PERSPECTIVE
published: 24 September 2020
doi: 10.3389/fendo.2020.559673
Frontiers in Endocrinology | www.frontiersin.org 1 September 2020 | Volume 11 | Article 559673
Edited by:
Ines Pineda-Torra,
University College London,
United Kingdom
Reviewed by:
Andrew Alt,
University of Michigan, United States
Andrew C. B. Cato,
Karlsruhe Institute of Technology
(KIT), Germany
*Correspondence:
Karolien De Bosscher
Specialty section:
This article was submitted to
Molecular and Structural
Endocrinology,
a section of the journal
Frontiers in Endocrinology
Received: 06 May 2020
Accepted: 26 August 2020
Published: 24 September 2020
Citation:
Van Moortel L, Gevaert K and
De Bosscher K (2020) Improved
Glucocorticoid Receptor Ligands:
Fantastic Beasts, but How to Find
Them? Front. Endocrinol. 11:559673.
doi: 10.3389/fendo.2020.559673
Improved Glucocorticoid Receptor
Ligands: Fantastic Beasts, but How
to Find Them?
Laura Van Moortel
1,2,3
, Kris Gevaert
2,3
and Karolien De Bosscher
1,2,3
*
1
Translational Nuclear Receptor Research (TNRR) Laboratory, VIB, Ghent, Belgium,
2
VIB Center for Medical Biotechnology,
VIB, Ghent, Belgium,
3
Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
Exogenous glucocorticoids are widely used in the clinic for the treatment of inflammatory
disorders and hematological cancers. Unfortunately, their use is associated wit h
debilitating side effects, including hyperglycemia, osteoporosis, mood swings, and
weight gain. Despite the continued efforts of pharma as well as academia, the search
for so-called selective glucocorticoid receptor modulators (SEGRMs), compounds with
strong anti-i nflammatory or anti-cancer properties but a reduced number or level of side
effects, has had limited success so far. Although monoclonal antibody therapies have
been successfully introduced for the treatment of certain disorders (such as anti- TNF for
rheumatoid arthritis), glucocorticoids remain the first-in-line option for many other chronic
diseases including asthma, multiple sclerosis, and multiple myeloma. This perspective
offers our opinion on why a continued search for SEGRMs remains highly relevant in an
era where small molecules are sometimes unrightfully considered old-fashioned. Besides
a discussion on which bottlenecks and pitfalls might have been overlooked in the past, we
elaborate on potential solutions and recent developments t hat may push future research
in the right direction.
Keywords: glucocorticoids, glucocorticoid receptor, selective GR modulators, drug discovery, inflammation, assay
development, GR, SEGRM
INTRODUCTION
Glucocorticoids (GCs) are endogenous steroidal hormones involved in metabolism, stress,
development, and immunity (
1). They exert their effects by binding the glucocorticoid receptor
(GR), a nuclear receptor (NR) consisting of an intrinsically disordered N-terminal domain (NTD),
a central DNA binding domain (DBD), a hinge region (HR), and a C-terminal ligand-binding
domain (LBD) (2). Upon ligand binding, GR typically translocates from the cytoplasm to
the nucleus where it acts as a genuine transcription factor to regulate target gene expression
via multiple mechanisms (Figure 1A), which are discussed in detail in (3). The discovery of
the anti-inflammatory effects of endogenous GCs preceded the development of synthetic GCs,
which are used to treat, among others, inflammatory disorders, and hematological cancers (4).
Unfortunately, the therapeutic efficacy of such exogenous GCs is, particularly for systemic use,
overshadowed by an unacceptably high number of undesired side effects such as hyperglycemia,
osteoporosis, mood swings, and weight gain (
5).
Van Moortel et al. Improved Glucocorticoid Receptor Ligands
FIGURE 1 | Overview of glucocorticoid receptor activity with classic glucocorticoids and selective GR modulators. (A) General action mechanism of the glucocorticoid
receptor (GR). Glucocorticoids (GCs) diffuse through the cellular membrane and bind GR. The latter dissociates from its chaperone complex and migrates to the
nucleus. There, it dimerizes and binds glucocorticoid response elements (GREs) to upregulate downstream target genes. Monomeric GR also undergoes
protein-protein interactions with DNA-bound pro-inflammatory transcription factors (TFs) to downregulate their activity, or it binds directly to the TF response elements
(TF-RE). (B) Distinct actions of classic GCs and selective GR modulators (SEGRMs). In contrast to classic GCs, SEGRMs are hypothesized to reduce GR’s capacity to
dimerize and therefore reduce GRE-mediated transcription. Interference with TF activity is driven via monomeric GR and therefore maintained with SEGRMs.
Frontiers in Endocrinology | www.frontiersin.org 2 September 2020 | Volume 11 | Article 559673
Van Moortel et al. Improved Glucocorticoid Receptor Ligands
Some of these side effects stem from direct binding of
homodimeric GR to pseudopalindromic glucocorticoid
reponse elements (GREs) in the promoter regions of genes
controlling key metabolic pathways (Figure 1). The resulting
GRE-driven upregulation of tyrosine aminotransferase (TAT),
glucose 6-phosphatase (G6P) and phosphoenolpyruvate
carboxykinase (PEPCK) for instance leads to hyperglycemia
(
6). The suppression of nuclea r factor (NF)-κB- and activator
protein (AP)-1 activity on the other hand, is typically explained
via protein-protein interactions with monomeric GR (called
tethering) (7). Despite the controversies on the actual underlying
mechanism (see further), the targeting of activities of pro-
inflammatory transcription factors undoubtedly contributes
substantially to the anti-inflammatory actions of GCs.
The discrepancy between monomer- and dimer-driven effects
of GR was first suggested in 1994 with the demonstration that GR
with a dimerization-disrupting mutation in the DBD (GRdim) is
still able to repress AP-1-driven genes, while no longer able to
induce GRE-mediated activation (
8). Four years later, Reichardt
et al. established that mice carrying this homozygous mutation
were viable and healthy, in contrast to GR full knock-out mice
(9), arguing for an equally viable mechanistic basis to separate
beneficial from undesired effects. This was the starting point of
the sea rch for so-called dissociative or selective GR modulators
(SEGRMs), GR ligands that can still repress inflammation via
monomer-driven NF-κB, and AP-1 inhibition, while no longer
inducing GRE-driven side effects (Figure 1B).
In the meantime, a few shades of gray have been added
to the original black-and-white monomer-dimer paradigm.
First of all, dimer-mediated gene activation also contributes
to the anti-inflammatory effects of GCs via the upregulation
of anti-inflammatory genes such as glucocorticoid-induced
leucine zipper (GILZ) and dual specificity phosphatase (DUSP1)
(
10). This helps explaining why GRdim mice show increased
sensitivity to acute inflammation such as septic shock (11).
Secondly, it was shown in cellulo that Dex still promotes
dimerization of the GRdim mutant (12). However, introducing
an extra point mutation in th e GR LBD almost completely
disrupted dimerization and abrogated GRE-driven activity, but
preser ved the inhibition of NF-κB activity. Thirdly, monomeric
GR was shown to bind directly to genomic NF-κB and AP-1
response elements, without the need for the transcription factor
itself (
13, 14). This finding challenges the original tethering
hypothesis but still supports the notion that suppression of NF-
κB and AP-1 activities does not require GR dimerization. Taken
together, given the bodies of evidence on a large contribution of
dimeric GR to particular side effects vs. the role of GR monomers
to support anti-inflammatory actions in a chronic setting, the
notion that compounds that favor signaling via monomeric GR
can hold a therapeutic benefit against persistent inflammation,
still stands.
The development of successful SEGRMs has proven to be a
long and extremely bumpy road. Many compounds that showed
promising initial results (listed in Table 1) never got past the
pre-clinical stage or failed la ter on in clinical trials. It is well-
known that the success rate for the development of any kind of
small molecule drug from bench to clinic is very low, typically
starting from 10,000 compounds to end up with one market-
approved drug (44, 45). In addition, we believe that in the case of
GR, multiple technical, and biological f actors have been reducing
the prospect to success even further. Fortunately, molecules
are still being developed, trying to meet the hope of many
patients wh o would benefit from GR modulators. For instance,
AZD7495 (asthma, NCT03622112) and AZD9567 (rheumatoid
arthritis, NCT03368235) are currently under evaluation in
clinical trials.
This perspective offers our opinion as molecular biologists
on the rationale why a continued search for SEGRMs still
makes sense and bears significant relevance. We offer our
view on a number of bottlenecks and pitfalls that might
have hampered research progress in the past and elaborate on
which new developments and insights could help overcome
these issues.
SEGRMs: THE UNMET MEDICAL NEED
The need for more selective GR ligands remains highly
relevant. Although more targeted therapies have successfully
been introduced, such as anti-tumor necrosis factor (TNF)
for arthritic disorders and inflammatory bowel diseases (IBD),
these therapies are not without limitations. For one, anti-
TNF t herapy has been associated with a 250% increase in the
occurrence of tuberculosis (
46). Furthermore, these therapies
have been reported to trigger multiple sclerosis (MS) and other
demyelinating conditions (
4750). This is in line with the
reported disease worsening in patients with pre-existing MS in
clinical trials for Lenercept and cA2, two types of anti-TNF
therapy (51, 52). Beside such side effects, monoclonal therapies
are generally very expensive, laying a huge burden on hea lth
care systems, which will only increase with aging populations
in western countries. Their price also makes them unaffordable
for most people in low income countries, which is particularly
a problem for asthma, for which 80% of disease-related deaths
occur in low to low-medium income countries (53).
GCs on the other hand are generally much cheaper and
are still the first-line treatment for asthma, multiple sclerosis,
and multiple myeloma among others (
5456). However, their
side effects are a well-known problem and not necessarily
limited to patients receiving oral or intravenous GCs. While
topical skin treatments, especially with t he newest generation
glucocorticoids, impose a very low risk for systemic side effects
(5759), topic al eye treatments, and inhaled GCs (IGCs) have
both been associated with adrenal suppression (60, 61). This can
lead to growth retardation in infants and children, who form a
large cohort of t h e asthma patient population. The long-term use
of high doses IGCs has also been associated with decreased bone
mineral density in both children and adults (6264). Although
the benefits of ocular and IGCs usually outweigh the risks,
patients would still benefit from GCs with lower risks for syst emic
side effe c ts.
Taken together, the need for more selective GCs reaches
further than systemic treatments and is also high for ocular and
inhalation therapies.
Frontiers in Endocrinology | www.frontiersin.org 3 September 2020 | Volume 11 | Article 559673
Van Moortel et al. Improved Glucocorticoid Receptor Ligands
TABLE 1 | Available pre-clinical data of SEGRMs.
Compound In vitro assays and in cellulo
overexpre ssion a ssays
In cellulo assays for endogenous
anti-inflammatory and/or side effect
targets
Inflammatory animal models Status and latest
progress
References
LGD-5552 Ligand-binding assays GR, AR, MR,
PR
MMTV-luciferase in CV-1 cells
(overexpressed GR)
E-selectin-luciferase in CV-1 and
HepG2 cells (overexpressed GR)
IL-6-luciferase in HepG2 cells
(overexpressed GR)
Cofactor binding assays
PEPCK and PDK4 mRNA in H4IIE cells
COX2 and APOCIII mRNA in H4IIE cells
POMC mRNA in ATT20 cells
Collagen-induced arthritis in
mice
Freund’s complete
adjuvant-induced arthritis in rats
Experimental autoimmune
encephalomyelitis in rats
Discontinued (preclinical) (
15, 16)
AL-438 Ligand-binding assays GR, PR
RSV-LTR-GRE-luciferase in CV-1 cells
(overexpressed GR)
TAT-luciferase in HepG2 cells
(overexpressed GR)
E-selectin-luciferase in HepG2 cells
(overexpressed GR)
Cofactor binding assays
Eosinophil counts in BAL
Human PBMC cell and rat splenocyte T-cell
proliferation assays
Osteocalcin protein in MG-63 cells
Aromatase activity in hDSF cells
IL-6 release in HSKF1501 cells
Carrageen-induced paw edema
in rats
Freund’s complete
adjuvant-induced arthritis in rats
Discontinued (preclinical) (
1719)
MK-5932 MMTV-luciferase in A549 cells
MMTV-luciferase in HeLa cells
TNFα-β-lactamase in U937 cells
TNFα, IFNγ, IL-1β, IL-6 secretion in human
whole blood
TNFα, IL-6 secretion in rat whole blood
Oxazolone-induced contact
dermatitis in rats
Discontinued (preclinical) (
20, 21)
GW870086 Functional selectivity MR, AR, PR, ER
on MMTV-luciferase in CV-1 cells
MMTV-LTR-luciferase in A549 and
MG-63 cells
E-selectin-κB-RE-alkaline
phosphatase in A549 cells
Lymphotoxin-β, COX-2, Cyp24a1, MAP-7,
GPR64, GILZ, DUSP1, MICAL2, FKBP5,
CDKN1C, RGS2, SGK mRNA in A549 cells
Delayed-type oxazolone-induced
contact hypersensitivity in mice
Ovalbumin-induced airway
inflammation in mice
Discontinued
Phase II for asthma: no
difference with placebo
(NCT00945932)
Phase II for atopic
dermatitis: weaker effects
than fluticasone propionate
standard (NCT01299610)
(
2224)
(Continued)
Frontiers in Endocrinology | www.frontiersin.org 4 September 2020 | Volume 11 | Article 559673
Van Moortel et al. Improved Glucocorticoid Receptor Ligands
TABLE 1 | Continued
Compound In vitro assays and in cellulo
overexpre ssion a ssays
In cellulo assays for endogenous
anti-inflammatory and/or side effect
targets
Inflammatory animal models Status and latest
progress
References
BI653048 Ligand-binding assays GR, PR
MMTV-luciferase in HeLa cells
IL-6 release in CCD-1112Sk cells
Osteocalcin levels in MG-63 cells
Human ERG potassium channel inhibition in
Hek293T cells
Canine low dose endotoxemia
model
Discontinued
Phase I: no improvement on
side effect profile compared
to prednisolone
(NCT02217631,
NCT02224105, NCT02217644)
(
2527)
Mapracorat Ligand-binding assays GR, PR, AR, MR
MMTV-luciferase in HeLa cells
Collagenase-luciferase in HeLa cells
κB-RE-luciferase in SV-40 transformed
hCEpiC cells
TPA-RE-luciferase in SV-40 transformed
hCEpiC cells
TAT activity in HepG2 cells
IL-12p40, IFNγ secretion in PBMC cells
Eotaxin-1 (+/– GR siRNA), 3, CCL5 (+/– GR
siRNA), G-CSF, IL-6, IL-8, MCP-1 release in
hConF cells
Eotaxin-3, CCL5 (+/– GR siRNA), CCL27,
ICAM-1 (+/– GR siRNA), IL-6, IL-8, MCP-1,
TNFα release in hCEpiC cells
IL-6, MCP-1 release and (p)p38, (p)JNK protein
in SV-40 transformed hCEpiC cells
IL-6, IL-8 release in hONA cells
IL-1β, ICAM-1 release in hREC cells
IL-6, IL-12p40, MCP-1 release in THP-1 cells
(p)JNK, (p)p65, (p)p38, IκBα levels in hCEpiC
cells
MYOC levels in mkTM cells
Migration, apoptosis, IL-8 release, annexin-1,
and CXCR4 expression in human eosinophils
IL-6, IL-8, CCL5, TNFα release in hMC-1 cells
GM-CSF, TNFα, PGE2 production and COX-2,
(p)p38, (p)MK2, DUSP1 protein in Raw 264.7
cells
IL-6, IL-8, MCP-1, PGE2 release, COX-2, RelB,
(p)IκBα protein, RelA and RelB DNA binding in
human keratinocytes
Croton oil-induced irritant
contact dermatitis in mice and
rats
Dinitrofluorobenzene
(DNFB)-induced allergic contact
dermatitis in mice and rats Dry
eye model in rabbits
Paracentesis model in rabbits
Ovalbumin-induced allergic
conjunctivitis in guinea pigs
Compound 48/80-induced
wheal and erythema skin
inflammation in beagles
Discontinued
Phase III for cataract
surgery, no results reported
(NCT01591655)
Phase I for psoriasis, no
results reported
(NCT03399526)
Phase I to assess corneal
endothelial cell changes, no
results
reported (NCT01736462)
(
2836)
(Continued)
Frontiers in Endocrinology | www.frontiersin.org 5 September 2020 | Volume 11 | Article 559673
Van Moortel et al. Improved Glucocorticoid Receptor Ligands
TABLE 1 | Continued
Compound In vitro assays and in cellulo
overexpre ssion a ssays
In cellulo assays for endogenous
anti-inflammatory and/or side effect
targets
Inflammatory animal models Status and latest
progress
References
(Fos)dagrocorat Gal4-RE-luciferase with
Gal4-DBD-LBD in Huh7 cells
Cofactor binding assays
IL-6 release in A549 cells
IFNγ in human whole blood assays
Human pre-adipocyte differentiation
FABP4 mRNA in adipocytes
TAT, PEPCK in human primary adipocytes
Osteocalcin levels in human
primary osteoblasts
Murine LPS-induced
endotoxemia model
Discontinued
Phase II for rheumatoid
arthritis: no improved
benefit-risk ratio compared
to
prednisone (NCT01393639)
(
37, 38)
AZD5423 Ligand-binding assays GR, MR, PR,
AR, ERα, ERβ
TPA-RE-β-galactosidase stable in
ChaGoK1 cells
TNFα-release in hPBMC cells Sephadex-induced airway
inflammation in rats
Discontinued
Phase II for asthma
(NCT01225549)
Phase II for
COPD (NCT01555099)
(
3941)
AZD7594 Ligand-binding assays GR, MR, PR,
AR, ERα, ERβ
TPA-RE-β-galactosidase stable in
ChaGoK1 cells
TNFα-release in hPBMC cells Sephadex-induced airway
inflammation in rats
Ongoing
Second phase II for asthma
completed 11/2019
(NCT03622112)
Phase I in adolescents
ongoing (NCT03976869)
(
39, 42)
AZD9567 Ligand-binding assays GR, MR, PR,
AR, ERα, ERβ
MMTV-β-galactosidase stable in
ChaGoK1 cells
TPA-RE-β-galactosidase stable in
ChaGoK1 cells
Cofactor binding assays
TAT in primary hepatocytes
Osteoprotegerine in human fetal osteoblasts
Streptococcal cell wall
reactivation arthritis model in rats
Ongoing
Phase II for rheumatoid
arthritis completed
11/2019 (NCT03368235)
(
43)
Information on clinical trials was retrieved from clinicaltrials.gov. A549, human lung epithelial carcinoma cell line; APOCIII, apolipoprotein C18; AR, androgen receptor; ATT20, mouse pituitary tumor cell line; BAL, bronchoalveolar
lavage; C C D-1112Sk, human foreskin fibroblast cell line; CCL (5), C-C motif chemokine (5); CDKN1C, cyclin-dependent kinase inhibitor 1C; hCEpiC, human corneal epithelial cells; ChaGoK1, human bronchogenic carcinoma cell line;
hConF, human conjunctival fibroblasts; COX-2, cyclo-oxygenase 2; CV-1, African green monkey kidney cell line; Cyp24a1, vitamin D (3) 24-hydroxylase; CXCR4, C-X-C chemokine receptor type 4; hDSF, human dermal skin fibroblasts;
ER, estrogen receptor; hERG, human ether-a-go-go potassium channel; FKBP5, 51 kDa FK506-binding protein; G(M)-CSF, granulocyte (macrophage) colony-stimulating factor; GPR-64, G-protein coupled receptor 64; H4IIE, rat
hepatocellular carcinoma cell line; Hek293T, human embryonic kidney cell line; HeLa, human cervical adenocarcinoma cell line; HepG2, human hepatocellular carcinoma cell line; HSKF1501, human foreskin fibroblast cell line; Huh7,
human hepatocellular carcinoma cell line; ICAM-1, intracellular adhesion molecule 1; IFNγ , interferon γ ; Iκ Bα, NF-kappa-B inhibitor α; IL(-12p40), interleukin (12 subunit p40); (p)JNK, (phospho-)c-Jun N-terminal kinase; κ B-RE, NF-κ B
response element; LTR, long terminal repeat; MAP-7, microtubule-associated protein 7; hMC-1, human mast cell line; MCP-1, monocyte chemotactic protein 1; MG-63, human osteosarcoma cell line; MICAL2, molecule interacting
with CasL protein 2; (p)MK-2, (phospho-)mitogen-activated protein kinase-activated protein kinase 2; MMTV, mouse mammary tumor virus; MR, mineralocorticoid receptor; MYOC, myocillin; hONA, human optic nerve astrocytes;
(p)p38, (phospho-)mitogen activated protein kinase p38; (p)p65, (phospho-)nuclear factor kappa B subunit p65; PBMC, peripheral blood mononuclear cells; PDK4, pyruvate dehydrogenase kinase 4; PGE2, prostaglandin E2; POMC,
pro-opiomelanocortin; PR, progesterone receptor; Raw264.7, mouse leukemia macrophage cell line; hREC, human retinal endothelial cells; RGS2, regulator of G-protein signaling 2; RSV, Rous sarcoma virus; SGK, serum/glucocorticoid
regulated kinase; THP-1, human leukemic monocyte cell line; mkTM, monkey trabecular meshwork cells; TPA-RE, 12-O-Tetradecanoylphorbol-13-acetate response element; U937, human histiocytic lymphoma cell line.
Frontiers in Endocrinology | www.frontiersin.org 6 September 2020 | Volume 11 | Article 559673
Van Moortel et al. Improved Glucocorticoid Receptor Ligands
BOTTLENECKS AND PITFALLS
OBSERVED IN THE PAST
Current tools for screening potential SEGRMs suffer from
shortcomings and do not always capture the complexity of
GR signaling. First of all, the lack of three-dimensional
structures of full-length GR highly restricts our knowledge of
GRs structure-activity relationship and decreases the predictive
power of molecular modeling and docking studies. Additionally,
all existing crystal structures of ligands in complex with
GRs LBD were obtained upon the introduction of one or
more mutations in this LBD. Although these mutations were
predicted not to influence the LBD structure, this can never
be claimed with absolute certainty. F602S for instance, one
of the most commonly used GR mutations allowing growth
of LBD crystals, causes chemical shift perturbations in LBD
nuclear magnetic resonance spectra compared to wild-type
LBD (
65). Furthermore, GR is allosterically regulated through
interactions with its corresponding response elements and
cofactors (
66), and more general also for other NR members,
conformational changes in one NR domain can allosterically
alter the conformation of another domain within the same NR
molecule (67). Thus, most probably the conformation of the LBD
studied in isolation is an incorrect reflection of this domain’s
conformation in the full-length protein.
Further, while high affinity and selectivity for GR can be
captured using in vitro ligand-binding assa ys, confirmations in
a cellular or in vivo context are sometimes lacking. This harbors
an inherent risk to miss out on off-target effects of the compound
in question. Therefore, the confirmation of GR dependency in
a cellular and an in vivo context is still an important valida tion
to make, for instance by testing compounds in wild-type vs. GR
knock-out models.
Table 1 provides an overview of the assays typically carried
out to characterize GR-mediated actions of a set of well-
known SEGRMs. To our opinion, a lack of predictive power
is one of the problems most difficult to solve, especially
when moving from simplified assays to more complex biology.
Direct GRE-driven activity, potentially leading to side effects,
is almost universally monitored via reporters driven by a
mouse mammary tumor virus (MMTV) promoter. Although
a fast and straightforward and thus defendable method for
initial compound characterization, a GRE-driven reporter assay
can be a poor predictor for regulation of endogenous GRE-
driven genes, as was also observed for MMTV (
15, 22).
The effects of GCs are highly gene-specific and GRE-driven
activity can differ depending on the sequence of the GRE
and the surrounding chromatin environment (6870). The use
of overexpressed GR should also be avoided in such assays,
as this may lead to compound potencies and efficacies t hat
are not necessarily representative for an endogenous context.
Additionally, not all side effects are dimer-driven and are
therefore not predict able v ia GRE-driven reporters. Mimicking
the right gene- and context-specificity of GR activity remains one
of the greatest challenges. Making a switch from reporters driven
by minimal recombinant promoters to more physiologically
relevant promoters could already offer some benefit. These
promoters would ideally belong to genes that are confirmed
mediators of underlying therapeutic - or side effects. Validation
on a well-representative set of relevant endogenous target genes is
even more important (see below, section Potential Solutions: The
Way Forward).
Cell- and tissue-specificity of GC actions is another variable
parameter. The MMTV-driven reporter for instance showed
stronger upregulation by GW870086 in bone osteosarcoma
cells compared to lung epithelial carcinoma cells (
22). It thus
remains essential to screen compounds in cell types that are the
best proxies for the underlying therapeutic and/or side effects
in vivo, for instance the use of hepatocy tes to study effects
on glucose and lipid metabolism, or th e use of osteoblast or
osteoclast cell lines for drugs that would be used in arthritis
patients. Further, although characterization of compound activity
in cellulo is essential, this will always be an oversimplification of
the situation in a living organism. Therefore, validation of an
improved therapeutic benefit depends on representative animal
models. While this is readily implemented for anti-inflammatory
effect scoring, concomitant testing of side effect parameters
(such as glucose tolerance, insulin tolerance, cortisol levels,
bone mineral density) presents a bottleneck, because a longer
treatment protocol may be needed to surpass the thresholds of
measurable results for these parameters or because of species
differences (see below) (
22, 43).
Lack of translatability from animal models to human patients
is yet another hurdle to overcome. Differences in ligand activity
between species can be an underlying cause, as observed for AL-
438 and MK-5932, which both had stronger anti-inflammatory
effects in rat vs. human blood (
17, 20). While it would be
recommended to perform initial cellular tests in human cells
as much as possible, in vivo interspecies differences remain a
hurdle in the entire field of drug discovery and are currently
difficult to overcome. Another concern is when animal models
used to study a particular disease insufficiently mimic the
pathology observed in humans. A careful design and set-up
of animal models remains key to study anti-inflammatory as
well as side effects. If a well-known side effect (marker) of
a classic GC in man is not observed in the animal model
used, this model will obviously have no predictive power on
(markers of) this particular side effect in patients and will
therefore be unsuited to evaluate the improved benefit-risk
ratio of SEGRMs over classic GCs. For instance, in a canine
model of low dose endotoxemia used to investigate the anti-
inflammatory and bone-sparing effects of BI653048, neither
BI653048 nor prednisolone treatment affected osteocalcin levels
(25). However, prednisolone does reduce bone mineral density
in dogs and decreases bone formation markers in humans after
1 day (71, 72). Indeed, in a phase I clinical trial, BI653048,
and prednisolone both caused decreased serum osteocalcin
levels (26). Studies with other SEGRMs also concluded that
osteocalcin levels in cellulo do not always reflect in vivo decreases
in bone density (18, 26, 27, 37, 38), casting doubts on the
value of osteocalcin as proxy for the in vivo reduction of bone
mineral density.
Lastly, notwithstanding the notion that dissociating GCs may
improve the benefit-risk ratio in chronic inflammatory disorders,
Frontiers in Endocrinology | www.frontiersin.org 7 September 2020 | Volume 11 | Article 559673
Van Moortel et al. Improved Glucocorticoid Receptor Ligands
a portion of the anti-inflammatory effects of GR does remain
dimer-driven (73). Hence, the likelihood decreases for truly
dissociating compounds to match the therapeutic efficacy of
the strongest classic GCs. Taking into account that some side
effects, such as osteoporosis, are at least partially mediated by
monomeric GR (
18), makes the quest to find a SEGRM that
scores better on multiple side effects even more challenging.
POTENTIAL SOLUTIONS: THE WAY
FORWARD
Although pre-clinical characterization of compounds will
never suffice to accurately predict their effects in patients,
particular improvements on current screenings could increase
the predictive power. First of all, reporter genes driven by
physiological promoters relevant for the clinical context of the
tested SEGRM should be preferred over artificial promoters. An
example could be the use of the G6P- or PEPCK-promoters in
liver cells to monitor hyperglycemia (
74, 75), or a Runt-related
transcription factor (Runx)2-driven promoter in osteoblasts
or Smad-driven promoters in osteoclasts as markers for GC-
induced osteoporosis (76, 77). A consistent and thorough
screening of endogenous targets in a relevant human cellular
context adds to importance. While monitoring GR activity in
every targeted pathway for every compound is impossible to
achieve, identification of reliable in cellulo biomarkers with a
higher predictive power for species-independent in vivo anti-
inflammatory and/or side effects would be a tremendous help.
This requires a full understanding of the molecular mechanisms
driving both anti-inflammatory and side effects in human tissues
as well as in animal models. This is, particularly for side
effects, not always the case. Continued efforts to unravel the
underlying molecular mechanisms driving particular GC side
effects are therefore crucial. However, some important side effect
mediators have already been identified and could be suitable
markers. Examples are muscle ring finger (MuRF)1, atrogin-
1, and Krüppel-like factor (KLF)15 in muscle atrophy (
78),
regulated in development and DNA damage response (REDD)1
in skin (and muscle) atrophy (79, 80), and G6P and PEPCK in
liver (
74, 75). In bone, the upregulation of cleaved caspase 3
and 9 or the reduction of bone morphogenetic protein (BMP)2
and Runx2 activity are important predictors for reduction in
osteoblast numbers (81, 82), while upregulation of receptor
activator of nucle ar factor-κB ligand (RANKL)-RANK signaling
and cathepsin K activity are important markers for increased
osteoclast differentiation and activity (respectively) (76, 83).
Reduction of publication bias toward “negative results and
joining forces between pharmaceutical companies and academic
groups should push the current boundaries and drive research
forward. At times, underlying reasons for discontinuation
of (pre-)clinical research remain enigmatic. As one concrete
example of many other examples that can be brought forward,
results from t hree completed phase III clinical trials on the
use of Mapracorat for post-operative treatment of cataract
surgery (NCT01230125, NCT01591161, and NCT01591655)
await publication, leaving fundamental scientists on the sideline
wondering why Mapracorat was never market approved. More
insights on where exactly discontinued SEGRMs failed, if those
reasons are on the scientific level, will encourage academic
labs with the right expertise to dig deeper into the underlying
causes, and create feedback-knowledge that may flow back to
industrial programs.
Even though fully dissociating SEGRMs might never reach the
therapeutic efficacy of the most potent classic GCs, they can still
offer relevant therapeutic benefit. Many inflammatory disorders
are characterized by a disease course that alternates between
periods of remission and exacerbation or relapse. SEGRMs might
not trigger the full-on anti-inflammatory casc ade that is required
to suppress an exacerbation, but might be ideal for maintenance
therapy. To maintain disease control, lower GC doses often
suffice. SEGRMs could match the anti-inflammatory efficacy of
the lower dose classic GCs while still showing a reduced side
effect burden. Combination of classic GCs with SEGRMs or
other therapeutic agents is another strategy to increase benefit-
risk ratios. Combination of Dex with CpdA was for instance
shown to increase anti-inflammatory effects while reducing
GRE-driven signaling in cellulo (84). Finally, the development
of compounds that do not bind the classic ligand-binding
pocket but instead target the dimerization interface might be an
interesting alternative strategy to disrupt GRE-driven signaling.
The intrinsically disordered nature of the GR NTD (85), has
so far prohibited resolving a crystal structure of full-length GR.
However, some smaller (however technically still challenging)
advances could already lead to important new insights. Crystal
structures of wild-type LBD in absence of stabilizing muta tions
would already give more confidence in the reliability of current
docking approaches. Secondly, crystal structures of the DBD-
hinge-LBD portion would not only lead to a better understanding
of the structure-activity relationship of GR, but might pose extra
advantages for molecular modeling or docking studies. Since
efficient GR dimerization seems to require both DBD and LBD
(
12), crystal structures of at least the DBD-hinge-LBD portion of
GR should improve predictions on those molecular entities that
are truly dimer-disrupting.
Another important emerging strategy to find more efficacious
GCs is to minimize exposure to non-inflamed tissues. IGCs can
for instance be optimized to undergo rapid elimination once
they enter the systemic circulation, a strategy that was applied
for the development of AZD7594 (
39). For systemic GCs, the
use of liposomal formulations is showing very promising results.
While these not only improve distribution to tissues that are
anatomically difficult to reach (8688), they can lower the side
effects of systemic GCs by maximizing concentrations at the
inflamed tissues while minimizing distribution to other tissues
(89, 90).
While it may be utopia to try and develop compounds that
alleviate all side effects, improved profiles for particular side
effects may be achievable. Skin thinning and ocular hypertension
are for instance among the most problematic side effects for
topical and ocular GC treatments, respectively (5). For systemic
treatments, liposomal formulations in combination with selective
improvement of particular side effects may be a viable way
forward. Liposomal SEGRMs that do not affect bone metabolism
Frontiers in Endocrinology | www.frontiersin.org 8 September 2020 | Volume 11 | Article 559673
Van Moortel et al. Improved Glucocorticoid Receptor Ligands
might for instance have an increased benefit-risk ratio over classic
GCs for the treatment of arthritic disorders.
CONCLUSION
While there still seems a long road ahead toward SEGRMs
with a real improved benefit-risk ratio, there is light at the
end of the tunnel. The pipeline of SEGRM compounds under
clinical evaluation is not empty and new insights from ongoing
(or future) research is expected to lead to optimized screening
tools with maximized predictive power. Additionally, strategies
to limit exposure to off-targets tissues, such as liposomal
formulations for systemic treatments, show promising results
(
8690). Combination of these approaches with the identification
of reliable markers to predict on-target side effects, (e.g., ocular
hypertension in ocular tre atment , osteoporosis in rheumatoid
arthritis, skin thinning in topic applications) may be an effective
and achievable leap forward.
DATA AVAILABILITY STATEMENT
The original contributions presented in the study are included
in the article/supplementary material, further inquiries can be
directed to t h e corresponding author/s.
AUTHOR CONTRIBUTIONS
LVM wrote the manuscript with contributions from KG and
KDB. LVM made the artwork with contributions from KG and
KDB. All authors approved the final version.
FUNDING
LVM was supported by a strategic basic research fellowship
of the Research Foundation-Flanders (FWO-Vlaanderen), grant
number 1S14720N.
REFERENCES
1. Tan C, Wahli W. A trilogy of glucocorticoid receptor actions. Proc Natl Acad
Sci USA. (2016) 113:1115–7. doi: 10.1073/pnas.1524215113
2. Oakley R, Cidlowski J. The biology of the glucocorticoid receptor: new
signaling mechanisms in health and disease. J Allergy Clin Immunol. (2013)
132:1033–44. doi: 10.1016/j.jaci.2013.09.007
3. Ramamoorthy S, Cidlowski JA. Corticosteroids. mechanisms of action
in health and disease. Rheum Dis Clin N Am. (2016) 42:15–31.
doi: 10.1016/j.rdc.2015.08.002
4. Caratti G, Matthews L, Poolman T, Kershaw S, Baxter M, Ray D.
Glucocorticoid receptor function in health and disease. Clin Endocrinol.
(2015) 83:441–8. doi: 10.1111/cen.12728
5. Schäcke H, Döcke W-D, Asadullah K. Mechanisms involved in the
side effects of glucocorticoids. Pharmacol Ther. (2002) 96:23–43.
doi: 10.1016/S0163-7258(02)00297-8
6. Ratman D, Vanden Berghe W, Dejager L, Libert C, Tavernier J, Beck I, et al.
How glucocorticoid receptors modulate the activity of other transcription
factors: a scope beyond tethering. Mol Cell Endocrinol. (2013) 380:41–54.
doi: 10.1016/j.mce.2012.12.014
7. De Bosscher K, Haegeman G. Minireview: latest perspectives on
antiinflammatory actions of g lucocorticoids. Mol Endocrinol. (2009)
23:281–91. doi: 10.1210/me.2008-0283
8. Heck S, Kullmann M, Gast A, Ponta H, Rahmsdorf HJ, Herrlich P, et al.
A distinct modulating domain in glucocorticoid receptor monomers in the
repression of activity of the transcription factor AP-1. EMBO J. (1994)
13:4087–95. doi: 10.1002/j.1460-2075.1994.tb06726.x
9. Reichardt HM, Kaestner KH, Tuckermann J, Kretz O, Wessely O, Bock R, et al.
DNA binding of the glucocorticoid re ceptor is not essential for survival. Cell.
(1998) 93:531–41. doi: 10.1016/S0092-8674 (00)8 11 8 3- 6
10. Clark A, Belvisi M. Maps and legends: the quest for dissociated
ligands of the glucocorticoid receptor. Pharmacol Ther. (2012) 134:54–67.
doi: 10.1016/j.pharmthera.2011.12.004
11. Kleiman A, Hübner S, Parkitna JMR, Neumann A, Hofer S, Weigand MA,
et al. Glucocorticoid receptor dimerization is required for survival in septic
shock via suppression of interleukin-1 in macrophages. FASEB J. (2012)
26:722–9. doi: 10.1096/fj.11-192112
12. Presman DM, Ogara MF, Stortz M, Alvarez LD, Pooley JR, S chiltz RL,
et al. Live cell imaging unveils multiple domain requirements for in vivo
dimerization of the glucocorticoid receptor. PLoS Biol. (2014) 12:e1001813.
doi: 10.1371/journal.pbio.1001813
13. Weikum ER, de Vera IMS, Nwachukwu JC, Hudson WH, Nettles KW, Kojetin
DJ, et al. Tethering not required : the glucocorticoid receptor binds directly to
activator protein-1 recognition motifs to repress inflammatory genes. Nucleic
Acids Res. (2017) 45:8596–608. doi: 10.1093/nar/gkx509
14. Hudson WH, de Vera IMS, Nwachukwu JC, Weikum ER, Herbst AG,
Yang Q, et al. Cryptic glucocorticoid receptor-binding sites pervade
genomic NF-κB response elements. Nat Commun. (2018) 9:1337.
doi: 10.1038/s41467-018-03780-1
15. Miner JN, Ardecky B, Benbatoul K, Griffiths K, Larson CJ, Mais DE, et al.
Antiinflammatory glucocorticoid receptor ligand with reduced side effects
exhibits an altered protein-protein interaction profile. Proc Natl Acad Sci USA.
(2007) 104:19244–9. doi: 10.1073/pnas.0705517104
16. López F, Ardecky R, Bebo B, Benbatoul K, de Grandpre L, Liu S, et al. LGD-
5552, an antiinflammatory glucocorticoid receptor ligand with reduced side
effects, in vivo. Endocrinology. (2008) 149:2080–8 9. doi: 10.1210/en.2007-1353
17. Kym PR, Kort ME, Coghlan MJ, Moore JL, Tang R, Ratajczyk JD,
et al. Nonsteroidal selective glucocorticoid modulators: the effect of C-10
substitution on receptor selectivity and functional potency of 5-allyl-2,5-
dihydro-2,2,4-trimethyl-lh-[1]benzopyrano[3,4-f]quinolines. J Med Chem.
(2003) 46:1016–30. doi: 10.1021/jm020335m
18. Coghlan M, Jacobson P, L ane B, Nakane M, Lin CW, Elmore S, et al. A novel
antiinflammatory maintains glucocorticoid efficacy with reduced side effects.
Mol Endocrinol. (2003) 17:860–9. doi: 10.1210/me.2002-0355
19. Elmore SW, Miner JN, Jacobson PB, Wilcox DM, Lane BC, Coghlan MJ,
et al. Nonsteroidal selective glucocorticoid modulators: the effect of C-5
alkyl substitution on the transcriptional activation/repression profile of 2,5-
dihydro-10-methoxy-2,2,4-trimethyl-1H-[1]benzopyrano[3,4-f]quinolines. J
Med Chem. (2001) 44:4481–91. doi: 10.1021/jm010367u
20. Brandish PE, Anderson K, Baltus GA, Bai C, Bungard CJ, Bunting P, et al.
The preclinical efficacy, selectivity and pharmacologic profile of MK-5932, an
insulin-sparing selective glucocorticoid receptor modulator. Eur J Pharmacol.
(2014) 724:102–11. doi: 10.1016/j.ejphar.2013.12.031
21. Bungard CJ, Hartman GD, Manikowski JJ, Perkins JJ, Bai C, Brandish PE, et al.
Discovery of selective glucocorticoid receptor modulator MK-5932. Bioorg
Med Chem. (2011) 19:7374–86. doi: 10.1016/j.bmc.2011.10.054
22. Uings IJ, Needham D, Matthews J, Haase M, Austin R, Angell D,
et al. Discovery of GW870086: a potent anti-inflammatory steroid with
a unique pharmacological profile. Br J Pharmacol. (2013) 169:1389–40 3.
doi: 10.1111/bph.12232
23. Bareille P, Hardes K, Donald AC. Efficacy and safety of once-daily
GW870086 a novel selective glucocorticoid in mild-moderate asthmatics: a
randomised, two-way crossover, controlled clinical trial. J Asthma. (2013)
50:1077–82. doi: 10.3109/02770903.2013.837480
24. Dölle S, Hielscher N, Bareille PJ, Hardes K, Robertson J, Worm M. Clinical
efficacy and tolerability of a novel selective corticosteroid in atopic dermatitis
Frontiers in Endocrinology | www.frontiersin.org 9 September 2020 | Volume 11 | Article 559673
Van Moortel et al. Improved Glucocorticoid Receptor Ligands
- two randomised controlled trials. Skin Pharmacol Physiol. (2015) 28:159–66.
doi: 10.1159/000367696
25. Bartko J, Derhaschnig U, Neels T, Nabozny GH, Harcken C, Leuschner J,
et al. Selective glucocorticoid receptor modulation inhibits cytokine responses
in a canine model of mild endotoxemia. Pharmacol Res. (2017) 12 5 :21 5–2 3 .
doi: 10.1016/j.phrs.2017.09.006
26. Harcken C, Scholl P, Nabozny G, Thomson D, Bianchi D. Clinical profile
of the functionally selective glucocorticoid receptor agonist BI 653048
in healthy male subjects. Expert Opin Investig Drugs. (2019) 28:489–96.
doi: 10.1080/13543784.2019.1599859
27. Harcken C, Riether D, Liu P, Razavi H, Patel U, Lee T, et al. Optimization
of drug-like properties of nonsteroidal glucocorticoid mimetics and
identification of a clinical candidate. ACS Med Chem Lett. (201 4) 5:1318–23.
doi: 10.1021/ml500387y
28. Pfeffer BA, DeWitt CA, Salvador-Silva M, Cavet ME, López FJ, Ward
KW. Reduced myocilin expression in cultured monkey trabecular
meshwork cells induced by a selective glucocorticoid receptor agonist:
comparison with steroids. Invest Ophthalmol Vis Sci. (2010) 51:437–46.
doi: 10.1167/iovs.09-4202
29. Shafiee A, Bucolo C, Budzynski E, Ward KW, López FJ. In vivo ocular efficacy
profile of mapracorat, a novel selective glucocorticoid receptor agonist, in
rabbit models of ocular disease. Invest Ophthalmol Vis Sci. (2011) 52:1422–30.
doi: 10.1167/iovs.10-5598
30. Schäcke H, Zollner T, Döcke W-D, Rehwinkel H, Jaroch S, Skuballa W,
et al. Characterization of ZK 245186, a novel, selective glucocorticoid
receptor agonist for the topical treatment of inflammatory skin diseases. Br
J Pharmacol. (2009) 158:1088–1103. doi: 10 .11 11 /j .14 76 -5 38 1 .200 9 .00 23 8.x
31. Zhang J-Z, Cavet ME, vanDerMeid KR, Salvador-Silva M, López FJ, Ward
KW. BOL-303242-X, a novel selective glucocorticoid receptor agonist,
with full anti-inflammatory properties in human ocular cells. Mol Vis.
(2009) 15:2606–16.
32. Baiula M, Spartà A, Bedini A, Carbonari G, Bucolo C, Ward KW, et al.
Eosinophil as a cellular target of the ocular anti-allergic action of mapracorat,
a novel selective glucocorticoid receptor agonist. Mol Vis. (2011) 17:3208–23.
33. Baiula M, Bedini A, Baldi J, Cavet ME, Govoni P, Spampinato S . Mapracorat,
a selective g lucocorticoid receptor agonist, causes apoptosis of eosinophils
infiltrating the conjunctiva in late-phase experimental ocular allergy. Drug Des
Devel Ther. (2014) 8:745–5 7 . doi: 10.2147/DDDT.S62659
34. Cavet ME, Harrington KL, Ward KW, Zhang J-Z. Mapracorat, a novel
selective glucocorticoid receptor agonist, inhibits hyperosmolar-induced
cytokine release and MAPK pathways in human corneal epithelial cells. Mol
Vis. (2010) 16:1791–800.
35. Spinelli SL, Xi X, McMillan DH, Woeller CF, Richardson ME, Cavet ME, et al.
Mapracorat, a selective glucocorticoid receptor agonist, upregulates RelB, an
anti-inflammatory nuclear factor-kappaB protein, in human ocular cells. Exp
Eye Res. (2014) 127:290–8. doi: 10.1016/j.exer.2014.07.013
36. Vollmer TR, Stockhausen A, Zhang J-Z. Anti-inflammatory effects of
mapracorat, a novel selective glucocorticoid receptor agonist, is partially
mediated by MAP kinase phosphatase-1 (MKP-1). J Biol Chem. (2012)
287:35212–21. doi: 10.1074/jbc.M112.40067 1
37. Hu X, Du S, Tunca C, Braden T, Long K, Lee J, et al. The antagonists but not
partial agonists of glucocorticoid re ceptor ligands show substantial side effect
dissociation. Endocrinology. (2011) 152:3123–3 4 . doi: 10.1210/en.2010-1447
38. Buttgereit F, Strand V, Lee EB, Simon-Campos A, McCabe D, Genet A, et al.
Fosdagrocorat (PF-04171327) versus prednisone or placebo in rheumatoid
arthritis: a randomised, double-blind, multicentre, phase IIb study. RMD
Open. (2019) 5:e000889. doi: 10 .11 36 /rmdopen-2 01 8- 00 0 88 9
39. Hemmerling M, Nilsson S. Selective Nonsteroidal Glucocorticoid Receptor
Modulators for the Inhaled Treatment of Pulmonary Diseases | Journal of
Medicinal Chemistry (2017). Available online at: https://pubs.acs.org/doi/full/
10.1021/acs.jmedchem.7b01215 (accessed April 2, 2020).
40. Kuna P, Aurivillius M, Jorup C, Prothon S, Taib Z, Edsbäcker S.
Efficacy and tolerability of an inhaled selective glucocorticoid receptor
modulator AZD5423 in chronic obstructive pulmonary disease patients:
Phase II study results. Basic Clin Pharmacol Toxicol. (2017) 121:279–89.
doi: 10.1111/bcpt.12768
41. Gauvreau GM, Boulet L-P, Leigh R, Cockcroft DW, Killian KJ, Davis BE,
et al. A nonsteroidal glucocorticoid receptor agonist inhibits allergen-induced
late asthmatic responses. Am J Respir Crit Care Med. (2014) 191:161–7.
doi: 10.1164/rccm.201404-0623OC
42. Brown MN, Fuhr R, Beier J, Su H-L, Chen Y, Forsman H, et al. Efficacy
and safety of AZD7594, an inhaled non-steroidal selective glucocorticoid
receptor modulator, in patients with asthma: a phase 2a randomized,
double blind, placebo-controlled crossover trial. Respir Res. (2019) 20:37.
doi: 10.1186/s12931-019-1000-7
43. Ripa L, Edman K, Dearman M, Edenro G, Hendrickx R, Ullah V, et al.
Discovery of a novel oral glucocorticoid receptor modulator (AZD9567)
with improved side effect profile. J Med Chem. (2018) 61:1785–99.
doi: 10.1021/acs.jmedchem.7b01690
44. Brake K, Gumireddy A, Tiwari A, Chauhan H, Kumari D. In vivo studies for
drug development via oral delivery: challenges, animal models and techniques.
Pharm Anal Acta. (2017) 8:1–11. doi: 10.4172/2153-2435.1000560
45. Takebe T, Imai R, Ono S. The current status of drug discovery and
development as originated in United States academia: the influence of
industrial and academic collaboration on drug discovery and development.
Clin Transl Sci. (2018) 11:597–606. doi: 10.11 1 1/cts.1 25 77
46. Minozzi S, Bonovas S, Lytras T, Pecoraro V, González-Lorenzo M,
Bastiampillai AJ, et al. Risk of infections using anti-TNF agents in
rheumatoid arthritis, psoriatic arthritis, and ankylosing spondylitis: a
systematic review and meta-analysis. Expert Opin Drug Saf. (2016) 15:11–34.
doi: 10.1080/14740338.2016.1240783
47. Kemanetzoglou E, Andreadou E. CNS demyelination with TNF-α
blockers. Curr Neurol Neurosci Rep. (2017) 17:36. doi: 10 .10 07 /s 11 91 0- 0 17 -
0742-1
48. Dreyer L, Magyari M, Laursen B, Cordtz R, Sellebjerg F, Locht H.
Risk of multiple sclerosis during tumour necrosis factor inhibitor
treatment for arthritis: a population-based study from DANBIO and
the Danish multiple sclerosis registry. Ann Rheum Dis. (2016) 75:785–6.
doi: 10.1136/annrheumdis-2015-208490
49. Engel S, Luessi F, Mueller A, Schopf RE, Zipp F, Bittner S. PPMS
onset upon adalimumab treatment extends the spectrum of anti-TNF-α
therapy-associated demyelinating disorders. Ther Adv Neurol Disord. (2020)
13:1756286419895155. doi: 10.1177/1756286419895155
50. Honda Y, Otsuka A, Egawa G, Inoue Y, Kuzuya A, Takahashi R, et al. Multiple
neurological abnormalities, including pontine hemorrhage, multiple sclerosis
and aseptic meningitis, during anti-TNF-α therapy in psoriatic arthritis. Eur J
Dermatol. (2015) 25:487–8. doi: 10.1684/ejd.2015.2558
51. The Lenercept Multiple Sclerosis Study Group and The University of British
Columbia MS/MRI Analysis Group. TNF neutralization in MS: results
of a randomized, placebo-controlled multicenter study. Neurology. (2011)
77:1382. doi: 10.1212/01.wnl.0000406608.85830.82
52. van Oosten BW, Barkhof F, Truyen L, Boringa JB, Bertelsmann FW, von
Blomberg BME, et al. Increased MRI activity and immune activation
in two multiple sclerosis patients treated with the monoclonal anti-
tumor necrosis factor antibody cA2. Neurology. (1996) 47:1531–4.
doi: 10.1212/WNL.47.6.1531
53. WHO. Global Surveillance, Prevention and Control of Chronic Respiratory
Diseases (2007). Available online at: https://www.who.int/respiratory/
publications/global_surveillance/en/ (accessed April 23, 2020).
54. Nazareth T, Datar M, Yu T-C. Treatment effectiveness for resolution of
multiple sclerosis relapse in a US health plan population. Neurol Ther. (2019)
8:383–95. doi: 10.1007/s40120-019-00156-5
55. Burwick N, Sharma S. Glucocorticoids in multiple myeloma: past, present, and
future. Ann Hematol. (2019) 98:19–28. doi: 10.1007/s00 2 77 -0 18 - 34 65 - 8
56. Global Initiative for Asthma. Global Strategy for Asthma Management and
Prevention. Global Initiative for Asthma - GINA. (2019). Available online at:
https://ginasthma.org/gina-reports/ (accessed April 3, 2020).
57. Mooney E, Rademaker M, Dailey R, Daniel BS, Drummond C, Fischer
G, et al. Adverse effects of topical corticosteroids in paediatric eczema:
Australasian consensus statement. Australas J Dermatol. (2015) 56:241–51.
doi: 10.1111/ajd.12313
58. Daniel BS, Orchard D. Ocular side–effects of topical corticosteroids: what
a dermatologist needs to know. Australas J Dermatol. (2015) 56:164–9.
doi: 10.1111/ajd.12292
59. Torrelo A. Methylprednisolone aceponate for atopic dermatitis. Int J
Dermatol. (2017) 56:691–7. doi: 10.1111/ijd.13485
Frontiers in Endocrinology | www.frontiersin.org 10 September 2020 | Volume 11 | Article 559673
Van Moortel et al. Improved Glucocorticoid Receptor Ligands
60. Bangsgaard R, Main KM, Boberg-Ans G, la Cour M, Forman JL,
Haargaard B, et al. Adrenal suppression in infants treated with
topical ocular glucocorticoids. Ophthalmology. (2018) 12 5:16 3 8– 43 .
doi: 10.1016/j.ophtha.2018.04.035
61. Hawcutt DB, Francis B, Carr DF, Jorgensen AL, Yin P, Wallin N,
et al. Susceptibility to corticosteroid-induced adrenal suppression: a
genome-wide association study. Lancet Respir Med. (2018) 6:442–50.
doi: 10.1016/S2213-2600(18)30058-4
62. Gonzalez AV, Coulombe J, Ernst P, Suissa S. Long-term use of inhaled
corticosteroids in COPD and the risk of fracture. Chest. (2018) 153:321–8.
doi: 10.1016/j.chest.2017.07.002
63. Skoner DP. Inhaled corticosteroids: effects on growth and bone health. Ann
Allergy Asthma Immunol. (2016) 11 7:5 95 –6 00 . doi: 10.1016/j .anai.2016.07.0 43
64. Wolfgram PM, Allen DB. Effects of inhaled corticosteroids on growth,
bone metabolism, and adrenal function. Adv Pediatr. (2017) 64:331–45.
doi: 10.1016/j.yapd.2017.03.006
65. Köhler C, Carlström G, Tångefjord S, Papavoine T, L epistö M, Edman
K, et al. Backbone 1H, 13C, and 15N resonance assignments of the
ligand binding domain of the human wildtype glucocorticoid receptor
and the F602S mutant variant. Biomol NMR Assign. (2 01 8 ) 12:263–8.
doi: 10.1007/s12104-018-9820-9
66. Weikum ER, Knuesel MT, Ortlund EA, Yamamoto KR. Glucocorticoid
receptor control of transcription: precision and plasticity via allostery. Nat Rev
Mol Cell Biol. (2017) 18:159–74. doi: 10.1038/nrm.2016.152
67. Ricci CG, Silveira RL, Rivalta I, Batista VS, Skaf MS. Allosteric pathways
in the PPARγ-RXRα nuclear receptor complex. Sci Rep. (2016) 6:19940.
doi: 10.1038/srep19940
68. Meijsing S, Pufall M, So A, Bates D, Chen L, Yamamoto K. DNA binding site
sequence directs glucocorticoid receptor structure and activity. Science. (2009)
324:407–10. doi: 10.1126/science.1164265
69. Telorac J, Prykhozhij S, Schöne S, Meierhofer D, Sauer S, Thomas-Chollier
M, et al. Identification and characterization of DNA sequences that prevent
glucocorticoid receptor binding to nearby response elements. Nucleic Acids
Res. (2016) 44:6142–56. doi: 10.1093/nar/gkw203
70. Schöne S, Jurk M, Helabad M, Dror I, Lebars I, Kieffer B, et al. Sequences
flanking the core-binding site modulate glucocorticoid receptor structure and
activity. Nat Commun. (2016) 7:12621. doi: 10.1038/ncomms12621
71. Kauh E, Mixson L, Malice M-P, Mesens S, Ramael S, Burke J, et al. Prednisone
affects inflammation, glucose tolerance, and bone turnover within hours
of treatment in healthy individuals. Eur J Endocrinol. (2012) 166:459–6 7.
doi: 10.1530/EJE-11-0751
72. Costa LAVS, Lopes BF, Lanis AB, Oliveira DCD, Giannotti JG, Costa
FS. Bone demineralization in the lumbar spine of dogs submitted
to prednisone therapy. J Vet Pharmacol Ther. (2010) 33:583–6.
doi: 10.1111/j.1365-2885.2010.01174.x
73. Desmet S, De Bosscher K. Glucocorticoid receptors : finding the middle
ground. J Clin Invest. (2017) 127:1136–45. doi: 10.1172/JCI88886
74. Imai E, Stromstedt PE, Quinn PG, Carlstedt-Duke J, Gust afsson JA, Granner
DK. Characterization of a complex glucocorticoid response u nit in t he
phosphoenolpyruvate carboxykinase gene. Mol Cell Biol. (1990) 10:4712–9.
doi: 10.1128/MCB.10.9.4712
75. Schmoll D, Allan BB, Burchell A. Cloning and sequencing of the 5
region of
the human glucose-6-phosphatase gene: transcriptional regulation by cAMP,
insulin and glucocorticoids in H4IIE hepatoma cells. FEBS Lett. (1996)
383:63–6. doi: 10.1016/0014-5793(96)00224-4
76. Fennen M, Pap T, Dankbar B. Smad-dependent mechanisms of
inflammatory bone destruction. Arthritis Res Ther. (2016) 18:279.
doi: 10.1186/s13075-016-1187-7
77. Morimoto E, Li M, Khalid AB, Krum SA, Chimge NO, Frenkel B.
Glucocorticoids Hijack Runx2 to stimulate Wif1 for suppression of
osteoblast g rowth and differentiation. J Cell Physiol. (2017) 232:145–53.
doi: 10.1002/jcp.25399
78. Shimizu N, Yoshikawa N, Ito N, Maruyama T, Suzuki Y, Takeda SI, et al.
Crosstalk between glucocorticoid receptor and nutritional sensor mTOR
in skeletal muscle. Cell Metab. (2011) 13:170–82. doi: 10.1016/j.cmet.2011.
01.001
79. Baida G, Bhalla P, Kirsanov K, Lesovaya E, Yakubovskaya M, Yuen K,
et al. REDD1 functions at the crossroads between the therapeutic and
adverse effects of topical glucocorticoids. EMBO Mol Med. (2015) 7:42–58.
doi: 10.15252/emmm.201404601
80. Britto FA, Begue G, Rossano B, Docquier A, Vernus B, Sar C, et al.
REDD1 deletion prevents dexamethasone-induced skeletal muscle atrophy.
AJP Endocrinol Metab. (2014) 307:E983–93. doi: 10.1152/ajpendo.00234.2014
81. Lin H, Gao X, Chen G, Sun J, Chu J, Jing K, et al. Indole-3-carbinol as
inhibitors of glucocorticoid-induced apoptosis in osteoblastic cells through
blocking ROS-mediated Nrf2 pathway. Biochem Biophys Res Commun. (2015)
460:422–7. doi: 10.1016/j.bbrc.2015.03.049
82. Liu K, Jing Y, Zhang W, Fu X, Zhao H, Zhou X, et al. Silencing miR-
106b accelerates osteogenesis of mesenchymal stem cells and rescues against
glucocorticoid-induced osteoporosis by targeting BMP2. Bone. (2017) 97 :13 0–
8. doi: 10.1016/j.bone.2017.01.014
83. Lin NY, Chen CW, Kagwiria R, Liang R, Beyer C, Distler A, et al.
Inactivation of autophagy ameliorates glucocorticoid-induced and
ovariectomy-induced bone loss. Ann Rheum Dis. (2015) 75:1203–10.
doi: 10.1136/annrheumdis-2015-207240
84. Desmet SJ, Bougarne N, Van Moortel L, de Cauwer L, Thommis J, Vuylsteke
M, et al. Compound A influences gene regulation of the Dexamethasone-
activated glucocorticoid receptor by alternative cofactor recruitment. Sci Rep.
(2017) 7:e8063. doi: 10.1038/s41598-017- 07 9 41 -y
85. Khan SH, McLaughlin WA, Kumar R. Site-specific phosphorylation
regulates the structure and function of an intrinsically disordered
domain of the glucocorticoid receptor. Sci Rep. (2017) 7:15440.
doi: 10.1038/s41598-017-15549-5
86. Mamelle E, Kechai NE, Granger B, Sterkers O, Bochot A, Agnely F, et al.
Effect of a liposomal hyaluronic acid gel loaded wit h dexamethasone in a
guinea pig model after manual or motorized cochlear implantation. Eur Arch
Otorhinolaryngol. (2017) 274:729–36. doi: 10.1007/s00405-016-4331-8
87. Altamirano-Vallejo JC, Navarro-Partida J, Gonzalez-De la Rosa A, Hsiao
JH, Olguín-Gutierrez JS, Gonzalez-Villegas AC, et al. Characterization
and pharmacokinetics of triamcinolone acetonide-loaded liposomes topical
formulations for vitreoretinal drug delivery. J Ocul Pharmacol Ther. (2018)
34:416–25. doi: 10.1089/jop.2017.0099
88. Deshantri AK, Fens MH, Ruiter RWJ, Metselaar JM, Storm G, van Bloois
L, et al. Liposomal dexamethasone inhibits tumor g rowth in an advanced
human-mouse hybrid model of multiple myeloma. J Controlled Release.
(2019) 296:232–40. doi: 10.1016/j.j conrel.201 9.01 .02 8
89. Meka RR, Venkatesha SH, Acharya B, Moudgil KD. Peptide-targeted
liposomal delivery of dexamethasone for arthritis therapy. Nanomed. (2019)
14:1455–69. doi: 10.2217/nnm-2018-0501
90. Moallem E, Koren E, Ulmansky R, Pizov G, Barlev M, Barenholz Y, et al.
A liposomal steroid nano-drug for treating systemic lupus erythematosus.
Lupus. (2016) 25:1209–16. doi: 10 .117 7 /09 6 12 03 3 16 63 6 46 8
Conflict of Interest: The authors declare that the research was conducted in the
absence of any commercial or financial relationships that could be construed as a
potential conflict of interest.
Copyright © 2020 Van Moortel, Gevaert and De Bosscher. This is an open-access
article distributed under the terms of the Creative Commons Attribution License (CC
BY). The use, distribution or reproduction in other forums is permitted, provided
the original author(s) and the copyright owner(s) are credited and that the original
publication in this journal is cited, in accordance with accepted academic practice.
No use, distribution or reproduction is permitted which does not comply with these
terms.
Frontiers in Endocrinology | www.frontiersin.org 11 September 2020 | Volume 11 | Article 559673