissue and play an important role in the pathophysiology of GO
has led to the development of ANTAG3 (NCGC00242364, [18]. The population of GOFs that are studied in vitro may be
 ▶  Fig. 1) which exhibits 2 properties that are important for drug
● derived from resident orbital fibroblasts and fibroblasts derived
treatment in humans. First, ANTAG3 is selective for TSHR and from fibrocytes. To our knowledge, however, the data published
does not affect signaling by LH or FSH receptors. Secondly, on fibrocytes has not yet been confirmed by other investigators.
ANTAG3 inhibits TSHR activation in mice in vivo [50]. This selec-

Fig. 2  ANTAG3 inhibits the increases in the levels


of serum free T4 and TPO and NIS mRNAs in the
thyroid in mice stimulated by a single injection
of TSAb M22. a T3 (5 μg/day for 4 days) was
administered by daily intraperitoneal (IP) injection
and ANTAG3 was given for 3 days (2 mg/day) via
osmotic pump. On day 4, the animals were given
an IP injection of vehicle or ANTAG3 (2 mg) and
4 h later an IP injection of M22 (0.5 μg) or vehicle.
The animals were euthanized 24 h after the M22
injection on day 5. b Serum free T4 (FT4) levels.
c mRNA levels of TPO and NIS in thyroid gland
lysates (reproduced from [50]).

Neumann S et al. Future Treatment of Graves’ Disease  …  Horm Metab Res 2015; 47: 789–796
Review 793

Models of GO have been reported in animal studies [53, 54], but troversial. Varewijck et al. [59] concluded that in a subset of
none have been proven to be adequate models that reproducibly patients with GO, circulating antibodies may stimulate the IGF-
or consistently recapitulate the disease. Therefore, the majority 1R and therefore may be involved in GO pathogenesis. In con-
of the studies of GO have been performed in tissue culture using trast, Minich et al. [60] detected antibodies to the IGF-1 receptor
GOFs obtained from patients with GO at orbital decompression in 11 % of healthy controls and 10 % of patients with GO. IGF-1R
surgery. Experimental evidence supports the idea that activa- antibodies failed to stimulate IGF-1R signaling but inhibited IGF-
tion of TSHR by TSAbs leads to proliferation of preadipocyte 1-induced activation of the receptor. The authors concluded that
fibroblasts or adipocytes, increase in HA production and differ- IGF-1R antibodies do not contribute to the pathogenesis of GO.
entiation of a subset of preadipocytes into mature adipocytes. Serum IGF-1, IGF-II and IGF-binding proteins are present at nor-
Therefore, showing that TSHR antagonists inhibit activation of mal levels in GO patients [61]. Therefore, one hypothesis is that
TSHRs on GOFs would be an important step in the development local production of IGF-1, acting in autocrine or paracrine fash-
of a medical therapy for GO. ion, might be involved in the autoimmune reaction [10]. In sum-
Two studies demonstrated in parallel that SMANTAGs can inhibit mary, the major contributing IGF-1R activating ligand in the
TSHR activation in GOFs. Van Zeijl et al. showed that Org pathogenesis of GO, that is, IGF-1R-stimulating antibodies or
274179–0 inhibited TSH-, TSAb- and M22-stimulated cAMP pro- endogenous IGF-1, has yet to be determined ( ● ▶  Fig. 3, see below).

duction by GOFs that had been made to differentiate into adipo- Strategies for interrupting IGF-1R signaling directly at the recep-
cytes in vitro [9]. Our group determined the effects of ANTAG2 tor are being developed ( ● ▶  Table 1). Teprotumumab (RV001,

on undifferentiated GOFs and GOFs differentiated into adipo- R1507) is a human monoclonal anti-IGF-1R antibody that antag-

Downloaded by: Collections and Technical Services Department. Copyrighted material.


cytes. GOFs exhibited higher absolute levels of basal and forsko- onizes receptor activation by binding to the extracellular
lin-stimulated cAMP production than differentiated adipocytes. α-subunit domain [62]. In fibrocytes, Teprotumumab reduced
Consistent with previous findings, TSH stimulated cAMP pro- cell surface expression of IGF-1R and TSHR, inhibited IGF-1 and
duction in the majority of differentiated adipocyte strains and TSH-induced Akt phosphorylation, and reduced TSHR-mediated
less consistently in GOFs. ANTAG2 reduced both TSH- and IL-6 and IL-8 expression [63]. These findings are another impor-
M22-stimulated signaling in both cell types. Moreover, ANTAG2 tant indication for crosstalk between TSHR and IGF-1R and sup-
was a selective TSHR antagonist in these GOFs because it did not port the therapeutic potential of this blocking IGF-1R antibody.
inhibit signaling by activation of prostaglandin D2 receptors, A multi-center placebo-controlled therapeutic trial specifically
which use the same cAMP signaling pathway as TSHRs [42]. directed at examining the impact of teprotumumab on the activ-
A follow-up study demonstrated that ANTAG2 inhibited TSAb- ity and progression of moderate to severe GO is being conducted.
induced HA production in undifferentiated GOFs [55]. The find- The currently performed phase 2 clinical trial will evaluate the
ings reported with ANTAG2 and Org274179–0 support a safety, tolerability and effectiveness of teprotumumab in the
potential role of SMANTAGs in the treatment of GO. treatment of GO (https://clinicaltrials.gov/show/NCT01868997).
In summary, TSHR SMANTAGs have potential as treatment for Teprotumumab is administered every 3 weeks by intravenous
Graves’ hyperthyroidism and GO. In comparison to antithyroid infusion. Considering parenteral administration and high pro-
drugs, SMANTAGs have the advantage of targeting the TSHR duction costs for a therapeutically used antibody, a small mole-
directly and therefore, the same drug could be used to treat both cule antagonist for IFG-1R may exhibit important advantages
major components of GD, hyperthyroidism and orbitopathy. due to oral administration and ease of production ( ● ▶  Table 1).

These advantages are also valid for the use of monoclonal block- Linsitinib (OSI-906) is an orally bioavailable small-molecule IGF-
ing TSHR antibodies (as described above). While SMANTAGs 1R kinase inhibitor [64]. Linsitinib potently inhibits ligand-
have a short half-life compared to antibodies, these small organic dependent auto-phosphorylation of IGF-1R in cells, while
molecules have the advantages of oral administration and rela- displaying a high degree of selectivity vs. a wide panel of protein
tive ease of production and quality control ( ● ▶  Table 1). Since kinases. Linsitinib prevents ligand-induced activation of down-
chronic treatment with TSHR antagonists might be necessary to stream pathways including pAKT and pERK1/2 and, therefore,
control hyperthyroidism and associated orbitopathy, an adjusted inhibits proliferation in a variety of tumor cell lines and has
lower dose of a SMANTAG could be administered to control demonstrated in vivo efficacy in tumor models [64]. Phase 2
hyperthyroidism and to avoid hypothyroidism. clinical trials with linsitinib for the treatment of small cell lung
cancer, ovarian cancer and prostate cancer, and a phase 3 clinical
trial for the treatment of patients with adrenocortical carcinoma
IGF-1R Antagonists have been completed (https://clinicaltrials.gov/ct2/results?term 
▼ = OSI-906).
The IGF-1R is a transmembrane receptor that belongs to the A major challenge in the development of IGF-1R inhibitors is to
class of tyrosine kinase receptors and is activated and auto- avoid blocking the closely related insulin receptor (IR) that regu-
phosphorylated following IGF-1 binding [56]. The IGF-1R is lates glucose levels in the blood. Linsitinib is a dual inhibitor of
expressed in orbital fibroblasts and circulating fibrocytes, its IGF-1R and IR [64]. Activation of the insulin receptor induces
expression is increased in GO fibroblasts compared to orbital glucose uptake, and a decrease in IR signaling leads to hypergly-
fibroblasts from non-Graves’ patients, and may be acting in con- cemia. Two phase 1 trials have tested continuous or intermittent
cert with the TSHR in the pathophysiology of GO [18, 32]. The dosing of Linsitinib in patients with advanced solid tumors and
potential role of IGF-1R-mediated signaling in GO was first sug- hyperglycemia has been reported as an adverse side effect
gested when antibodies from GO patients were found to com- [65, 66]. Therefore, selective targeting of the IGF-1R with small
pete with binding of radiolabeled IGF-1 to the surface of GOFs molecule ligands is the major challenge due to the close rela-
[57]. It has been postulated that stimulating antibodies against tionship with the IR. An additional challenge for the use of IGF-1
the IGF-1R [11] or a subset of TSAbs that stimulate the IGF-1R inhibitors in the treatment of GO is the fact that the IGF-1R sign-
are contributing to GO pathogenesis [58] but data remain con- aling axis plays many essential roles in the growth, differentia-

Neumann S et al. Future Treatment of Graves’ Disease  …  Horm Metab Res 2015; 47: 789–796
794 Review

Downloaded by: Collections and Technical Services Department. Copyrighted material.


Fig. 3  Combination therapy that antagonizes TSHR and IGF-1R may be the most effective treatment of Graves’ orbitopathy (GO). Left panel. TSHR is
activated by thyroid stimulating antibodies (TSAbs). IGF-1R may be activated via cross-talk by TSHR, IGF-1R-activating antibodies or endogenous IGF-1.
Middle panels: TSAb-stimulated hyaluronan secretion by Graves’ orbital fibroblasts is inhibited by antagonizing either TSHR with a SMANTAG or IGF-1R with
linsitinib. TSHR and IGF-1R could also be inhibited by monoclonal TSHR blocking antibodies and monoclonal blocking IGF-1R antibodies, respectively (not
shown). Right panel: Combined treatment with submaximal doses of a TSHR antagonist and an IGF-1R antagonist may provide the most effective medical
therapy for GO with the fewest side effects.

tion and development of mammalian tissues [67]. Therefore,


inhibition of IGF-1R-mediated signaling by antibodies or small 1000
Control
molecule antagonists might lead to undesirable side effects in
normal tissue. 800 Linsitinib
ANTAG2
HA (µg/ml)

600
Future Therapy for GO – Hypothesis
▼ 400
Recent findings support the idea that targeting both TSHR and
IGF-1R may be advantageous in treating patients with GO. We
200
have shown that simultaneous stimulation of GOFs by TSH and
IGF-1 synergistically increased HA secretion [17] ( ● ▶  Fig. 3, left

panel); that is, these receptors are functionally interdependent. 0


Bidirectional crosstalk between TSHR and IGF-1R in GOFs Basal IGF-1 M22
induced by the TSAb M22 appears to both directly activate TSHR
and indirectly trigger IGF-1R signaling supporting a strategy for Fig. 4  Inhibition of IGF-1- and M22-stimulated HA secretion from GOFs
by TSHR antagonist ANTAG2 and IGF-1R antagonist linsitinib. GOFs were
targeting TSHR as the primary approach for GO therapy. How-
incubated in medium containing M22 (2 nM) or IGF-1 (13 nM) with or
ever, a combination therapy that antagonizes the activation of
without ANTAG2 (10 μM) or linsitinib (10 μM). After 5 days, the mediums
both receptors might be even more effective. were collected and HA was assayed. The maximally effective dose of
By using primary GOF cells in culture as an in vitro model for GO, linsitinib causes partial inhibition, whereas ANTAG2 completely inhibited
we have shown that antagonizing either IGF-1R or TSHR leads to M22-stimulated HA secretion.
inhibition of HA secretion stimulated by monoclonal TSAb M22
▶  Fig. 4; adapted from [17]). Our data show that a maximally
( ●
effective dose of linsitinib causes partial inhibition of HA secre- lower doses and fewer potential side effects as compared to
tion whereas ANTAG2, a TSHR antagonist, completely inhibited maximally effective doses of either drug alone. We, therefore,
HA secretion. These findings suggest that antagonism of TSHR is suggest that combined treatment with a TSHR antagonist and an
more effective in inhibiting HA secretion by TSAbs than antago- IGF-1R antagonist may provide the most effective medical ther-
nism of IGF-1R. However, since we have shown that there is apy for GO.
cross-talk between TSHR and IGF-1R on HA secretion when
GOFs are stimulated by TSAbs ( ● ▶  Fig. 3), it is possible that co-

treatment with IGF-1R inhibitors may further augment the ther- Acknowledgements
apeutic potential of targeted TSHR therapies. In preliminary ▼
experiments, we have found that co-treatment with SMANTAGs This work was supported by the Intramural Research Program of
and linsitinib have cooperative drug effects allowing improved the National Institute of Diabetes and Digestive and Kidney
efficacy at reduced doses when used in combination ( ● ▶  Fig. 3) ­Diseases, National Institutes of Health (Z01 DK011006, Z01
(unpublished data). This strategy would allow disease control at DK047044).

Neumann S et al. Future Treatment of Graves’ Disease  …  Horm Metab Res 2015; 47: 789–796
Review 795

Conflict of Interest 22 Azizi F, Yousefi V, Bahrainian A, Sheikholeslami F, Tohidi M, Mehrabi Y.


Long-term continuous methimazole or radioiodine treatment for
▼ hyperthyroidism. Arch Iran Med 2012; 15: 477–484
Nova Therapeutics LLC holds licenses for intellectual property 23 Elbers L, Mourits M, Wiersinga WM. Outcome of very long-term treat-
related to TSH receptor small molecule ligand technologies. Rob- ment with antithyroid drugs in Graves’ hyperthyroidism associated
ert F. Place is employed at Nova Therapeutics LLC. Susanne Neu- with Graves’ orbitopathy. Thyroid 2011; 21: 279–283
24 Laurberg P, Berman DC, Andersen S, Bülow Pedersen I. Sustained con-
mann, Christine C. Krieger, and Marvin C. Gershengorn have no trol of Graves’ hyperthyroidism during long-term low-dose antithy-
conflict of interest. roid drug therapy of patients with severe Graves’ orbitopathy. Thyroid
2011; 21: 951–956
25 Abraham P, Avenell A, McGeoch SC, Clark LF, Bevan JS. Antithyroid drug
References regimen for treating Graves’ hyperthyroidism. Cochrane Database Syst
1 Vassart G, Dumont JE. The thyrotropin receptor and the regulation of Rev 2010; CD003420
thyrocyte function and growth. Endocr Rev 1992; 13: 596–611 26 Bartalena L, Baldeschi L, Dickinson A, Eckstein A, Kendall-Taylor P, Mar-
2 Williams GR. Extrathyroidal expression of TSH receptor. Ann Endo- cocci C, Mourits M, Perros P, Boboridis K, Boschi A, Curro N, Daumerie
crinol (Paris) 2011; 72: 68–73 C, Kahaly GJ, Krassas GE, Lane CM, Lazarus JH, Marino M, Nardi M,
3 Davies TF, Ando T, Lin RY, Tomer Y, Latif R. Thyrotropin receptor- Neoh C, Orgiazzi J, Pearce S, Pinchera A, Pitz S, Salvi M, Sivelli P, Stahl
associated diseases: from adenomata to Graves disease. J Clin Invest M, von Arx G, Wiersinga WM. Consensus statement of the European
2005; 115: 1972–1983 Group on Graves’ orbitopathy (EUGOGO) on management of GO. Eur
4 Brent GA. Clinical practice. Graves’ disease. N Engl J Med 2008; 358: J Endocrinol 2008; 158: 273–285
2594–2605 27 Salvi M, Vannucchi G, Campi I, Curro N, Dazzi D, Simonetta S, Bonara P,
5 Bartalena L. Graves’ orbitopathy: imperfect treatments for a rare dis- Rossi S, Sina C, Guastella C, Ratiglia R, Beck-Peccoz P. Treatment of
ease. Eur Thyroid J 2013; 2: 259–269

Downloaded by: Collections and Technical Services Department. Copyrighted material.


Graves’ disease and associated ophthalmopathy with the anti-CD20
6 Perros P, Krassas GE. Graves orbitopathy: a perspective. Nat Rev Endo- monoclonal antibody rituximab: an open study. Eur J Endocrinol
crinol 2009; 5: 312–318 2007; 156: 33–40
7 Feliciello A, Porcellini A, Ciullo I, Bonavolonta G, Avvedimento EV, Fenzi G. 28 Salvi M, Vannucchi G, Beck-Peccoz P. Potential utility of rituximab for
Expression of thyrotropin-receptor mRNA in healthy and Graves’ dis- Graves’ orbitopathy. J Clin Endocrinol Metab 2013; 98: 4291–4299
ease retro-orbital tissue. Lancet 1993; 342: 337–338 29 Salvi M, Vannucchi G, Curro N, Campi I, Covelli D, Dazzi D, Simonetta S,
8 Valyasevi RW, Erickson DZ, Harteneck DA, Dutton CM, Heufelder AE, Guastella C, Pignataro L, Avignone S, Beck-Peccoz P. Efficacy of B-cell
Jyonouchi SC, Bahn RS. Differentiation of human orbital preadipocyte targeted therapy with rituximab in patients with active moderate
fibroblasts induces expression of functional thyrotropin receptor. to severe Graves’ orbitopathy: a randomized controlled study. J Clin
J Clin Endocrinol Metab 1999; 84: 2557–2562 Endocrinol Metab 2015; 100: 422–431
9 van Zeijl CJ, van Koppen CJ, Surovtseva OV, de Gooyer ME, Plate R, 30 Stan MN, Garrity JA, Carranza Leon BG, Prabin T, Bradley EA,
Conti P, Karstens WJ, Timmers M, Saeed P, Wiersinga WM, Miltenburg AM, Bahn RS. Randomized controlled trial of rituximab in patients with
Fliers E, Boelen A. Complete inhibition of rhTSH-, Graves’ disease IgG-, Graves’ orbitopathy. J Clin Endocrinol Metab 2015; 100: 432–441
and M22-induced cAMP production in differentiated orbital fibro- 31 Bahn RS. Emerging pharmacotherapy for treatment of Graves’ disease.
blasts by a low-molecular-weight TSHR antagonist. J Clin Endocrinol Expert Rev Clin Pharmacol 2012; 5: 605–607
Metab 2012; 97: E781–E785 32 Smith TJ, Hegedus L, Douglas RS. Role of insulin-like growth factor-1
10 Wiersinga WM. Autoimmunity in Graves’ ophthalmopathy: the result (IGF-1) pathway in the pathogenesis of Graves’ orbitopathy. Best Pract
of an unfortunate marriage between TSH receptors and IGF-1 recep- Res Clin Endocrinol Metab 2012; 26: 291–302
tors? J Clin Endocrinol Metab 2011; 96: 2386–2394 33 Kleinau G, Neumann S, Gruters A, Krude H, Biebermann H. Novel
11 Smith TJ. Insulin-like growth factor-I regulation of immune function: insights on thyroid-stimulating hormone receptor signal transduc-
a potential therapeutic target in autoimmune diseases? Pharmacol tion. Endocr Rev 2013; 34: 691–724
Rev 2010; 62: 199–236 34 Boutin A, Eliseeva E, Gershengorn MC, Neumann S. beta-Arrestin-1
12 Pritchard J, Han R, Horst N, Cruikshank WW, Smith TJ. Immunoglobulin mediates thyrotropin-enhanced osteoblast differentiation. FASEB
activation of T cell chemoattractant expression in fibroblasts from J 2014; 28: 3446–3455
patients with Graves’ disease is mediated through the insulin-like 35 Kleinau G, Biebermann H. Constitutive activities in the thyrotropin
growth factor I receptor pathway. J Immunol 2003; 170: 6348–6354 receptor: regulation and significance. Adv Pharmacol 2014; 70: 81–119
13 Tramontano D, Cushing GW, Moses AC, Ingbar SH. Insulin-like growth 36 Furmaniak J, Sanders J, Rees Smith B. Blocking type TSH receptor anti-
factor-I stimulates the growth of rat thyroid cells in culture and syn- bodies. Autoimmune Highlights 2013; 4: 11–26
ergizes the stimulation of DNA synthesis induced by TSH and Graves’- 37 Sanders J, Miguel RN, Furmaniak J, Rees Smith B. TSH receptor monoclo-
IgG. Endocrinology 1986; 119: 940–942 nal antibodies with agonist, antagonist, and inverse agonist activities.
14 Tsui S, Naik V, Hoa N, Hwang CJ, Afifiyan NF, Sinha HA, Gianoukakis Methods Enzymol 2010; 485: 393–420
AG, Douglas RS, Smith TJ. Evidence for an association between thyroid- 38 Gershengorn MC, Neumann S. Update in TSH receptor agonists and
stimulating hormone and insulin-like growth factor 1 receptors: a antagonists. J Clin Endocrinol Metab 2012; 97: 4287–4292
tale of 2 antigens implicated in Graves’ disease. J Immunol 2008; 181: 39 Sanders J, Evans M, Betterle C, Sanders P, Bhardwaja A, Young S, Roberts E,
4397–4405 Wilmot J, Richards T, Kiddie A, Small K, Platt H, Summerhayes S, Harris
15 Krieger CC, Gershengorn MC. A modified ELISA accurately measures R, Reeve M, Coco G, Zanchetta R, Chen S, Furmaniak J, Rees Smith B.
secretion of high molecular weight hyaluronan (HA) by Graves’ disease A human monoclonal autoantibody to the thyrotropin receptor with
orbital cells. Endocrinology 2014; 155: 627–634 thyroid-stimulating blocking activity. Thyroid 2008; 18: 735–746
16 Smith TJ, Hoa N. Immunoglobulins from patients with Graves’ disease 40 Evans M, Sanders J, Tagami T, Sanders P, Young S, Roberts E, Wilmot J,
induce hyaluronan synthesis in their orbital fibroblasts through the Hu X, Kabelis K, Clark J, Holl S, Richards T, Collyer A, Furmaniak J, Rees
self-antigen, insulin-like growth factor-I receptor. J Clin Endocrinol Smith B. Monoclonal autoantibodies to the TSH receptor, one with
Metab 2004; 89: 5076–5080 stimulating activity and one with blocking activity, obtained from the
17 Krieger CC, Neumann S, Place RF, Marcus-Samuels B, Gershengorn MC. same blood sample. Clin Endocrinol (Oxf) 2010; 73: 404–412
Bidirectional TSH and IGF-1 receptor cross-talk mediates stimulation 41 Neumann S, Eliseeva E, McCoy JG, Napolitano G, Giuliani C, Monaco F,
of hyaluronan secretion by Graves’ disease immunoglobins. J Clin Huang W, Gershengorn MC. A new small-molecule antagonist inhibits
Endocrinol Metab 2014; 100: 1071–1077 Graves’ disease antibody activation of the TSH receptor. J Clin Endo-
18 Smith TJ. TSH-receptor-expressing fibrocytes and thyroid-associated crinol Metab 2011; 96: 548–554
ophthalmopathy. Nat Rev Endocrinol 2015; 11: 171–181 42 Neumann S, Pope A, Geras-Raaka E, Raaka BM, Bahn RS, Gershengorn
19 Mai VQ, Burch HB. A stepwise approach to the evaluation and treat- MC. A drug-like antagonist inhibits thyrotropin receptor-mediated
ment of subclinical hyperthyroidism. Endocr Pract 2012; 18: 772–780 stimulation of cAMP production in Graves’ orbital fibroblasts. Thy-
20 Sundaresh V, Brito JP, Wang Z, Prokop LJ, Stan MN, Murad MH, roid 2012; 22: 839–843
Bahn RS. Comparative effectiveness of therapies for Graves’ hyper- 43 van Koppen CJ, de Gooyer ME, Karstens WJ, Plate R, Conti PG, van Achter-
thyroidism: a systematic review and network meta-analysis. J Clin berg TA, van Amstel MG, Brands JH, Wat J, Berg RJ, Lane JR, Milten-
Endocrinol Metab 2013; 98: 3671–3677 burg AM, Timmers CM. Mechanism of action of a nanomolar potent,
21 Cooper DS. Antithyroid drugs. N Engl J Med 2005; 352: 905–917 allosteric antagonist of the thyroid-stimulating hormone receptor. Br
J Pharmacol 2012; 165: 2314–2324

Neumann S et al. Future Treatment of Graves’ Disease  …  Horm Metab Res 2015; 47: 789–796
796 Review

44 Neumann S, Kleinau G, Costanzi S, Moore S, Jiang JK, Raaka BM, Thomas 56 LeRoith D, Werner H, Beitner-Johnson D, Roberts CT Jr. Molecular and
CJ, Krause G, Gershengorn MC. A low-molecular-weight antagonist for cellular aspects of the insulin-like growth factor I receptor. Endocr
the human thyrotropin receptor with therapeutic potential for hyper- Rev 1995; 16: 143–163
thyroidism. Endocrinology 2008; 149: 5945–5950 57 Weightman DR, Perros P, Sherif IH, Kendall-Taylor P. Autoantibodies to
45 Neumann S, Huang W, Titus S, Krause G, Kleinau G, Alberobello AT, IGF-1 binding sites in thyroid associated ophthalmopathy. Autoim-
Zheng W, Southall NT, Inglese J, Austin CP, Celi FS, Gavrilova O, Thomas munity 1993; 16: 251–257
CJ, Raaka BM, Gershengorn MC. Small-molecule agonists for the thyro- 58 Gianoukakis AG, Douglas RS, King CS, Cruikshank WW, Smith TJ.
tropin receptor stimulate thyroid function in human thyrocytes and Immunoglobulin G from patients with Graves’ disease induces inter-
mice. Proc Natl Acad Sci USA 2009; 106: 12471–12476 leukin-16 and RANTES expression in cultured human thyrocytes: a
46 Boutin A, Allen MD, Geras-Raaka E, Huang W, Neumann S, Gershengorn MC. putative mechanism for T-cell infiltration of the thyroid in autoim-
Thyrotropin receptor stimulates internalization-independent persis- mune disease. Endocrinology 2006; 147: 1941–1949
tent phosphoinositide signaling. Mol Pharmacol 2011; 80: 240–246 59 Varewijck AJ, Boelen A, Lamberts SW, Fliers E, Hofland LJ, Wiersinga WM,
47 Neumann S, Huang W, Eliseeva E, Titus S, Thomas CJ, Gershengorn MC. Janssen JA. Circulating IgGs may modulate IGF-I receptor stimulating
A small molecule inverse agonist for the human thyroid-stimulating activity in a subset of patients with Graves’ ophthalmopathy. J Clin
hormone receptor. Endocrinology 2010; 151: 3454–3459 Endocrinol Metab 2013; 98: 769–776
48 Jaschke H, Neumann S, Moore S, Thomas CJ, Colson AO, Costanzi S, Kle- 60 Minich WB, Dehina N, Welsink T, Schwiebert C, Morgenthaler NG,
inau G, Jiang JK, Paschke R, Raaka BM, Krause G, Gershengorn MC. A low Kohrle J, Eckstein A, Schomburg L. Autoantibodies to the IGF1 receptor
molecular weight agonist signals by binding to the transmembrane in Graves’ orbitopathy. J Clin Endocrinol Metab 2013; 98: 752–760
domain of thyroid-stimulating hormone receptor (TSHR) and lutein- 61 Krassas GE, Pontikides N, Kaltsas T, Dumas A, Frystyk J, Chen JW, Fly-
izing hormone/chorionic gonadotropin receptor (LHCGR). J Biol Chem vbjerg A. Free and total insulin-like growth factor (IGF)-I, -II, and IGF
2006; 281: 9841–9844 binding protein-1, -2, and -3 serum levels in patients with active thy-
49 Kleinau G, Krause G. Thyrotropin and homologous glycoprotein hor- roid eye disease. J Clin Endocrinol Metab 2003; 88: 132–135

Downloaded by: Collections and Technical Services Department. Copyrighted material.


mone receptors: structural and functional aspects of extracellular 62 Schnitzer T, Kuenkele KP, Rebers F, Van Vugt M, Klein C, Lanzendor-
signaling mechanisms. Endocr Rev 2009; 30: 133–151 fer M, Mundigl O, Parren PWHI, van de Winkel JGJV, Schumacher R.
50 Neumann S, Nir EA, Eliseeva E, Huang W, Marugan J, Xiao J, Dulcey AE, Characterization of a recombinant, fully human monoclonal antibody
Gershengorn MC. A selective TSH receptor antagonist inhibits stimu- directed against the human insulin-like growth factor-1 receptor. EJC
lation of thyroid function in female mice. Endocrinology 2014; 155: Suppl 2006; 4: 66–67
310–314 63 Chen H, Mester T, Raychaudhuri N, Kauh CY, Gupta S, Smith TJ, Douglas
51 Hamidi S, Aliesky H, Chen CR, Rapoport B, McLachlan SM. Variable sup- RS. Teprotumumab an IGF-1R blocking monoclonal antibody inhibits
pression of serum thyroxine in female mice of different inbred strains TSH and IGF-1 action in fibrocytes. J Clin Endocrinol Metab 2014;
by triiodothyronine administered in drinking water. Thyroid 2010; 99: E1635–E1640
20: 1157–1162 64 Mulvihill MJ, Cooke A, Rosenfeld-Franklin M, Buck E, Foreman K, Land-
52 Sanders J, Evans M, Premawardhana LD, Depraetere H, Jeffreys J, Rich- fair D, O’Connor M, Pirritt C, Sun Y, Yao Y, Arnold LD, Gibson NW, Ji QS.
ards T, Furmaniak J, Rees Smith B. Human monoclonal thyroid stimu- Discovery of OSI-906: a selective and orally efficacious dual inhibitor
lating autoantibody. Lancet 2003; 362: 126–128 of the IGF-1 receptor and insulin receptor. Future Med Chem 2009;
53 Moshkelgosha S, So PW, Deasy N, Diaz-Cano S, Banga JP. Cutting edge: 1: 1153–1171
retrobulbar inflammation, adipogenesis, and acute orbital congestion 65 Jones RL, Kim ES, Nava-Parada P, Alam S, Johnson FM, Stephens AW, Sim-
in a preclinical female mouse model of Graves’ orbitopathy induced by antov R, Poondru S, Gedrich R, Lippman SM, Kaye SB, Carden CP. Phase
thyrotropin receptor plasmid-in vivo electroporation. Endocrinology I study of intermittent oral dosing of the insulin-like growth factor-1
2013; 154: 3008–3015 and insulin receptors inhibitor OSI-906 in patients with advanced
54 Wiesweg B, Johnson KT, Eckstein AK, Berchner-Pfannschmidt U. Current solid tumors. Clin Cancer Res 2015; 21: 693–700
insights into animal models of Graves’ disease and orbitopathy. Horm 66 Puzanov I, Lindsay CR, Goff L, Sosman J, Gilbert J, Berlin J, Poondru S,
Metab Res 2013; 45: 549–555 Simantov R, Gedrich R, Stephens A, Chan E, Evans TR. A phase I study
55 Turcu AF, Kumar S, Neumann S, Coenen M, Iyer S, Chiriboga P, Gersh- of continuous oral dosing of OSI-906, a dual inhibitor of insulin-like
engorn MC, Bahn RS. A small molecule antagonist inhibits thyrotro- growth factor-1 and insulin receptors, in patients with advanced solid
pin receptor antibody-induced orbital fibroblast functions involved in tumors. Clin Cancer Res 2015; 21: 701–711
the pathogenesis of Graves ophthalmopathy. J Clin Endocrinol Metab 67 Tognon CE, Sorensen PH. Targeting the insulin-like growth factor 1
2013; 98: 2153–2159 receptor (IGF1R) signaling pathway for cancer therapy. Expert Opin
Ther Targets 2012; 16: 33–48

Neumann S et al. Future Treatment of Graves’ Disease  …  Horm Metab Res 2015; 47: 789–796

You might also like