ArticlePDF AvailableLiterature Review

JAK/STAT pathway targeting in primary Sjögren syndrome

Authors:

Abstract and Figures

Primary Sjögren's syndrome (pSS) is an autoimmune systemic disease mainly affecting exocrine glands and resulting in disabling symptoms, as dry eye and dry mouth. Mechanisms underlying pSS pathogenesis are intricate, involving multiplanar and, at the same time, interlinked levels, e.g., genetic predisposition, epigenetic modifications and the dysregulation of both immune system and glandular-resident cellular pathways, mainly salivary gland epithelial cells. Unravelling the biological and molecular complexity of pSS is still a great challenge but much progress has been made in recent years in basic and translational research field, allowing the identification of potential novel targets for therapy development. Despite such promising novelties, however, none therapy has been specifically approved for pSS treatment until now. In recent years, growing evidence has supported the modulation of Janus kinases (JAK) - signal transducers and activators of transcription (STAT) pathways as treatment strategy immune mediated diseases. JAK-STAT pathway plays a crucial role in autoimmunity and systemic inflammation, being involved in signal pathways of many cytokines. This review aims to report the state-of-the-art about the role of JAK-STAT pathway in pSS, with particular focus on available research and clinical data regarding the use of JAK inhibitors in pSS.
This content is subject to copyright. Terms and conditions apply.
Review • DOI: 10.2478/rir-2022-0017 • 3(3) • 2022 • 95–102
RHEUMATOLOGY AND IMMUNOLOGY RESEARCH
95
Introduction
Primary Sjögren’s syndrome (pSS) is an autoimmune systemic
disease which mainly a󰀨ects exocrine glands and causes dis-
abling symptoms, such as dry eye and dry mouth.[1,2] Besides
sicca, fatigue and pain are the other most common symptoms
in pSS patients, together with a wide range of possible addi-
tional and highly heterogeneous clinical extra-glandular mani-
festations, potentially involving any organ.[1,2] Furthermore,
pSS patients are at higher risk than general population to
develop lymphoma, mainly a non-Hodgkin B cell lymphoma of
mucosa-associated lymphoid tissue (MALT) type.[3,4] Females
are most a󰀨ected by pSS than males, with a ratio 9:1, but an
increased risk of lymphoma development has been recent-
ly reported for the latter.[5] The histological hallmark of pSS,
i.e., the focal lymphocytic sialadenitis, observed in speci-
mens obtained through minor salivary gland (MSG) biopsy,
still represents, together with anti-SSA (Sjögren’s-syndrome-
related antigen A) autoantibodies, a crucial pillar for pSS
JAK/STAT pathway targeting in primary Sjögren
syndrome
Saviana Gandolfo1, Francesco Ciccia2,*
1Rheumatology Unit, Department of Internal Medicine, San Giovanni Bosco Hospital, Naples, Italy
2Department of Precision Medicine, Università della Campania Luigi Vanvitelli, Naples, Italy
Received May 18, 2022 accepted June 25, 2022
Primary Sjögren’s syndrome (pSS) is an autoimmune systemic disease mainly a󰀨ecting exocrine glands and resulting
in disabling symptoms, as dry eye and dry mouth. Mechanisms underlying pSS pathogenesis are intricate, involving
multiplanar and, at the same time, interlinked levels, e.g., genetic predisposition, epigenetic modications and the
dysregulation of both immune system and glandular-resident cellular pathways, mainly salivary gland epithelial cells.
Unravelling the biological and molecular complexity of pSS is still a great challenge but much progress has been made
in recent years in basic and translational research eld, allowing the identication of potential novel targets for therapy
development. Despite such promising novelties, however, none therapy has been specically approved for pSS treat-
ment until now. In recent years, growing evidence has supported the modulation of Janus kinases (JAK) - signal trans-
ducers and activators of transcription (STAT) pathways as treatment strategy immune mediated diseases. JAK-STAT
pathway plays a crucial role in autoimmunity and systemic inammation, being involved in signal pathways of many
cytokines. This review aims to report the state-of-the-art about the role of JAK-STAT pathway in pSS, with particular
focus on available research and clinical data regarding the use of JAK inhibitors in pSS.
Sjögren’s syndrome • JAK molecules • STAT molecules • cytokines
Abstract
Keywords
Address for correspondence:
*Francesco Ciccia, MD, PhD, Professor of Rheumatology, University della
Campania L. Vanvitelli, Via S. Pansini 5, 80127 Naples Italy. Telephone and
Fax: +390815666752. E-mail: francesco.ciccia@unicampania.it
classication.[6]
Mechanisms underlying pSS pathogenesis are intricate, in-
volving multiplanar and, at the same time, interlinked levels,
e.g., genetic predisposition, epigenetic modications, and
the dysregulation of both the immune system and glandular-
resident cellular pathways, mainly salivary gland epithelial
cells (SGECs).[1,7–9] SGECs are not only damaged by the in-
ammatory process but are leading actors actively involved in
several immune pathophysiology pSS processes.[7,8] Infectious
and/or exogenous agents might be involved in triggering the
disease in predisposed individuals, enhanced by endogenous
factors.[10] Unraveling the biological and molecular complexity
of pSS is still a great challenge, but much progress has been
made in recent years in the basic and translational research
eld, allowing the identication of potential novel targets for
therapy development. Despite such promising novelties,[11]
however, no therapy has been specically approved for pSS
treatment until now.[12]
In recent years, growing evidence has supported the modula-
tion of Janus kinases (JAK)–signal transducers and activators
of transcription (STAT) pathways as a treatment strategy for
96
RHEUMATOLOGY AND IMMUNOLOGY RESEARCH
Review • DOI: 10.2478/rir-2022-0017 • 3(3) • 2022 • 95–102
rheumatoid arthritis (RA) and spondyloarthritis (SpA), lead-
ing to the development of clinical trials and, nally, to the ap-
proval of di󰀨erent JAK inhibitors (JAK-i) agents for clinical use
in RA and SpA.[13–15] JAK-STAT pathway plays a crucial role in
autoimmunity and systemic inammation and are involved in
signal pathways of many cytokines, and, for this reason, an
increasing number of clinical trials with JAK-i have been per-
formed even on other immune-mediated systemic diseases
beyond RA and SpA, such as connective tissue diseases.[16]
This review aims to report the state of the art about the role of
JAK-STAT pathway in pSS, with particular focus on available
research and clinical data regarding the use of JAK-i in pSS.
JAK-STAT Pathway and pSS Cytokine Landscape
The family of JAK-STAT molecules, including 4 JAK intracel-
lular tyrosine kinases (i.e., JAK1, JAK2, JAK3, and TYK2)
and seven transcription factors STAT (STAT1, STAT2, STAT3,
STAT4, STAT5a and 5b, and STAT6), represents a crucial
system involved in the transduction of signaling of several
cytokines related to immune responses, inammation, cell
activation, and survival.[17] Following the cytokine binding to
its receptor on the cell membrane, JAK transfers phosphates
from ATP (adenosine triphosphate) to intracellular domains
of the cytokine receptor, to JAK members themselves, and
to other downstream signaling molecules, such as STAT, that
translocate to the nucleus and modulate the expression of
dened gene sets.[17]
JAK-STAT dysregulation has been associated with di󰀨erent
immune system disorders.[18] Therefore, JAK-i, which causes
competitive ATP binding and blocks the abovementioned cas-
cade of events and gene expression has become an excel-
lent novel therapeutic strategy in rheumatology.
Several JAK-i have been or are being developed, includ-
ing rst-generation pan-JAK-i, i.e., tofacitinb, baricitinib,
ruxolitinib, and pefacitinib and second-generation more se-
lective JAK-i, i.e., lgotinib, upadacitinib, and decernotinib.
Tofacitinib acts by blocking JAK1 and JAK3 but has also a
role in JAK2 and TYK2 inhibition and is the rst oral JAK-i
approved for the treatment of RA[19–21] and psoriatic arthritis
(PsA).[22] Baricitinib is a JAK1 and JAK2 inhibitor used in RA
treatment,[23] such as lgotinib[24,25] and upadacitinb,[26,27] that
inhibits more selectively JAK1. Given the high number of cy-
tokines signaling through JAK-STAT, the revolutionary impact
of oral anti-JAK small molecule therapy lies in being able to
simultaneously block multiple cytokines and the related path-
ways, e󰀨ectively overcoming the direct block of single cyto-
kines that is at the basis of therapies with biotechnological
anti-cytokine drugs (Figure 1). In the context of pSS, this also
translates into the possibility of nding and developing ef-
fective therapies that target multiple pathways with a single
drug, whereas anti-cytokine biotechnological therapies have
instead dramatically failed in several clinical trials.[12]
The cytokine landscape characterizing pSS is extremely com-
plex and heterogeneous. Among cytokines signaling through
JAK-STAT, IL-6, IL-7, IL-21, and IL-23 have been demonstrated
to be potentially involved in pSS pathogenesis. The classical
pro-inammatory cytokine IL-6 has been found to be increased
in serum, saliva, and tears of patients with pSS and linked to
the pathogenesis of the disease.[28–30] Despite the pathogenetic
contribution of IL-6 in crucial immune processes such as the
di󰀨erentiation and activation of both B and T cells, the use of
tocilizumab, a recombinant humanized monoclonal antibody act-
ing as an IL-6 receptor antagonist, did not improve systemic in-
volvement and symptoms over 24 weeks of treatment compared
with placebo in a multicenter double-blind randomized placebo-
controlled trial in pSS patients.[31] IL-21 is a key cytokine in the type
I interferon (IFN) signaling pathway, in the generation of follicu-
lar subtypes, and IL-17–producing T helper (Th) cells, as well as
in plasma cell di󰀨erentiation and B-cell activation. High levels of
IL-21 have been demonstrated in the pSS sera, which are cor-
related with lower memory B-cell and higher naïve B-cell per-
centages.[32,33] RNA sequencing of MSG also demonstrated
signicantly increased levels of IL-21 and IL-21-inducible genes
such as IL-21R, JAK3, STAT1, HLA-B, CCR7, and C-X-C motif
ligand 10 (CXCL10) in pSS patients.[34] The increased IL-21
signature gene expression was associated with an increased
EULAR Sjögren’s Syndrome Disease Activity Index score
(ESSDAI)[35] and increased enrichment of B cells, memory B
cells, CD4+ T cells, and CD8+ T cells.[34] Interestingly, the ex-
pansion of IL-21 T-follicular-helper (Tfh) under the control of
ICOS (inducible co-stimulator) has been demonstrated as a
characteristic of pSS with ectopic germinal centers and MALT
lymphoma.[36]
IL-23A is a pro-inammatory cytokine required for Th17 and
innate lymphoid cells (ILC) 3 maintenance and expansion
and the production of type 3 cytokines such as IL-17 and
IL-22. Intense IL-23 expression has been demonstrated,
Figure 1: Main cytokines signaling through JAK-STAT involved in pSS
pathogenesis.
97
RHEUMATOLOGY AND IMMUNOLOGY RESEARCH
Review • DOI: 10.2478/rir-2022-0017 • 3(3) • 2022 • 95–102
of the immune system.[51] The hyperexpression of IFN sys-
tems in pSS is sustained by chronic reverberating processes,
such as autoantigenic overload and lack of control mecha-
nisms, that act together with genetic susceptibility and epi-
genetic modications.[52] Despite the di󰀨erence between IFNs
in signaling via specic cell surface receptor complexes,
they activate some common downstream pathways, where
JAK-STAT is one of the most crucial ones among them.[51]
Targeting JAK-STAT can therefore result in the modulation
of biological consequences of IFN activation. In pSS, type
I IFN plays a central role in initiating and enhancing inam-
mation in the context of salivary glands, where it is mainly
released by plasmacytoid dendritic cells (pDCs) and SGECs
after an exogenous, likely viral or bacterial, or endogenous,
e.g., autoantigens, trigger.[52] By the expression of more than
2000 genes, the biological e󰀨ects of type I IFN are impressive
and range from innate responses, such as the maturation
of DC with expression of costimulatory molecules, antigen
presentation, and upregulation of chemokines, to T and B
lymphocyte stimulation and activation.[51,52] Among others, in
fact, following type I IFN release, crucial adaptive immune re-
sponses occur, e.g., the stimulation of CD8+ lymphocytes, the
di󰀨erentiation of Th1 and Th17 lymphocytes, the suppression
of T regulatory (Treg) lymphocyte activity, and the induction
of B-cell hyperactivity, by both enhancing the B-cell activating
factor (BAFF) release and lowering of B-cell receptor (BCR)
threshold required for B-cell activation.[52] B-cell dysregula-
tion is one of the most impacting pillars in pSS pathogenesis
leading to both autoimmunity and lymphoproliferation that
accounts for many clinical manifestations and the risk of lym-
phoma evolution in about 5%–10% of pSS patients.[3,4] Type
II IFN sources in pSS are mainly T lymphocytes, but also NK,
B cells, macrophages and, to a less extent, DC, being this
system involved in antimicrobial protection, apoptosis, in-
ammation and tissue damage.[51] Very novel data report the
presence of an amplication loop between interleukin IL-7,
a pivotal cytokine in T-cell responses and T-cell–dependent
activation of SGECs and B lymphocytes in pSS, and IFN-γ,
supporting the role of the IFN system as a bridge milestone
connecting leading actors in pSS pathogenesis, i.e., SGECs,
T and B cells.[45] Recently, many studies also focused on type
III IFN, with increasing evidence of possible contribution to
both autoimmune and malignant disorders.[53,54]
Classically, pSS has been indicated as a type I IFN-driven
disease,[52] but discrepancies in di󰀨erent IFN signatures
expression between peripheral blood and MSG biopsies
have been reported also in the same patient, with a predomi-
nant type I IFN signature in the former and type II in the lat-
ter samples.[46] Higher IFN-γ transcriptional levels were ob-
served in MSG biopsies of patients developing lymphoma,[46]
suggesting a possible contribution of IFN systems also in
the prediction of the risk of lymphoma evolution in pSS.[46,55]
Other authors reported 3 di󰀨erent subsets of IFN expression
in MSG biopsies from pSS patients, i.e., purely type I, purely
by immunohistochemical staining, in submandibular glands
of C57BL/6.NOD-Aec1Aec2 mice and in pSS salivary gland
biopsies within lymphocytic foci and on epithelial tissues.[37,38] IL-
23 increased expression is associated in pSS salivary glands
with the increased expression of IL-17 and IL-17 producing
cells, mainly Th17 and ILC3.[37] According to the increased
IL-23 expression, IL-22 and STAT3 are also signicantly
increased at both protein and mRNA levels in the inamed
salivary glands of patients with pSS and accompanied by the
expansion of IL-22-producing cells.[37] Interestingly, Barone
et al.[39] by using a virus-induced model of autoantibody for-
mation in the salivary glands of adult mice conrmed the role
of IL-22 in pSS pathogenesis, demonstrating that IL-22 pro-
vides a mechanistic link between mucosal infection, B-cell
recruitment, and humoral autoimmunity. IL-22 receptor en-
gagement was in fact necessary and su󰀩cient to promote
di󰀨erential expression of CXCL12 and CXCL13 in epithelial
and broblastic stromal cells that, in turn, is pivotal for B-cell
recruitment and organization of the TLOs (tertiary lymphoid
organs). Accordingly, IL-22 blockade impairs and reverses
TLO formation and autoantibody production.
An increasing body of evidence suggests also a signicant
role of IL-7 axis in pSS, by driving both T-cell responses
and B lymphoneogenesis. IL-7 and its receptor IL-7Rα lev-
els have been demonstrated to be increased in pSS salivary
glands, the latter correlating with the severity of sialadenitis,
and involved in the development of pSS ectopic lymphoid
structures.[40] Interestingly, the exogenous administration of
IL-7 was able to accelerate pSS onset in a mouse model,
whereas pSS development was prevented by blocking IL-
7Rα signal, suggesting that therapeutic intervention on this
axis, by a direct block or an indirect inhibition of downstream
molecules, e.g., JAK-STAT, might be useful in pSS.[41,42] By
signaling through its receptor featuring the common IL-7Rα
chain, in addition to a specic chain named TSLPR (Thymic
stromal lymphopoietin receptor), the pathway of thymic stro-
mal lymphopoietin (TSLP), a novel biomarker for pSS and
related lymphoproliferation,[43,44] also involves JAK-STAT, and
its e󰀨ects might be modulated by JAK-i agents. Furthermore,
very recent data reported that IL-7 secreted by SGECs under
IFN inuence may activate T cells in pSS, which in turn se-
crete IFN-γ, enhancing a vicious circle that amplies one of
the main pathogenetic system in connective tissue diseases,
i.e., IFN itself.[45]
Among such a rich cytokine networking in pSS, growing
evidence supports a prominent role of type I IFN (mainly
IFN-α and IFN-β), type II (IFN-γ), and, more recently, type III
(IFN-λ) also in pSS pathogenesis, based on studies report-
ing the upregulation of IFN-regulated genes (IRGs, proling
the so-called IFN signatures) both in peripheral blood and in
MSG biopsy specimens.[46–50] IFNs are primarily involved in
host defense against infections but also in cell di󰀨erentiation,
proliferation, survival, and death and are crucial regulators
98
RHEUMATOLOGY AND IMMUNOLOGY RESEARCH
Review • DOI: 10.2478/rir-2022-0017 • 3(3) • 2022 • 95–102
cells, broblasts, and endothelial cells.[63] CXCL10 and its re-
ceptor CXCR3 have been involved in the pathogenesis of
pSS since they are up-regulated in pSS MSG and contrib-
ute to the chemotaxis of immune cells and their accumula-
tion in the context of inamed salivary glands.[64–66] Beside
the strong expression of JAK1 and JAK2 in MSG biopsies of
pSS patients,[57] Aota et al.[57] demonstrated that baricitinib
signicantly inhibited IFN-γ-induced CXCL10 production in
an immortalized human salivary gland ductal-cell clone and
by a western blot analysis showed also its ability to strongly
inhibit the phosphorylation of STAT1 and, to a less extent,
of STAT3. These data suggested a possible role of JAK-i
baricitinib in pSS treatment.
Very recently, the e󰀩cacy and safety of baricitinib for active
pSS patients have been explored in a pilot non-controlled
trial.[67] This study enrolled 11 pSS patients, all fullling 2016
ACR (American college of rheumatology)/EULAR (European
Alliance of Associations for Rheumatology) classication cri-
teria for pSS, showing a ESSDAI of at least 5. The improve-
ment of ESSDAI of at least 3 points has been considered as
the minimal clinical improvement expected. Other measures,
such as European pSS Patient Reported Index (ESSPRI),
Phisycian Global Assessment Score (PGA), Immunoglobulin
G (IgG), and remission/improvement of single-organ mani-
festations have been also collected and evaluated. Patients
were treated with baricitinib 2 mg per day and followed up at
3 months and 6 months after starting the therapy. Baricitinib
treatment led to a signicant ESSDAI reduction, as did with
regard to the ESSPRI and PGA. At 6 months, 88.9% patients
achieved minimal clinical improvement of ESSDAI.[67] A de-
creasing trend in IgG and ESR (Erythrocyte sedimentation
rate) levels was also observed. Main clinical manifestations
showing improvement compared with baseline were skin
rash and arthritis, consistent with the study of baricitinib in ac-
tive SLE patients,[68] weight loss, anemia, and cytopenia. Two
pSS patients with interstitial lung disease (ILD) and cough,
short of breath, and dyspnea after exertion were relieved in
symptoms, together with an improvement of lung involve-
ment in follow-up HRCT (High-resolution computed tomog-
raphy) scan. A are of HBV (hepatitis B virus) was the only
adverse event reported.[67] Besides major limitations of the
study, mainly the absence of a control group, the treatment
with baricitinib appears promising in pSS, and high-quality
randomized controlled clinical trials are needed to conrm
these preliminary results.
Filgotinib
Lee et al.[69] demonstrated that lgotinib, a selective JAK1
inhibitor, suppressed in pSS the expression of IFN-related
genes and of BAFF in human SGECs, as well as the BAFF
production in salivary gland organoids. In addition, after l-
gotinib administration in mice models, both increased sali-
vary ow rates and amelioration of lymphocytic inltration of
type II, and a mixed type I/type II pattern and described a
link between these di󰀨erent patterns and peculiar clinical
manifestations.[47] More recently, a very innovative study[48]
proposed a new classication of pSS according to results ob-
tained from a huge multi-omic analysis performed on periph-
eral blood of pSS patients and healthy volunteers, highlight-
ing a clear centrality of IFN signatures in molecular subsetting
of pSS patients. Among 4 di󰀨erent identied clusters, namely
C1, C2, C3, and C4, those pSS patients belonging to C1,
C3, and C4 were sharply characterized by specic combi-
nations between di󰀨erent magnitudes of type I and type II
IFN expression.[48] Although an integration between periph-
eral blood- and tissue-derived data is mandatory in the next
future, considering that all crucial events for pSS develop-
ment and maintenance occur in salivary glands, this novel
approach is the rst step for developing precision medicine-
driven therapies overcoming heterogeneity issues of pSS
that led in the past to the failure of many clinical trials.[12,48,56]
With regards to IFN-λ, a higher expression in MSG from pSS,
compared with non-pSS sicca control subjects, has been
demonstrated,[49] and, more recently, a synergistic e󰀨ect be-
tween IL-29, which belongs to type III IFN system, and IFN-α
in the induction of BAFF and CXCL10 by prolonged STAT1
phosphorylation in salivary gland epithelium has been also
reported.[50]
Data regarding JAK and/or STAT expression in pSS salivary
glands are limited. Aota et al.[57] demonstrated a strong JAK1
and JAK2 expression, respectively, in ductal and acinar cells
of MSG biopsies of pSS patients by immunohistochemi-
cal analysis. STAT1[58,59] and STAT3[37,60,61] expression has
been found also to be increased in pSS MSG biopsies and
linked to IFN-α, IFN-γ, and IL-6 stimulation of the former[58,59]
and to IL-22 and IL-17 overexpression[37,60,61] of the latter.
Furthermore, very recent data demonstrated that STAT3 is
also implicated in epigenetic DNA methylation/hydroxymeth-
ylation processes in pSS, mostly a󰀨ecting IFN-α- and IFN-
γ-regulated genes, as well as the oxidative stress pathways,
and that JAK-i agents (AG490 and ruxolitinib) were able to
reverse the global DNA hydroxymethylation mediated by
IFNα, IFNγ, and H2O2 in human SGECs.[62]
JAK Inhibition in pSS
Baricitinib
There are no many basic research data about JAK-i in pSS.
A recent study[57] demonstrated that baricitinib, a JAK1 and
JAK2 inhibitor, suppressed the destruction of acinar cells in
the salivary gland of pSS patients by abrogating IFN-γ-induced
CXCL10 expression and CXCL10-dependent immune cell
inltration in human salivary gland ductal cells. CXCL10 is
a chemokine induced by IFN-γ via JAK-STAT during Th1 im-
mune responses, released by peripheral blood mononuclear
99
RHEUMATOLOGY AND IMMUNOLOGY RESEARCH
Review • DOI: 10.2478/rir-2022-0017 • 3(3) • 2022 • 95–102
salivary glands were reported,[69] suggesting that lgotinib
might be a novel therapy to directly target salivary gland
inammation and, possibly, lymphoproliferation in pSS
patients.
Results from a multicenter, double-blind placebo-controlled
randomized phase 2 clinical trial including an arm with
lgotinib, besides other agents (i.e., lanraplenib and tirabru-
tinib), in order to assess both safety and e󰀩cacy in patients
with active pSS (ESSDAI ≥ 5), have been very recently pub-
lished.[70] Patients randomized to the lgotinib arm received
the dosage of 200 mg per day for 48 weeks. The primary
endpoint was dened as the proportion of patients fullling
at week 12 both protocol-specied improvement and non-
worsening criteria, based on C-reactive protein (CRP) and
pSS-related symptoms, assessed by the visual analog scale
(VAS) of global disease, pain, oral dryness, ocular dryness,
and fatigue. Change in ESSPRI and ESSDAI was included as
a secondary endpoint and assessed at week 12 and week 24.
Exploratory e󰀩cacy endpoints included objective tests, such
as Schirmer’s test and salivary ow (unstimulated and stimu-
lated), treatment response on specic ESSDAI domains, and
ESSDAI score change from baseline in subgroups of patients.
Exploratory biomarker-related endpoint was the change from
baseline for selected peripheral biomarkers, e.g., IgA, IgG,
IgM, rheumatoid factor (RF) and CRP, B cell and plasma cell
subset, and IFN signature, for each patient at week 4, 12,
and 24.[70]
At week 12, 43.3% of the lgotinib group achieved the primary
endpoint, although no statistically signicant di󰀨erence was
found compared with the placebo arm.[70] Neither secondary
endpoint was met. However, some notable evidence emerged
from the trial. Change of ESSDAI appeared more pronounced
after lgotinib treatment in the pSS subgroup of patients with
baseline ESSDAI ≥ 14 or without disease-modifying antirheu-
matic drugs/corticosteroids. Moreover, by week 24, greater
decreases in RF, IgM, IgG, and IgA were seen in the lgotinib
group compared with placebo, and, very interestingly, the IFN
activity was signicantly reduced from baseline at week 4 and
week 12. Cytosolic DNA sensing and chemokine signaling
pathways were also reduced by lgotinib therapy. In explor-
atory analyses, the salivary rate and tear production resulted
stabilized at similar levels compared with baseline during the
treatment with lgotinib.[70] Finally, most adverse events were
not severe, and overall safety and tolerability were consistent
with the already known safety prole. In light of this, although
primary and secondary endpoints were not met, these obser-
vations support post hoc analyses in specic subgroups of
pSS patients, possibly guided by peculiar biomarkers, which,
together with a general deep revision of pSS outcome mea-
sures for clinical trials, already ongoing,[71] might lead to tar-
get more accurately pSS patients and potentially to prove e󰀩-
cacy of promising novel therapeutic agents, such as lgotinib,
for use in clinical practice.[70]
Tofacitinib
Autophagy is one additional altered mechanism in pSS,
which is involved in several homeostatic functions and both
in innate and adaptive immune responses.[72,73] Deciency of
autophagy has been associated with increased inammation
by IL-6 and accumulation of JAK-STAT components.[74] In a
recent study,[75] Barrera et al.[75] evaluated autophagy dysreg-
ulation in pSS and its link with JAK-STAT system by analyzing
MSG biopsies from both pSS patients and control subjects
and by generating autophagy-decient (ATG5 knockdown)
3 dimensional (3D) salivary glands acini, which were then
incubated in the presence or in absence of the JAK1 and
JAK3 blocking agent tofacitinib. MSG from pSS patients
showed decreased ATG5 expression, correlating negatively
with increased activation of STAT1 and STAT3. Increased ex-
pression of STAT1 and IL-6 correlated with ESSDAI and the
presence of anti-SSA antibodies.[75] ATG5-decient 3D-acini
showed also increased expression of pro-inammatory cyto-
kines such as IL-6, interestingly reversed by tofacitinib.[75]
Ruxolitinib
The biological e󰀨ects of ruxolitinib, a JAK1 and JAK2 inhibi-
tor, on mesenchymal stromal cells (MSCs), isolated from sali-
vary glands of both pSS patients and controls, have been
recently evaluated in vitro.[76] A ruxolitinib-mediated inhibi-
tion of IFN-γ-induced expression of MSC immunomodulatory
markers, such as HLA-DR (Major histocompatibility complex
(MHC) II cell surface receptor) expression, has emerged by
these experimental studies, together with the block of CD4+
T cell chemotaxis through the inhibition of MSC production of
CXCL9, CXCL10, and CXCL11, suggesting potential implica-
tions for ruxolitinib in pSS therapy.[76]
Conclusions
JAK-STAT inhibition has become in recent last years a new
therapeutic option approved for clinical use in immune-
mediated disorders and tested in an increasing number of
clinical trials for other autoimmune diseases, including pSS.
The variegate cytokine landscape signaling through the JAK-
STAT system and, among others, the prominent importance
of IFN pathways in both pSS pathogenesis and patient sub-
setting suggest the potential role of JAK-i in treating pSS by
modulating crucial molecular and biological events for the
disease development and maintenance. In vitro and in vivo
data seem to support this hypothesis, together with encour-
aging results from clinical trials, despite the inadequacy of
outcome measures available at the moment. However, many
e󰀨orts have still to be dedicated to more clearly elucidate the
e󰀨ects of blocking JAK-STAT pathways in pSS, not only in
the perspective of controlling inammation but also in that of
rescuing the homeostatic complex functions of the salivary
gland epithelium.[77]
100
RHEUMATOLOGY AND IMMUNOLOGY RESEARCH
Review • DOI: 10.2478/rir-2022-0017 • 3(3) • 2022 • 95–102
[1] Moutsopoulos HM. Sjogren’s Syndrome: Autoimmune Epithelitis.
Clin Immunol Immunopathol. 1994;72:162–165.
[2] Mariette X, Criswell LA. Primary Sjogren’s Syndrome. N Engl J
Med. 2018;378:931–939.
[3] Zintzaras E, Voulgarelis M, Moutsopoulos HM. The Risk of Lym-
phoma Development in Autoimmune Diseases: A Meta-Analysis.
Arch Intern Med. 2005;165:2337–2344.
[4] Voulgarelis M, Dafni UG, Isenberg DA, et al. Malignant Lymphoma
in Primary Sjogren’s Syndrome: A Multicenter, Retrospective, Clinical
Study by the European Concerted Action on Sjogren’s Syndrome.
Arthritis Rheum. 1999;42:1765–1772.
[5] Chatzis L, Pezoulas VC, Ferro F, et al. Sjogren’s Syndrome: The
Clinical Spectrum of Male Patients. J Clin Med. 2020;9.
[6] Shiboski CH, Shiboski SC, Seror R, et al. 2016 American College
of Rheumatology/European League against Rheumatism Classica-
tion Criteria for Primary Sjogren’s Syndrome: A Consensus and Data-
Driven Methodology Involving Three International Patient Cohorts.
Arthritis Rheumatol. 2017;69:35–45.
[7] Mitsias DI, Kapsogeorgou EK, Moutsopoulos HM. The Role of
Epithelial Cells in the Initiation and Perpetuation of Autoimmune
Lesions: Lessons from Sjogren’s Syndrome (Autoimmune Epithelitis).
Lupus. 2006;15:255–261.
[8] Verstappen GM, Pringle S, Bootsma H, et al. Epithelial- Immune
Cell Interplay in Primary Sjogren Syndrome Salivary Gland
Pathogenesis. Nat Rev Rheumatol. 2021;17:333–348.
[9] Riviere E, Pascaud J, Tchitchek N, et al. Salivary Gland
Epithelial Cells from Patients with Sjogren’s Syndrome Induce B-
Lymphocyte Survival and Activation. Ann Rheumatol Dis. 2020;79:
1468–1477.
[10] Bjork A, Mofors J, Wahren-Herlenius M. Environmental Factors
in the Pathogenesis of Primary Sjogren’s Syndrome. J Intern Med.
2020;287:475–492.
[11] Bowman SJ, Fox R, Dorner T, et al. Safety and E󰀩cacy of
Subcutaneous Ianalumab (VAY736) in Patients with Primary Sjo-
gren’s Syndrome: A Randomised, Double-Blind, Placebo-Controlled,
Phase 2b Dose-Finding Trial. Lancet. 2022;399:161–171.
[12] Gandolfo S, De Vita S. Emerging Drugs for Primary Sjogren’s
Syndrome. Expert Opin Emerg Drugs. 2019;24:121–132.
[13] Wang F, Sun L, Wang S, et al. E󰀩cacy and Safety of Tofaci-
tinib, Baricitinib, and Upadacitinib for Rheumatoid Arthritis: A Sys-
tematic Review and Meta-Analysis. Mayo Clin Proc. 2020;95:
1404–1419.
[14] Keeling S, Maksymowych WP. JAK Inhibitors, Psoriatic Arthritis,
and Axial Spondyloarthritis: A Critical Review of Clinical Trials. Expert
Rev Clin Immunol. 2021;17:701–715.
[15] Crispino N, Ciccia F. JAK/STAT Pathway and Nociceptive
Cytokine Signalling in Rheumatoid Arthritis and Psoriatic Arthritis.
Clin Exp Rheumatol. 2021;39:668–675.
[16] You H, Xu D, Zhao J, et al. JAK Inhibitors: Prospects in
Connective Tissue Diseases. Clin Rev Allergy Immunol. 2020;59:
334–351.
[17] Villarino AV, Kanno Y, O’Shea JJ. Mechanisms and Conse-
quences of Jak-STAT Signaling in the Immune System. Nat Immunol.
2017;18:374–384.
[18] Hu X, Li J, Fu M, et al. The JAK/STAT Signaling Pathway: From
Bench to Clinic. Signal Transduct Target Ther. 2021;6:402.
[19] van Vollenhoven RF, Fleischmann R, Cohen S, et al. Tofacitinib
or Adalimumab versus Placebo in Rheumatoid Arthritis. N Engl J
Med. 2012;367:508–519.
[20] Lee EB, Fleischmann R, Hall S, et al. Tofacitinib versus
Methotrexate in Rheumatoid Arthritis. N Engl J Med. 2014;370:
2377–2386.
[21] Fleischmann R, Mysler E, Hall S, et al. E󰀩cacy and Safety of
Tofacitinib Monotherapy, Tofacitinib with Methotrexate, and Adalim-
umab with Methotrexate in Patients with Rheumatoid Arthritis (ORAL
Strategy): A Phase 3b/4, Double-Blind, Head-to-Head, Randomised
Controlled Trial. Lancet. 2017;390:457–468.
[22] Mease P, Hall S, FitzGerald O, et al. Tofacitinib or Adalimumab
versus Placebo for Psoriatic Arthritis. N Engl J Med. 2017;377:1537–
1550.
[23] Taylor PC, Keystone EC, van der Heijde D, et al. Baricitinib ver-
sus Placebo or Adalimumab in Rheumatoid Arthritis. N Engl J Med.
2017;376:652–662.
[24] Combe B, Kivitz A, Tanaka Y, et al. Filgotinib versus Placebo or
Adalimumab in Patients with Rheumatoid Arthritis and Inadequate
Response to Methotrexate: A Phase III Randomised Clinical Trial.
Ann Rheum Dis. 2021;80:848–858.
[25] Genovese MC, Kalunian K, Gottenberg JE, et al. E󰀨ect of
Filgotinib vs Placebo on Clinical Response in Patients with Moderate
to Severe Rheumatoid Arthritis Refractory to Disease-Modifying An-
tirheumatic Drug Therapy: The FINCH 2 Randomized Clinical Trial.
JAMA. 2019;322:315–325.
[26] Fleischmann R, Pangan AL, Song IH, et al. Upadacitinib versus
Placebo or Adalimumab in Patients with Rheumatoid Arthritis and
an Inadequate Response to Methotrexate: Results of a Phase III,
Double-Blind, Randomized Controlled Trial. Arthritis Rheumatol.
2019;71:1788–1800.
[27] Smolen JS, Pangan AL, Emery P, et al. Upadacitinib as
Monotherapy in Patients with Active Rheumatoid Arthritis and
Inadequate Response to Methotrexate (SELECT-MONOTHERAPY):
A Randomised, Placebo-Controlled, Double-Blind Phase 3 Study.
Lancet. 2019;393:2303–2311.
[28] Grisius MM, Bermudez DK, Fox PC. Salivary and Serum Interleu-
kin 6 in Primary Sjogren’s Syndrome. J Rheumatol. 1997;24:1089–
1091.
[29] Hulkkonen J, Pertovaara M, Antonen J, et al. Elevated Inter-
Conict of Interest
Francesco Ciccia is an Editorial Board Member of the journal. This article was subject to the journal’s standard procedures, with peer review
handled independently of this member and his research group.
References
101
RHEUMATOLOGY AND IMMUNOLOGY RESEARCH
Review • DOI: 10.2478/rir-2022-0017 • 3(3) • 2022 • 95–102
Exp Rheumatol. 2019;37 Suppl 118:55–64.
[44] Gandolfo S, Fabro C, Kapsogeorgou E, et al. Validation
of Thymic Stromal Lymphopoietin as a Biomarker of Primary
Sjogren’s Syndrome and Related Lymphoproliferation: Results in
Independent Cohorts. Clin Exp Rheumatol. 2020;38 Suppl 126:
189–194.
[45] Riviere E, Pascaud J, Virone A, et al. Interleukin-7/Interferon
Axis Drives T Cell and Salivary Gland Epithelial Cell Interactions in
Sjogren’s Syndrome. Arthritis Rheumatol. 2021;73:631–640.
[46] Nezos A, Gravani F, Tassidou A, et al. Type I and II Interferon
Signatures in Sjogren’s Syndrome Pathogenesis: Contributions in
Distinct Clinical Phenotypes and Sjogren’s Related Lymphomagen-
esis. J Autoimmun. 2015;63:47–58.
[47] Hall JC, Baer AN, Shah AA, et al. Molecular Subsetting of In-
terferon Pathways in Sjogren’s Syndrome. Arthritis Rheumatol.
2015;67:2437–2446.
[48] Soret P, Le Dantec C, Desvaux E, et al. A New Molecular Classi-
cation to Drive Precision Treatment Strategies in Primary Sjogren’s
Syndrome. Nat Commun. 2021;12:3523.
[49] Apostolou E, Kapsogeorgou EK, Konsta OD, et al. Expression
of Type III Interferons (IFNlambdas) and Their Receptor in Sjogren’s
Syndrome. Clin Exp Immunol. 2016;186:304–312.
[50] Ha YJ, Choi YS, Kang EH, et al. Increased Expression of In-
terferon-Lambda in Minor Salivary Glands of Patients with Primary
Sjogren’s Syndrome and Its Synergic E󰀨ect with Interferon-Alpha on
Salivary Gland Epithelial Cells. Clin Exp Rheumatol. 2018;36 Suppl
112:31–40.
[51] Negishi H, Taniguchi T, Yanai H. The Interferon (IFN) Class of
Cytokines and the IFN Regulatory Factor (IRF) Transcription Factor
Family. Cold Spring Harb Perspect Biol. 2018;10:a028423
[52] Nordmark G, Alm GV, Ronnblom L. Mechanisms of Disease: Pri-
mary Sjogren’s Syndrome and the Type I Interferon System. Nat Clin
Pract Rheumatol. 2006;2:262–269.
[53] Lasfar A, Zloza A, Silk AW, et al. Interferon Lambda: To-
ward a Dual Role in Cancer. J Interferon Cytokine Res. 2019;39:
22–29.
[54] Lazear HM, Schoggins JW. Diamond MS. Shared and Distinct
Functions of Type I and Type III Interferons. Immunity. 2019;50:907–
923.
[55] Nezos A, Makri P, Gandolfo S, et al. TREX1 Variants in Sjo-
gren’s Syndrome Related Lymphomagenesis. Cytokine. 2020;132:
154781.
[56] Quartuccio L, Mavragani CP, Nezos A, et al. Type I Interferon
Signature May Inuence the E󰀨ect of Belimumab on Immunoglobu-
lin Levels, Including Rheumatoid Factor in Sjogren’s Syndrome. Clin
Exp Rheumatol. 2017;35:719–720.
[57] Aota K, Yamanoi T, Kani K, et al. Inhibition of JAK-STAT Sig-
naling by Baricitinib Reduces Interferon-Gamma-Induced CXCL10
Production in Human Salivary Gland Ductal Cells. Inammation.
2021;44:206–216.
[58] Wakamatsu E, Matsumoto I, Yasukochi T, et al. Overexpression
of Phosphorylated STAT-1alpha in the Labial Salivary Glands of Pa-
tients with Sjogren’s Syndrome. Arthritis Rheumatol. 2006;54:3476–
3484.
[59] Pertovaara M, Silvennoinen O, Isomaki P. Cytokine-Induced
leukin-6 Plasma Levels Are Regulated by the Promoter Region
Polymorphism of the IL6 Gene in Primary Sjogren’s Syndrome and
Correlate with the Clinical Manifestations of the Disease. Rheumatol-
ogy (Oxford). 2001;40:656–661.
[30] Zhao H, Li Q, Ye M, et al. Tear Luminex Analysis in Dry Eye
Patients. Med Sci Monit. 2018;24:7595–7602.
[31] Felten R, Devauchelle-Pensec V, Seror R, et al. Interleukin 6
Receptor Inhibition in Primary Sjogren Syndrome: A Multicentre
Double-Blind Randomised Placebo-Controlled Trial. Ann Rheum Dis
2021;80:329-338.
[32] Kang KY, Kim HO, Kwok SK, et al. Impact of Interleukin-21 in
the Pathogenesis of Primary Sjogren’s Syndrome: Increased Serum
Levels of Interleukin-21 and Its Expression in the Labial Salivary
Glands. Arthritis Res Ther. 2011;13:R179.
[33] Loureiro-Amigo J, Franco-Jarava C, Perurena-Prieto J, et al. Se-
rum CXCL13, BAFF, IL-21 and IL-22 Levels Are Related to Disease
Activity and Lymphocyte Prole in Primary Sjogren’s Syndrome. Clin
Exp Rheumatol. 2021;39 Suppl 133:131–139.
[34] Chen X, Jiang S, Zhou Z, et al. Increased Expression of Inter-
leukin-21-Inducible Genes in Minor Salivary Glands Are Associated
with Primary Sjogren’s Syndrome Disease Characteristics. Rheuma-
tology (Oxford). 2021;60:2979–2989.
[35] Seror R, Bowman SJ, Brito-Zeron P, et al. EULAR Sjogren’s
Syndrome Disease Activity Index (ESSDAI): A User Guide. RMD
Open. 2015;1:e000022.
[36] Pontarini E, Murray-Brown WJ, Croia C, et al. Unique Expansion
of IL-21+ Tfh and Tph Cells under Control of ICOS Identies Sjogren’s
Syndrome with Etopic Germinal Centres and MALT Lymphoma. Ann
Rheumatol Dis. 2020;79:1588–1599.
[37] Ciccia F, Guggino G, Rizzo A, et al. Potential Involvement of
IL-22 and IL-22-Producing Cells in the Inamed Salivary Glands of
Patients with Sjogren’s Syndrome. Ann Rheumatol Dis. 2012;71:
295–301.
[38] Nguyen CQ, Hu MH, Li Y, et al. Salivary Gland Tissue Expres-
sion of Interleukin-23 and Interleukin-17 in Sjogren’s Syndrome:
Findings in Humans and Mice. Arthritis Rheumatol. 2008;58:
734–743.
[39] Barone F, Nayar S, Campos J, et al. IL-22 Regulates Lymphoid
Chemokine Production and Assembly of Tertiary Lymphoid Organs.
Proc Natl Acad Sci U S A. 2015;112:11024–11029.
[40] Bikker A, van Woerkom JM, Kruize AA, et al. Increased
Expression of Interleukin-7 in Labial Salivary Glands of Patients with
Primary Sjogren’s Syndrome Correlates with Increased Inammation.
Arthritis Rheumatol. 2010;62:969–977.
[41] Zhou J, Yu Q. Anti-IL-7 Receptor-Alpha Treatment Ameliorates
Newly Established Sjogren’s-Like Exocrinopathy in Non-obese
Diabetic Mice. Biochim Biophys Acta Mol Basis Dis. 2018;1864:2438–
2447.
[42] Jin JO, Kawai T, Cha S, et al. Interleukin-7 Enhances the Th1
Response to Promote the Development of Sjogren’s Syndrome-
Like Autoimmune Exocrinopathy in Mice. Arthritis Rheumatol.
2013;65:2132–2142.
[43] Gandolfo S, Bulfoni M, Fabro C, et al. Thymic Stromal
Lymphopoietin Expression from Benign Lymphoproliferation to
Malignant B-Cell Lymphoma in Primary Sjogren’s Syndrome. Clin
102
RHEUMATOLOGY AND IMMUNOLOGY RESEARCH
Review • DOI: 10.2478/rir-2022-0017 • 3(3) • 2022 • 95–102
Systemic Lupus Erythematosus: A Double-Blind, Ran-
domised, Placebo-Controlled, Phase 2 Trial. Lancet. 2018;392:
222–231.
[69] Lee J, Lee J, Kwok SK, et al. JAK-1 Inhibition Suppresses
Interferon-Induced BAFF Production in Human Salivary Gland:
Potential Therapeutic Strategy for Primary Sjogren’s Syndrome.
Arthritis Rheumatol. 2018;70:2057–2066.
[70] Price E, Bombardieri M, Kivitz A, et al. Safety and E󰀩cacy of
Filgotinib, Lanraplenib, and Tirabrutinib in Sjogren’s Syndrome:
Randomised, Phase 2, Double-Blind, Placebo-Controlled Study.
2022 Apr 4;keac167; Online ahead of print.
[71] Seror R, Baron G, Camus M, et al. Development and Preliminary
Validation of the Sjogren’s Tool for Assessing Response (STAR):
A Consensual Composite Score for Assessing Treatment E󰀨ect in
Primary Sjogren’s Syndrome. Ann Rheum Dis. 2022;81:979–989.
[72] Colafrancesco S, Barbati C, Priori R, et al. Maladaptive
Autophagy in the Pathogenesis of Autoimmune Epithelitis in Sjogren’s
Syndrome. Arthritis Rheumatol. 2022;74:654–664.
[73] Colafrancesco S, Vomero M, Iannizzotto V, et al. Autophagy Oc-
curs in Lymphocytes Inltrating Sjogren’s Syndrome Minor Salivary
Glands and Correlates with Histological Severity of Salivary Gland
Lesions. Arthritis Res Ther. 2020;22:238.
[74] Harris J, Lang T, Thomas JPW, et al. Autophagy and Inamma-
somes. Mol Immunol. 2017;86:10–15.
[75] Barrera MJ, Aguilera S, Castro I, et al. Tofacitinib Counteracts
IL-6 Overexpression Induced by Decient Autophagy: Implica-
tions in Sjogren’s Syndrome. Rheumatology (Oxford). 2021;60:
1951–1962.
[76] McCoy SS, Parker M, Gurevic I, et al. Ruxolitinib Inhibits IFN-
gamma-Stimulated Sjogren’s Salivary Gland MSC HLA-DR Expres-
sion and Chemokine-Dependent T-cell Migration. Rheumatology
(Oxford) 2022;61:4207-4218
[77] Pringle S, Wang X, Bootsma H, et al. Small-Molecule Inhibitors
and the Salivary Gland Epithelium in Sjogren’s Syndrome. Expert
Opin Investig Drugs. 2019;28:605–616.
STAT1 Activation Is Increased in Patients with Primary Sjogren’s
Syndrome. Clin Immunol. 2016;165:60–67.
[60] Ciccia F, Guggino G, Rizzo A, et al. Interleukin (IL)-22
Receptor 1 Is Over-expressed in Primary Sjogren’s Syndrome and
Sjogren- Associated Non-Hodgkin Lymphomas and Is Regulated by
IL-18. Clin Exp Immunol. 2015;181:219–229.
[61] Vartoukian SR, Tilakaratne WM, Seoudi N, et al. Dysregulation
of the Suppressor of Cytokine Signalling 3-Signal Transducer and
Activator of Transcription-3 Pathway in the Aetiopathogenesis of
Sjogren’s Syndrome. Clin Exp Immunol. 2014;177:618–629.
[62] Charras A, Arvaniti P, Le Dantec C, et al. JAK Inhibitors Sup-
press Innate Epigenetic Reprogramming: A Promise for Patients
with Sjogren’s Syndrome. Clin Rev Allergy Immunol. 2020;58:
182–193.
[63] Groom JR, Luster AD. CXCR3 Ligands: Redundant, Collab-
orative and Antagonistic Functions. Immunol Cell Biol. 2011;89:
207–215.
[64] Ogawa N, Ping L, Zhenjun L, et al. Involvement of the Inter-
feron-Gamma-Induced T Cell-Attracting Chemokines, Interferon-
Gamma-Inducible 10-kd Protein (CXCL10) and Monokine Induced
by Interferon-Gamma (CXCL9), in the Salivary Gland Lesions of
Patients with Sjogren’s Syndrome. Arthritis Rheumatol. 2002;46:
2730–2741.
[65] Aota K, Yamanoi T, Kani K, et al. Inverse Correlation between the
Number of CXCR3(+) Macrophages and the Severity of Inamma-
tory Lesions in Sjogren’s Syndrome Salivary Glands: A Pilot Study. J
Oral Pathol Med. 2018;47:710–718.
[66] Blokland SLM, Kislat A, Homey B, et al. Decreased Circulat-
ing CXCR3 + CCR9+T Helper Cells Are Associated with Elevated
Levels of Their Ligands CXCL10 and CCL25 in the Salivary Gland
of Patients with Sjogren’s Syndrome to Facilitate Their Concerted
Migration. Scand J Immunol. 2020;91:e12852.
[67] Bai W, Liu H, Dou L, et al. Pilot Study of Baricitinib for Active
Sjogren’s Syndrome. 2022;81:1050-1052.
[68] Wallace DJ, Furie RA, Tanaka Y, et al. Baricitinib for
... Advances in genetic, epigenetic, transcriptomic, and proteomic technologies have advanced our understanding of the pathogenesis of SjD. These studies have shown that multiple in ammatory cytokines are involved including: Type I and Type II interferons (IFNs: IFN , IFN , and IFNγ) 20,21 , interleukins (ILs: IL6, IL-7, IL-12, IL-21, and IL-23) 20,22 , and cytokines and chemokines (CXCL10, -11, -12) [23][24][25][26] . Emerging technologies (e.g., single cell and spatial transcriptomics) are unraveling disease-speci c cellular networks and signaling pathways, and spotlight effector cells and therapeutic pathways. ...
... Advances in genetic, epigenetic, transcriptomic, and proteomic technologies have advanced our understanding of the pathogenesis of SjD. These studies have shown that multiple in ammatory cytokines are involved including: Type I and Type II interferons (IFNs: IFN , IFN , and IFNγ) 20,21 , interleukins (ILs: IL6, IL-7, IL-12, IL-21, and IL-23) 20,22 , and cytokines and chemokines (CXCL10, -11, -12) [23][24][25][26] . Emerging technologies (e.g., single cell and spatial transcriptomics) are unraveling disease-speci c cellular networks and signaling pathways, and spotlight effector cells and therapeutic pathways. ...
Preprint
Full-text available
Sjögren's Disease (SjD) is a systemic autoimmune disease without a clear etiology or effective therapy. Utilizing unbiased single-cell and spatial transcriptomics to analyze human minor salivary glands in health and disease we developed a comprehensive understanding of the cellular landscape of healthy salivary glands and how that landscape changes in SjD patients. We identified novel seromucous acinar cell types and identified a population of PRR4+CST3+WFDC2- seromucous acinar cells that are particularly targeted in SjD. Notably, GZMK+CD8 T cells, enriched in SjD, exhibited a cytotoxic phenotype and were physically associated with immune-engaged epithelial cells in disease. These findings shed light on the immune response's impact on transitioning acinar cells with high levels of secretion and explain the loss of this specific cell population in SjD. This study explores the complex interplay of varied cell types in the salivary glands and their role in the pathology of Sjögren's Disease.
... IFN binds to its receptor and activates the JAK/STAT pathway. JAK inhibitors can treat pSS by downregulating the STAT pathway [55,56]. JAK inhibitors regulate pSTAT-1Y701 and pSTAT-3Y705 and activate STAT-3 proteins induced by IFN-α and IFN-γ in human SGECs. ...
... The results of a prospective, randomized, double-blind and multicenter clinical trial demonstrated that tofacitinib improves dry eyes in SS patients. Baritinib inhibits JAK/STAT signal transduction, IFN-γ-induced CXCL10 expression and immune cell chemotactic activity, indicating its effectiveness for the treatment of SS [55][56][57]. NOD.IFN-γ-/-, NOD.IFN-γ R-/-mice have been used to study the role of IFN-γ, in SS pathogenesis. Interestingly, NOD and NOD-derived mice develop disease symptoms spontaneously as a polygenic trait with a female predisposition, which resembles the development of SS in humans. ...
Article
Full-text available
Primary Sjögren’s syndrome (pSS) is a connective tissue disease characterized by a wide spectrum of clinical features, extending from a benign glandular disease to an aggressive systemic disorder and/or lymphoma. The pathogenesis of Sjögren’s syndrome (SS) is not completely understood, but it is assumed that pathogenesis of SS is multifactorial. The studies based on the animal models of SS provided significant insight in SS disease pathogenesis and management. The aim of this review is to summarize current studies on animal models with primary SS-like symptoms and discuss the impact of these studies on better understanding pathogenesis and management of Sjögren’s syndrome. Databases PubMed, Web of Science, Scopus and Cochrane library were searched for summarizing studies on animal models in SS. Available data demonstrate that animal models are highly important for our understanding of SS disease.
... [107][108][109][110][111][112][113][114][115] Due to the important role of JAK-STAT pathway in the pathogenesis of pSS and the success of Jakinibs for the treatment of RA and SLE, Jakinibs seem to be promising therapies for pSS patients. [101,116] At the cell level, JAK1/2 inhibitors can suppress JAK-STAT pathway activated by IFNa/g or H 2 O 2 . This suppression subsequently inhibits downstream effects, including the increase in aberrant DNA epigenetic reprograming, [77] high expression of tissue-type plasminogen activator, [97] and CXCL10 expression. ...
Article
Full-text available
Primary Sjögren's syndrome (pSS) is a systemic autoimmune disease with high prevalence and possible poor prognosis. Though the pathogenesis of pSS has not been fully elucidated, B cell hyperactivity is considered as one of the fundamental abnormalities in pSS patients. It has long been identified that Janus kinases-signal transducer and activator of transcription (JAK-STAT) signaling pathway contributes to rheumatoid arthritis and systemic lupus erythematosus. Recently, increasing numbers of studies have provided evidence that JAK-STAT pathway also has an important role in the pathogenesis of pSS via direct or indirect activation of B cells. Signal transducer and activator of transcription 1 (STAT1), STAT3, and STAT5 activated by various cytokines and ribonucleic acid contribute to pSS development, respectively or synergically. These results reveal the potential application of Janus kinase inhibitors for treatment of pSS, which may fundamentally improve the quality of life and prognosis of patients with pSS.
Article
Objectives: Inflammatory cytokines that signal through the Janus kinases-signal transducer and activator of transcription (JAK-STAT) pathway, especially interferons (IFNs), are implicated in Sjögren's disease (SjD). Although inhibition of JAKs is effective in other autoimmune diseases, a systematic investigation of IFN-JAK-STAT signalling and the effect of JAK inhibitor (JAKi) therapy in SjD-affected human tissues has not been fully investigated. Methods: Human minor salivary glands (MSGs) and peripheral blood mononuclear cells (PBMCs) were investigated using bulk or single-cell (sc) RNA sequencing (RNAseq), immunofluorescence (IF) microscopy and flow cytometry. Ex vivo culture assays on PBMCs and primary salivary gland epithelial cell (pSGEC) lines were performed to model changes in target tissues before and after JAKi. Results: RNAseq and IF showed activated JAK-STAT pathway in SjD MSGs. Elevated IFN-stimulated gene (ISGs) expression associated with clinical variables (eg, focus scores, anti-SSA positivity). scRNAseq of MSGs exhibited cell type-specific upregulation of JAK-STAT and ISGs; PBMCs showed similar trends, including markedly upregulated ISGs in monocytes. Ex vivo studies showed elevated basal pSTAT levels in SjD MSGs and PBMCs that were corrected with JAKi. SjD-derived pSGECs exhibited higher basal ISG expressions and exaggerated responses to IFN-β, which were normalised by JAKi without cytotoxicity. Conclusions: SjD patients' tissues exhibit increased expression of ISGs and activation of the JAK-STAT pathway in a cell type-dependent manner. JAKi normalises this aberrant signalling at the tissue level and in PBMCs, suggesting a putative viable therapy for SjD, targeting both glandular and extraglandular symptoms. Predicated on these data, a phase Ib/IIa randomised controlled trial to treat SjD with tofacitinib was initiated.
Article
Full-text available
Our goal was to investigate the effects of epidermal growth factor (EGF) and interferons (IFNs) on signal transducer and activator of transcription STAT1 and STAT4 mRNA and active phosphorylated protein expression in Sjögren’s syndrome cell culture models. iSGECs (immortalized salivary gland epithelial cells) and A253 cells were treated with EGF, IFN-alpha, -beta, -gamma, or mitogen-activated protein kinase p38 alpha (p38-MAPK) inhibitor for 0–24–48–72 h. STAT1 and STAT4 mRNA expression was quantified by qRT-PCR. Untreated and treated cells were compared using the delta-delta-CT method based on glyceraldehyde-3-phosphate dehydrogenase (GAPDH) normalized relative fold changes. phospho-tyrosine-701-STAT1 and phospho-serine-721-STAT4 were detected by Western blot analysis. STAT4 mRNA expression decreased 48 h after EGF treatment in A253 cells, immortalized salivary gland epithelial cells iSGECs nSS2 (sicca patient origin), and iSGECs pSS1 (anti-SSA negative Sjögren’s Syndrome patient origin). EGF and p38-MAPK inhibitor decreased A253 STAT4 mRNA levels. EGF combined with IFN-gamma increased phospho-STAT4 and phospho-STAT1 after 72 h in all cell lines, suggesting additive effects for phospho-STAT4 and a major effect from IFN-gamma for phospho-STAT1. pSS1 and nSS2 cells responded differently to type I and type II interferons, confirming unique functional characteristics between iSGEC cell lines. EGF/Interferon related pathways might be targeted to regulate STAT1 and STAT4 expression in salivary gland epithelial cells. Further investigation is required learn how to better target the Janus kinases/signal transducer and activator of transcription proteins (JAK/STAT) pathway-mediated inflammatory response in Sjögren’s syndrome.
Preprint
Full-text available
Objectives: Inflammatory cytokines that signal through the JAK- STAT pathway, especially interferons (IFNs), are implicated in Sjogrens Disease (SjD). Although inhibition of JAKs is effective in other autoimmune diseases, a systematic investigation of IFN-JAK-STAT signaling and effect of JAK inhibitor (JAKi) therapy in SjD-affected human tissues has not been reported. Methods: Human minor salivary glands (MSGs) and peripheral blood mononuclear cells (PBMCs) were investigated using bulk or single cell (sc) RNA sequencing (RNAseq), immunofluorescence microscopy (IF), and flow cytometry. Ex vivo culture assays on PBMCs and primary salivary gland epithelial cell (pSGEC) lines were performed to model changes in target tissues before and after JAKi. Results: RNAseq and IF showed activated JAK-STAT pathway in SjD MSGs. Elevated IFN-stimulated gene (ISGs) expression associated with clinical variables (e.g., focus scores, anti-SSA positivity). scRNAseq of MSGs exhibited cell-type specific upregulation of JAK-STAT and ISGs; PBMCs showed similar trends, including markedly upregulated ISGs in monocytes. Ex vivo studies showed elevated basal pSTAT levels in SjD MSGs and PBMCs that were corrected with JAKi. SjD-derived pSGECs exhibited higher basal ISG expressions and exaggerated responses to IFNβ, which were normalized by JAKi without cytotoxicity. Conclusions: SjD patients tissues exhibit increased expression of ISGs and activation of the JAK-STAT pathway in a cell type-dependent manner. JAKi normalizes this aberrant signaling at the tissue level and in PBMCs, suggesting a putative viable therapy for SjD, targeting both glandular and extraglandular symptoms. Predicated on these data, a Phase Ib/IIa randomized controlled trial to treat SjD with tofacitinib was initiated.
Article
Full-text available
Primary Sjogren syndrome (pSS) is the second most common autoimmune disorder worldwide, which, in the worst scenario, progresses to Non-Hodgkin Lymphoma (NHL). Despite extensive studies, there is still a lack of knowledge about developing pSS for NHL. This study focused on cells’ signaling in pSS progression to the NHL type of diffuse large B-cell lymphoma (DLBCL). Using bulk RNA and single cell analysis, we found five novel pathologic-independent clusters in DLBCL based on cells’ signaling. B-cell receptor (BCR) signaling was identified as the only enriched signal in DLBCL and pSS peripheral naive B-cells or salivary gland-infiltrated cells. The evaluation of the genes in association with BCR has revealed that targeting CD79A, CD79B, and LAMTOR4 as the shared genes can provide novel biomarkers for pSS progression into lymphoma.
Article
Full-text available
Purpose: This study aims to compare dry eye parameters before and after COVID-19 infection in dry eye patients. Methods: We included 44 dry eye patients (88 eyes) from our existing dry eye cohort, with 22 belonging to the post-COVID-19 group due to a prior COVID-19 infection and the other 22 forming the non-COVID-19 group as they had no history of COVID-19. We examined and compared the dry eye parameters of the post-COVID-19 group, including the ocular surface disease index (OSDI), Schirmer's test results (ST), non-invasive Keratography tear break-up time (NIKBUT), lipid layer thickness (LLT), Meibomian gland dysfunction (MGD), and the grading of papillae and follicles, both before and after the COVID-19 infection. We also compared the dry eye parameters difference of the post-COVID-19 group with the non-COVID-19 group. Results: The post-COVID-19 group was comprised of individuals with an average age of 38.36 ± 14.99 years, of which 82% were female. The time interval between the two tests was 16.92 ± 5.40 months, which did not differ significantly from the non-COVID-19 group. Compared to the pre-COVID-19 eyes, the post-COVID-19 eyes showed a significant decrease in the average LLT (52.86 ± 18.00 nm vs. 63.00 ± 22.40 nm, p < 0.001), as well as the maximum LLT (67.89 ± 20.81 nm vs. 78.48 ± 20.55 nm, p < 0.001). The MGD in both the upper (1.75 ± 0.84) and lower eyelids (1.43 ± 0.73) worsened after a COVID-19 infection. Additionally, the grading of papillae was worse following a COVID-19 infection (0.61 ± 0.69 vs. 0.16 ± 0.37, p < 0.001). The multivariate linear regression model revealed a negative association between COVID-19 infection and NIKBUT-average (β = -2.98, 95%CI: (-5.82, -0.15), p = 0.039), LLT-average (β = -14.12, 95%CI: (-22.66, -5.59), p = 0.001), and LLT max (β = -15.65, 95%CI: (-23.09, -8.20), p < 0.001). Conclusion: From preliminary results, we concluded that dry eye patients who have been infected with COVID-19 appear to have a more severe dry eye condition, as evidenced by lower LLT, worse papillae and MGD, and shorter NIKBUT. It is important to raise awareness of this potential long-term symptom of COVID-19, especially among existing dry eye patients.
Article
Full-text available
Sjögren's syndrome is a chronic and insidious auto-immune disease characterized by lymphocyte infiltration of exocrine glands. The patients typically present with ocular surface diseases related to dry eye and other systemic manifestations. However, due to the high prevalence of dry eye disease and the lack of objective and clinically reliable diagnostic tools, discriminating Sjögren's syndrome dry eye (SSDE) from non-Sjögren's syndrome dry eye (NSSDE) remains a challenge for clinicians. Diagnosing SS is important to improve the quality of life of patients through timely referral for systemic workups, as SS is associated with serious systemic complications such as lymphoma and other autoimmune diseases. The purpose of this article is to describe the current molecular understanding of Sjögren's syndrome and its implications for novel diagnostic modalities on the horizon. A literature review of the pre-clinical and clinical studies published between 2016 and 2022 was conducted. The SSDE pathophysiology and immunology pathways have become better understood in recent years. Novel diagnostic modalities, such as tear and saliva proteomics as well as exosomal biomarkers, provide hope on the horizon.
Article
Full-text available
Objective To develop a composite responder index in primary Sjögren’s syndrome (pSS): the Sjögren’s Tool for Assessing Response (STAR). Methods To develop STAR, the NECESSITY (New clinical endpoints in primary Sjögren’s syndrome: an interventional trial based on stratifying patients) consortium used data-driven methods based on nine randomised controlled trials (RCTs) and consensus techniques involving 78 experts and 20 patients. Based on reanalysis of rituximab trials and the literature, the Delphi panel identified a core set of domains with their respective outcome measures. STAR options combining these domains were proposed to the panel for selection and improvement. For each STAR option, sensitivity to change was estimated by the C-index in nine RCTs. Delphi rounds were run for selecting STAR. For the options remaining before the final vote, a meta-analysis of the RCTs was performed. Results The Delphi panel identified five core domains (systemic activity, patient symptoms, lachrymal gland function, salivary gland function and biological parameters), and 227 STAR options combining these domains were selected to be tested for sensitivity to change. After two Delphi rounds, a meta-analysis of the 20 remaining options was performed. The candidate STAR was then selected by a final vote based on metrological properties and clinical relevance. Conclusion The candidate STAR is a composite responder index that includes all main disease features in a single tool and is designed for use as a primary endpoint in pSS RCTs. The rigorous and consensual development process ensures its face and content validity. The candidate STAR showed good sensitivity to change and will be prospectively validated by the NECESSITY consortium in a dedicated RCT.
Article
Full-text available
Objective: To characterise filgotinib, lanraplenib, and tirabrutinib safety and efficacy in patients with active Sjögren's syndrome (SS). Methods: This multicentre, double-blind study randomised patients with active primary or secondary SS (European League Against Rheumatism [EULAR] SS disease activity index [ESSDAI] ≥5) to receive filgotinib 200 mg (Janus kinase-1 inhibitor), lanraplenib 30 mg (spleen tyrosine kinase inhibitor), tirabrutinib 40 mg (Bruton's tyrosine kinase inhibitor), or placebo. The composite primary end point was W12 proportion of patients fulfilling protocol-specified improvement criteria (based on C-reactive protein and SS-related symptoms). EULAR SS patient-reported index (ESSPRI) and ESSDAI change from baseline (CFB) were secondary endpoints. Exploratory endpoints included disease-related biomarkers. Treatment-emergent adverse events (AEs) represented safety outcomes. Results: Baseline mean ESSDAI was 10.1, and ESSPRI was 6.2 in the 150 patients who received study drug; 125 completed the 24-week placebo-controlled treatment period. At W12, 43.3% of the filgotinib group achieved the primary end point (p = 0.17 vs placebo) vs 42.3% (p = 0.16), 34.7% (p = 0.33), and 26.7% of lanraplenib, tirabrutinib, and placebo groups, respectively. Neither secondary end point was met. Biomarker reductions included immunoglobulins classically associated with SS disease activity. Filgotinib ESSDAI CFB appeared more pronounced in subgroups with baseline ESSDAI ≥14 or without disease-modifying antirheumatic drugs/corticosteroids. Most AEs were Grade 1 or 2. Conclusions: Three drugs with disparate mechanisms were tested, but no significant differences vs placebo in primary or secondary endpoints were observed. These results may be considered hypothesis-generating, given the drug tolerability, subgroup analysis, and biomarker findings. Trial registration: ClinicalTrials.gov, https://clinicaltrials.gov, NCT03100942.
Article
Full-text available
The Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling pathway was discovered more than a quarter-century ago. As a fulcrum of many vital cellular processes, the JAK/STAT pathway constitutes a rapid membrane-to-nucleus signaling module and induces the expression of various critical mediators of cancer and inflammation. Growing evidence suggests that dysregulation of the JAK/STAT pathway is associated with various cancers and autoimmune diseases. In this review, we discuss the current knowledge about the composition, activation, and regulation of the JAK/STAT pathway. Moreover, we highlight the role of the JAK/STAT pathway and its inhibitors in various diseases.
Article
Full-text available
Objective Salivary gland epithelial cells (SGECs) are key cellular drivers in the pathogenesis of primary Sjögren's syndrome (SS); however, the mechanisms sustaining SGEC activation in primary SS remain unclear. We undertook this study to determine the role of autophagy in the survival and activation of SGECs in primary SS. Methods Primary SGECs isolated from the minor SGs of patients with primary SS or sicca syndrome were evaluated by flow cytometry, immunoblotting, and immunofluorescence to assess autophagy (autophagic flux, light chain 3 IIB [LC3‐IIB], p62, LC3‐IIB+/lysosome‐associated membrane protein 1 [LAMP‐1] staining), apoptosis (annexin V/propidium iodide [PI], caspase 3), and activation (intercellular adhesion molecule, vascular cell adhesion molecule). Focus score and germinal center presence were assessed in the SGs from the same patients to assess correlation with histologic severity. Human SG (HSG) cells were stimulated in vitro with peripheral blood mononuclear cells (PBMCs) and serum from primary SS patients in the presence or absence of autophagy inhibitors to determine changes in autophagy and epithelial cell activation. Results SGECs from primary SS patients (n = 24) exhibited increased autophagy (autophagic flux [P = 0.001]; LC3‐IIB [P = 0.02]; p62 [P = 0.064]; and as indicated by LC3‐IIB/LAMP‐1+ staining), increased expression of antiapoptotic molecules (Bcl‐2 [P = 0.006]), and reduced apoptosis (annexin V/PI [P = 0.002]; caspase 3 [P = 0.057]), compared to samples from patients with sicca syndrome (n = 16). Autophagy correlated with histologic disease severity. In vitro experiments on HSG cells stimulated with serum and PBMCs from primary SS patients confirmed activation of autophagy and expression of adhesion molecules, which was reverted upon pharmacologic inhibition of autophagy. Conclusion In primary SS SGECs, inflammation induces autophagy and prosurvival mechanisms, which promote SGEC activation and mirror histologic severity. These findings indicate that autophagy is a central contributor to the pathogenesis of primary SS and a new therapeutic target.
Article
Full-text available
There is currently no approved treatment for primary Sjögren’s syndrome, a disease that primarily affects adult women. The difficulty in developing effective therapies is -in part- because of the heterogeneity in the clinical manifestation and pathophysiology of the disease. Finding common molecular signatures among patient subgroups could improve our understanding of disease etiology, and facilitate the development of targeted therapeutics. Here, we report, in a cross-sectional cohort, a molecular classification scheme for Sjögren’s syndrome patients based on the multi-omic profiling of whole blood samples from a European cohort of over 300 patients, and a similar number of age and gender-matched healthy volunteers. Using transcriptomic, genomic, epigenetic, cytokine expression and flow cytometry data, combined with clinical parameters, we identify four groups of patients with distinct patterns of immune dysregulation. The biomarkers we identify can be used by machine learning classifiers to sort future patients into subgroups, allowing the re-evaluation of response to treatments in clinical trials.
Article
Background Today, there are still no DMARDs licensed for primary Sjögren Syndrome (pSS) patients. Among the explanations, are the limitations of current outcome measures used as primary endpoints: e.g; high placebo response rate, evaluation of either symptoms or systemic activity, and important features not being assessed. The NECESSITY consortium ( https://www.necessity-h2020.eu/ ), including pSS experts from academia, pharmaceutical industry and patient groups formed to develop a new composite responder index, the Sjögren’s Tool for Assessing Response (STAR) that solve the issues of current outcome measures in pSS and is intended for use in clinical trials as an efficacy endpoint. Objectives To develop a composite responder index in primary Sjögren’s syndrome (pSS): the STAR. Methods To develop the STAR, the NECESSITY consortium used data-driven methods, based on 9 randomized controlled trials (RCTs), and consensus techniques, involving 78 experts and 20 patients. Based on reanalysis of rituximab trials (TRACTISS and TEARS) and literature review, the Delphi panel identified a core set of domains to include in the STAR, with their respective outcome measures. STAR options combining these domains were designed and proposed to the panel to select and improve them. For each STAR option, sensitivity to change was estimated by the C-index (derived from Effect size) in all 9 RCTs. Delphi rounds were run for selecting STAR among these options. The Delphi panel also voted to classify trials as positive, negative or “in between” in regards to primary but also key secondary endpoints. For the options remaining before the final vote, meta-analyses of the RCTs were performed separately for positive and “in between” trials together, and for negative trials. Results The Delphi panel identified 5 core domains (systemic activity, patient symptoms, lachrymal gland function, salivary gland function and biological parameters), and 227 STAR options, combining these domains, were selected to be tested for sensitivity to change. After two Delphi rounds, meta-analyses of the 20 remaining options were performed. The candidate STAR was selected by a final vote based on metrological properties and clinical relevance. In positive/in between trials, candidate STAR detected a difference between arms (OR 3.29, 95%-CI [2.07;5.22], whereas it did not in negative trials (OR 1.53, 95%-CI [0.81;2.91]). Conclusion The candidate STAR is a composite responder index, including in a single tool all main disease features, and is designed for use as a primary endpoint in pSS RCTs. Its rigorous and consensual development process ensures its face and content validity. The candidate STAR showed good sensitivity and specificity to change. The candidate STAR will be prospectively validated in a dedicated three arms RCT of the NECESSITY consortium that will evaluate combination of synthetic DMARDs (hydroxychloroquine + lefunomide or hydroxychloroquine + mycophenolate vs placebo). We encourage the use of STAR in any ongoing and future trials. Table 1. Candidate STAR Domain Point Definition of response Systemic activity 3 Decrease of clinESSDAI ≥ 3 Patient reported outcome 3 Decrease of ESSPRI ≥ 1 point or ≥ 15% Lachrymal gland function 1 Schirmer: If abnormal score at baseline: increase ≥ 5 mm from baseline If normal score at baseline: no change to abnormal Or Ocular Staining Score: If abnormal score at baseline: decrease ≥ 2 points from baseline If normal score at baseline: no change to abnormal Salivary gland function 1 Unstimulated Whole Salivary Flow: If score > 0 at baseline: increase ≥ 25% from baseline If score is 0 at baseline: any increase from baseline or Ultrasound: Decrease ≥ 25% in total Hocevar score from baseline Biological 1 Serum IgG levels: decrease ≥ 10% or Rheumatoid Factor levels: decrease ≥ 25% Candidate STAR responder ≥ 5 points ESSDAI: EULAR Sjögren syndrome disease activity index; ESSPRI: EULAR Sjögren syndrome patient reported index; IgG: Immunoglobulin G; Acknowledgements NECESSITY WP5 STAR development participants: Suzanne Arends (University Medical Center Groningen, Department of Rheumatology and Clinical Immunology, Groningen 9700 RB, Netherlands), Francesca Barone (Centre for Translational Inflammation Research, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK), Albin Björk (Division of Rheumatology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden), Coralie Bouillot (Association Française du Gougerot Sjögren et des Syndromes Secs, France), Guillermo Carvajal Alegria (University of Brest, Inserm, CHU de Brest, LBAI, UMR1227, Brest, France; Service de Rhumatologie, Centre de Référence Maladies Autoimmunes Rares CERAINO, CHU Cavale Blanche, Brest, France), Wen-Hung Chen (GlaxoSmithKline, Research Triangle Park, North Carolina, USA), Kenneth Clark (GlaxoSmithKline Medicines Research Centre, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, United Kingdom), Konstantina Delli (Department of Oral and Maxillofacial Surgery, University Medical Center Groningen (UMCG), University of Groningen, The Netherlands), Salvatore de Vita (Rheumatology Clinic, University Hospital of Udine, Italy), Liseth de Wolff (University Medical Center Groningen, Department of Rheumatology and Clinical Immunology, Groningen 9700 RB, Netherlands), Jennifer Evans (Novartis Pharmaceuticals corporation USA), Stéphanie Galtier (Institut de Recherches Internationales Servier (IRIS), Suresnes Cedex, France), Saviana Gandolfo (Rheumatology Clinic, Department of Medical area, University of Udine, ASUFC, 33100 Udine, Italy), Mickael Guedj (Institut de Recherches Internationales Servier (IRIS), Suresnes Cedex, France), Dewi Guellec (CHU de Brest, Service de Rhumatologie, Inserm, CIC 1412, Brest, France), Safae Hamkour (Center of Translational Immunology, Department of Immunology, University Medical Center Utrecht, Utrecht 3584 GA, Netherlands), Dominik Hartl (Novartis Institutes for BioMedical Research, Basel, Switzerland), Malin Jonsson (Section for Oral and Maxillofacial Radiology, Department of Clinical Dentistry, Faculty of Medicine and Dentistry, University of Bergen, Norway), Roland Jonsson (Broegelmann Research Laboratory, Department of Clinical Science, University of Bergen, Department of Rheumatology, Haukeland University Hospital, Bergen, Norway), Frans Kroese (University Medical Center Groningen, Department of Rheumatology and Clinical Immunology, Groningen 9700 RB, Netherlands), Aike Albert Kruize (University Medical Center Utrecht, Department Rheumatology and Clinical Immunology, Utrecht, Netherlands), Laurence Laigle (Institut de Recherches Internationales Servier (IRIS), Suresnes Cedex, France), Véronique Le Guern (AP-HP, Hôpital Cochin, Centre de référence maladies auto-immunes et systémiques rares, service de médecine interne, Paris, France), Wen-Lin Luo (Department of Biometrics and Statistical Science, Novartis Pharmaceuticals, East Hanover, New Jersey), Esther Mossel (University Medical Center Groningen, Department of Rheumatology and Clinical Immunology, Groningen 9700 RB, Netherlands), Wan-Fai Ng (Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne NE2 4HH, UK), Gaëtane Nocturne (Department of Rheumatology, Université Paris-Saclay, INSERM U1184: Centre for Immunology of Viral Infections and Autoimmune Diseases, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Le Kremlin Bicêtre, Paris, France), Marleen Nys (Global Biometric Sciences, Bristol Myers Squibb, Braine L’Alleud, Belgium), Roald Omdal (Clinical Immunology Unit, Department of Internal Medicine, Stavanger University Hospital, PO Box 8100, 4068, Stavanger, Norway), Jacques-Olivier Pers (LBAI, UMR1227, University of Brest, Inserm, Brest, France and CHU de Brest, Brest, France), Maggy Pincemin (Association Française du Gougerot Sjögren et des Syndromes Secs, France), Manel Ramos-Casals (Department of Autoimmune Diseases, Hospital Clinic de Barcelona Institut Clinic de Medicinai Dermatologia, Barcelona, Catalunya, Spain), Philippe Ravaud (Centre d’Epidémiologie Clinique, Hôpital Hôtel-Dieu, Assistance Publique-Hôpitaux de Paris, Paris, France), Neelanjana Ray (Global Drug Development - Immunology, Bristol Myers Squibb Company, Princeton, New Jersey, USA), Alain Saraux (HU de Brest, Service de Rhumatologie, Univ Brest, Inserm, UMR1227, Lymphocytes B et Autoimmunité, Univ Brest, Inserm, LabEx IGO, Brest, France), Athanasios Tzioufas (Rheumatology Clinic, Department of Medical area, University of Udine, ASUFC, 33100 Udine, Italy), Gwenny Verstappen (University Medical Center Groningen, Department of Rheumatology and Clinical Immunology, Groningen 9700 RB, Netherlands), Arjan Vissink, Marie Wahren-Herlenius (Division of Rheumatology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden). We thank the following experts: Esen Karamursel Akpek, Alan Baer, Chiara Baldini, Elena Bartoloni, Marí-Alfonso Begona, Johan Brun, Vatinee Bunya, Laurent Chiche, Troy Daniels, Paul Emery, Robert Fox, Roberto Giacomelli, John Gonzales, John Greenspan, Robert Moots, Susumu Nishiyama, Elizabeth Price, Christophe Richez, Caroline Shiboski, Roser Solans Laque, Muthiah Srinivasan, Peter Olsson, Tsutomu Takeuchi, Frederick Vivino, Paraskevi Voulgari, Daniel Wallace, Ava Wu, Wen Zhang. We thank the anonymous patients from the NECESSITY Patient Advisory Group and the Sjögren Foundation for their valuable contribution to the Delphi process. We thank EW StClair and AN Baer who generated the baminercept data and made them publicly available. Disclosure of Interests Raphaèle Seror Consultant of: GlaxoSmithKline, Boehringer, Janssen and Novartis, Grant/research support from: GlaxoSmithKline and Amgen, Gabriel Baron: None declared, Marine Camus: None declared, Divi Cornec Consultant of: GlaxoSmithKline, Bristol Myers Squibb, Janssen, Amgen, Pfizer and Roche, Elodie Perrodeau: None declared, Simon J. Bowman Consultant of: Abbvie, Astra Zeneca, Galapagos and Novartis Pharmaceuticals, Michele Bombardieri Consultant of: UCB, Amgen/Medimmune, Janssen, and GlaxoSmithKline, Grant/research support from: Amgen/Medimmune, Janssen, and GlaxoSmithKline, Hendrika Bootsma: None declared, Jacques-Eric Gottenberg Consultant of: AbbVie, Bristol Myers Squibb, Eli Lilly, Galapagos, Gilead, Pfizer, Roche, Sanofi, Novartis, MSD, CSL-Behring and Genzyme, Grant/research support from: Bristol Myers Squibb, Benjamin Fisher Speakers bureau: Bristol Myers Squibb and Novartis, Consultant of: Novartis, Bristol Myers Squibb, Janssen and Servier, Grant/research support from: Servier, Galapagos and Janssen, Wolfgang Hueber Shareholder of: Novartis Pharma, Employee of: Novartis Pharma, Joel van Roon: None declared, Valerie Devauchelle-Pensec: None declared, Peter Gergely Shareholder of: Novartis Pharma, Employee of: Novartis Pharma, Xavier Mariette Consultant of: Bristol Myers Squibb, Galapagos, GlaxoSmithKline, Janssen, Novartis, Pfizer and UCB, Grant/research support from: Ose Pharmaceuticals, Raphaël Porcher: None declared
Article
Objectives: Sjӧgren's disease (SjD) is a systemic autoimmune disease characterized by focal lymphocytic infiltrate of salivary glands (SGs) and high SG IFNγ, both of which are associated with elevated lymphoma risk. IFNγ is also biologically relevant to mesenchymal stromal cells (MSCs), a SG resident cell with unique niche regenerative and immunoregulatory capacities. In contrast to the role of IFNγ in SjD, IFNγ promotes an anti-inflammatory MSC phenotype in other diseases. The objective of this study was to define the immunobiology of IFNγ-exposed SG-MSCs with and without the JAK1 & 2 inhibitor, ruxolitinib. Methods: SG-MSCs were isolated from SjD and controls human subjects. SG-MSCs were treated with 10 ng/ml IFNγ +/- 1000 nM ruxolitinib. Experimental methods included flow cytometry, RNA-sequencing, chemokine array, ELISA, and transwell chemotaxis experiments. Results: We found that IFNγ promoted expression of SG-MSC immunomodulatory markers, including HLA-DR, and this expression was inhibited by ruxolitinib. We confirmed the differential expression of CXCL9, CXCL10, CXCL11, CCL2, and CCL7, initially identified with RNA-sequencing. SG-MSCs promoted CD4+ T cell chemotaxis when pre-stimulated with IFNγ. Ruxolitinib blocks chemotaxis through inhibition of SG-MSC production of CXCL9, CXCL10, and CXCL11. Conclusions: These findings establish that ruxolitinib inhibits IFNγ-induced expression of SG-MSC immunomodulatory markers and chemokines. Ruxolitinib also reverses IFNγ-induced CD4+ T cell chemotaxis, through inhibition of CXCL9, -10, and -11. Because IFNγ is higher in SjD than control SGs, we have identified SG-MSCs as a plausible pathogenic cell type in SjD. We provide proof of concept supporting further study of ruxolitinib to treat SjD.
Article
Objectives: To investigate the utility of serum BAFF, IL-17, IL-18, IL-21, IL-22, CXCL13, TNF-R2 and PD-L2 as biomarkers of disease activity in primary Sjögren's syndrome (pSS), their relationship with lymphocyte subpopulations and their accuracy to discriminate pSS from Sicca syndrome. Methods: We conducted an observational study on 66 pSS patients and 48 controls (25 with Sicca syndrome and 23 healthy volunteers). Serum levels of BAFF, IL-17 A/F, IL-18, IL-21, IL-22, CXCL13, TNF-R2 and PD-L2 were measured using a multiplex immunoassay. Lymphocyte subpopulations were analysed by flow cytometry. Disease activity of pSS was assessed with ESSDAI at study inclusion. Results: Patients with pSS presented higher serum CXCL13 (364.7 vs. 205.2 pg/mL), IL-21 (43.2 vs. 0 pg/mL) and BAFF (1646 vs. 1369 pg/mL), and lower PD-L2 levels (1950.8 vs. 2792.3 pg/mL) than controls. ESSDAI was associated with BAFF, IL-18 and IL-22. Patients with ESSDAI >0 exhibited higher CXCL13, IL-21, IL-22 and TNFR2 concentrations. IL-21 levels correlated with lower memory B-cell and higher naïve B-cell percentages and IL-22 levels correlated with increased circulating activated CD4+ T-cells. The combination of serum CXCL13, BAFF and PDL2 levels using the formula [ln(CXCL13)+ln(BAFF)]/ln(PDL2) exhibit an AUC of 0.854 (95% CI: 0.750-0.919) to discriminate between pSS and Sicca syndrome (sensitivity 77.2% and specificity 86.4% using a cut-off of 1.7). Conclusions: CXCL13, BAFF, IL-21, and IL-22 are potential biomarkers of pSS activity and IL-21 and IL-22 are associated with disturbances of lymphocyte subpopulations in pSS. The combination of serum CXCL13, BAFF, and PD-L2 levels allows discrimination between pSS and Sicca syndrome.
Article
Background Sjögren's syndrome is an autoimmune disease characterised by dry eyes and mouth, systemic features, and reduced quality of life. There are no disease-modifying treatments. A new biologic, ianalumab (VAY736), with two modes of suppressing B cells, has previously shown preliminary efficacy. This dose-finding trial aimed to assess the safety and efficacy of different subcutaneous doses of ianalumab in patients with moderate to severe primary Sjögren's syndrome. Methods VAY736A2201 was a randomised, parallel, double-blind, placebo-controlled, phase 2b dose-finding study done in 56 centres in 19 countries. Patients aged 18–75 years with primary Sjögren's syndrome with moderate to severe disease activity (European Alliance of Associations for Rheumatology [EULAR] Sjögren's Syndrome Disease Activity Index [ESSDAI] score ≥6) and symptom severity (EULAR Sjögren's Syndrome Patient Reported Index score ≥5) were eligible. Participants were randomly assigned (1:1:1:1) to receive subcutaneous placebo or ianalumab (5 mg, 50 mg, or 300 mg) every 4 weeks for 24 weeks using a secure, online randomisation system. Randomisation was stratified by the ESSDAI score at baseline (≥10 or <10). Study personnel and patients were masked to treatment assignment. The primary outcome was the change in ESSDAI score from baseline to 24 weeks in all randomly assigned patients. Dose-related change in disease activity (ESSDAI) from baseline at week 24 was assessed by multiple comparison procedure with modelling analysis. Safety was measured in all patients who received at least one dose of study drug. This trial is registered with ClinicalTrials.gov, NCT02962895. Findings Between June 27, 2017, and Dec 06, 2018, 293 patients were screened, 190 of whom were randomly assigned (placebo n=49, ianalumab 5 mg n=47, ianalumab 50 mg n=47, ianalumab 300 mg n=47). Statistically significant dose-responses were seen for overall disease activity (ESSDAI score) in four of the five dose-response models tested (p<0·025 in four models, p=0·060 in one model). The ESSDAI score decreased from baseline in all ianalumab groups, with the maximal ESSDAI score change from baseline observed in the ianalumab 300 mg group: placebo-adjusted least-squares mean change from baseline −1·92 points (95% CI −4·15 to 0·32; p=0·092). There were four serious adverse events in three patients considered treatment-related (pneumonia [n=1] and gastroenteritis [n=1] in the placebo group; appendicitis plus tubo-ovarian abscess in the same patient in the ianalumab 50 mg group). Interpretation The study met its primary objective, showing a dose-related decrease in disease activity as measured by ESSDAI at week 24. Overall, ianalumab was well tolerated and safe, with no increase in infections. To our knowledge, this is the first large, randomised, controlled trial in primary Sjögren's syndrome that met its primary endpoint, and its results mean there is potential for more studies of this mechanism in the future. Funding Novartis.