You are on page 1of 20

PRIMER

Sjögren syndrome
Pilar Brito-Zerón1–3, Chiara Baldini4, Hendrika Bootsma5, Simon J. Bowman6,
Roland Jonsson7,8, Xavier Mariette9,10, Kathy Sivils11, Elke Theander12,
Athanasios Tzioufas13 and Manuel Ramos-Casals2,3,14
Abstract | Sjögren syndrome (SjS) is a systemic autoimmune disease that primarily affects the exocrine
glands (mainly the salivary and lacrimal glands) and results in the severe dryness of mucosal surfaces,
principally in the mouth and eyes. This disease predominantly affects middle-aged women, but can
also be observed in children, men and the elderly. The clinical presentation of SjS is heterogeneous
and can vary from sicca symptoms to systemic disease (characterized by peri-epithelial lymphocytic
infiltration of the affected tissue or the deposition of the immune complex) and lymphoma.
The mechanism underlying the development of SjS is the destruction of the epithelium of the exocrine
glands, as a consequence of abnormal B cell and T cell responses to the autoantigens Ro/SSA and
La/SSB, among others. Diagnostic criteria for SjS include the detection of autoantibodies in patient
serum and histological analysis of biopsied salivary gland tissue. Therapeutic approaches for SjS
include both topical and systemic treatments to manage the sicca and systemic symptoms of disease.
SjS is a serious disease with excess mortality, mainly related to the systemic involvement of disease
and the development of lymphomas in some patients. Knowledge of SjS has progressed substantially,
but this disease is still characterized by sicca symptoms, the systemic involvement of disease,
lymphocytic infiltration to exocrine glands, the presence of anti-Ro/SSA and anti-La/SSB
autoantibodies and the increased risk of lymphoma in patients with SjS.

Sjögren syndrome (SjS) is a systemic autoimmune dis­ In the past 50 years, the term ‘secondary SjS’ has
ease with a specific predisposition for causing inflam­ been used in the research setting for patients diag­
mation of the exocrine glands, predominantly the nosed with SjS in combination with another systemic
salivary and lacrimal glands but also including the nose, autoimmune disease and ‘primary SjS’ for those whom
upper respiratory tract, oropharynx and, in women, the SjS is the only diagnosis. However, this terminology is
vagina. The main consequence of this inflammation is under debate, and standardized guidelines on when or
the develop­ment of sicca symptoms, such as dryness why they should be used are lacking. ‘Secondary SjS’
of the mucosal surfaces, principally in the mouth and is mainly used in patients with concomitant systemic
eyes1. Hendrik Sjögren was the first to establish that autoimmune diseases (such as rheumatoid arthritis,
typical sicca symptoms extended beyond glandular systemic sclerosis or systemic lupus erythematosus
­involvement and termed the sicca syndrome SjS. (SLE)) and is rarely used in patients with concomitant
The autoimmune aetiopathogenetic basis of SjS was organ-specific autoimmune diseases (for example,
confirmed in the 1960s and the presence of the auto­ ­autoimmune thyroiditis, primary biliary cholangitis
antibodies SjS-related antigen A (SSA; also known or autoimmune hepatitis). In addition, some authors
as anti-Ro/SSA antibodies) and SSB (also known as have used ‘secondary SjS’ for patients with clinical dry­
anti-­La/SSB antibodies), in addition to organ-specific ness who do not fulfil the criteria of SjS, whereas others
lymphocyte infiltration (for example, focal lymphocytic have proposed the use of different terms, such as ‘associ­
sialadenitis (FLS) or inflammation of the salivary glands) ated SjS’ for patients with SLE and ‘secondary SjS’ for
Correspondence to M.R.‑C. became central for the pathobiology and diagnosis of those with rheumatoid arthritis2. In fact, the distinc­
Department of Autoimmune this disease (FIG. 1). Since then, knowledge of SjS has pro­ tion between primary and secondary (or associated) SjS
Diseases, ICMiD, Hospital gressed substantially, but the five features of this disease reflects only the frequently reported clinical situation of
Clínic, C/Villarroel, 170,
that were initially defined in the early years (the develop­ the coexistence or overlap of SjS in patients with other
08036 Barcelona, Spain.
mramos@clinic.ub.es ment of sicca symptoms, systemic involvement of disease, autoimmune diseases1. With respect to patient manage­
lymphocytic infiltration to exocrine glands, the presence ment, dividing patients into ‘primary’ or ‘secondary’ SjS
Article number: 16047
doi:10.1038/nrdp.2016.47 of autoantibodies and the increased risk of ­lymphoma in makes no sense as the management of SjS is the same in
Published online 7 July 2016 patients with SjS) still characterize this disease. both patient populations.

NATURE REVIEWS | DISEASE PRIMERS VOLUME 2 | 2016 | 1


© 2016 Macmillan Publishers Limited. All rights reserved
PRIMER

Author addresses At the presentation of disease, the epidemiological


profile of SjS is very typical, which might aid early diag­
1
Autoimmune Diseases Unit, Department of Medicine, Hospital CIMA- Sanitas, nosis. First, SjS predominantly affects women. In fact, SjS
Barcelona, Spain. has the most unbalanced gender ratio out of all systemic
2
Sjögren Syndrome Research Group (AGAUR), Laboratory of Autoimmune Diseases Josep autoimmune diseases; close to a 10/1 female/male ratio
Font, IDIBAPS-CELLEX, Barcelona, Spain.
was reported in a big data study of >14,000 patients with
3
Department of Autoimmune Diseases, ICMiD, Hospital Clínic, C/Villarroel, 170,
08036 Barcelona, Spain. SjS9. Moreover, the phenotypic expression of SjS differs
4
Rheumatology Unit, University of Pisa, Pisa, Italy. in men and women; men present with severe ocular
5
Department of Rheumatology and Clinical Immunology, University of Groningen, involvement and less-pronounced systemic and immuno­
University Medical Center Groningen, Groningen, The Netherlands. logical involvement of disease compared with women10,11.
6
Rheumatology Department, University Hospitals Birmingham NHS Foundation Trust, Second, although SjS can occur at all ages, it is mainly
Birmingham, UK. diagnosed between 30 and 50 years of age. SjS is rare in
7
Broegelmann Research Laboratory, Department of Clinical Science, University of children and the female/male ratio is less evident in chil­
Bergen, Bergen, Norway. dren than in adults12. Enlargement of the parotid glands,
8
Department of Rheumatology, Haukeland University Hospital, Bergen, Norway. which is not one of the typical sicca symptoms, is the
9
Université Paris Sud, INSERM, Paris, France.
predominant feature observed at presentation in children
10
Center for Immunology of Viral Infections and Autoimmune Diseases, Assistance
Publique — Hôpitaux de Paris, Hôpitaux Universitaires Paris-Sud, Le Kremlin-Bicêtre, with SjS12, although salivary gland biopsy is just as useful
Paris, France. for SjS diagnosis in children as in adults13. Moreover, the
11
Oklahoma Sjögren’s syndrome Center of Research Translation, Oklahoma Medical most frequently reported autoantibody is anti-Ro/SSA
Research Foundation, Oklahoma City, Oklahoma, USA. antibody, but not antinuclear antibody (ANA), which
12
Department of Rheumatology, Malmö University Hospital, Lund University, Lund, Sweden. is the most common antibody in adults12. By contrast,
13
Department of Pathophysiology, School of Medicine, National University of Athens, parotid gland involvement and anti-Ro/SSA antibodies
Athens, Greece. are observed less frequently in older patients >70 years
14
Department of Medicine, University of Barcelona, Barcelona, Spain. of age than in those <70 years of age, and elderly patients
also have a higher frequency of pulmonary involvement11.
Scientific reports have mainly focused on primary SjS and other autoimmune diseases frequently
disease (thus avoiding the confounding effect of an ­coincide (associated SjS) in daily practice. In patients with
additional autoimmune disease) to provide a more systemic autoimmune diseases, the proportion of patients
homogeneous characterization of SjS, especially after with concomitant SjS differs between conditions; 14–18%
the introduction of the first set of worldwide classifica­ of patients with SLE14,15, 7–17% with rheumatoid arthri­
tion criteria in 1993 (REF. 3). Thus, the term SjS has been tis16,17 and 12% with systemic sclero­sis also have associ­
used throughout this Primer to refer to primary SjS and ated SjS18. In clinical practice, the manage­ment of
the term associated SjS used for studies that included associated SjS should be the same as the ­management
patients with concomitant autoimmune diseases. This of primary SjS.
Primer describes the epidemiology and aetiopatho­ SjS can be a serious disease with excess mortality.
genetic mechanisms of SjS, the global clinical picture The main causes of death traditionally associated with
including both glandular and systemic manifestations, excess mortality in patients with SjS are B cell lym­
the approaches to diagnosis and treatment, patient qual­ phoma, severe organ-specific features (principally,
ity of life and concludes by presenting an outlook on the interstitial lung disease, renal failure and severe cryo­
potential advances in the understanding of SjS. globulinaemic v­ asculitis), in addition to infections and
cardiovascular disease19.
Epidemiology
The reported incidence and prevalence of SjS varies Mechanisms/pathophysiology
according to both the study design and the classifica­ Multiple environmental factors that interact with an
tion criteria used4. The pooled prevalence rates in studies individual’s genetic susceptibility can influence SjS
that used the 1993 European Classification Criteria was develop­ment. Increased levels of cytokines and chemo­
12‑fold higher than the pooled prevalence rates in stud­ kines as a result of abnormal responses of both T cells
ies that used the 2002 American–European Consensus and B cells to autoantigens, such as the ribonucleo­
Group (AECG) criteria, whereas the pooled prevalence protein particles Ro/SSA and La/SSB expressed by the
rates reported in population-based epidemiological epithelium of the exocrine glands (such as the salivary
studies were slightly lower than that calculated in the and lacrimal glands), lead to chronic inflammation
total population4. The incidence of SjS ranges between of the exocrine glands and the eventual loss of their
3 and 11 cases per 100,000 individuals5,6, whereas the physio­logical functions20. Salivary gland epithelial cells
prevalence ranges between 0.01% and 0.72%7,8. It is also (SGECs) in patients with SjS drive and regulate local
likely that there are asymptomatic cases, which never autoimmune responses by actively mediating the accu­
become diagnosed. Only one study has evaluated the mulation, activation and differentiation of immune
influence of race or ethnicity on the prevalence of SjS. cells. Immune cells and the inflammatory micro­
This study found a significant twofold higher prevalence environment further activate epithelial cells or regulate
among individuals with a non-European background their survival, thus creating a vicious cycle of epithelial
than in those with a European background in the general cell and immune cell interaction that perpetuates the
population of the Greater Paris area7. ­autoimmune responses observed in SjS21.

2 | 2016 | VOLUME 2 www.nature.com/nrdp


© 2016 Macmillan Publishers Limited. All rights reserved
PRIMER

Aetiopathogenetic mechanisms TNF ligand superfamily member 6 (also known as FAS


The current theory explaining the pathogenesis of SjS — ligand), pro-inflammatory cytokines, cytokines involved
termed ‘autoimmune epithelitis’ in this context — sug­ in lymphoid cell differentiation and many chemokines
gests that the epithelium is the inflamed tissue in SjS22. that attract T cells and form the germinal centre (that
The exocrinopathy and involvement of functional is, sites in secondary lymphoid organs where B cells
parts of an organ (parenchyma) are associated with mature)20,25. Expression of these molecules in SGECs is
the development of lymphocytic infiltrates in epithe­ upregulated further by cytokines secreted by immune
lial cells surrounding or invading organs. According to cells, which infiltrate tissue lesions, suggesting abnor­
this theory, epithelial cells are thus central regulators of mal interactions between SGECs and immune cells in
the auto­immune response by acting as atypical antigen-­ SjS. SGECs have been shown to activate CD4+ T cells
presenting cells and are not just innocent bystanders in vitro and mediate their differentiation into follicular
affected by infiltrating immune cells (FIG. 2). However, helper T cells27, which in turn enhances the survival
auto­immune epithelitis is only one explanation for the of B cells20,25,28. In addition, SGECs have been shown
immuno­pathology of SjS, although it is the theory with to directly interact with B cells and can promote their
the most support. Several studies have also suggested a differentiation towards B cell subtypes, similar to those
role for a neuroendocrine mechanism that is related to observed in salivary gland lesions of patients with SjS29.
the influence of hormones and neuropeptides in the func­ The constitutive expression of most immune-­
tion of the exocrine glands23 that could explain why some competent molecules was significantly increased and
patients with SjS present with severe sicca symptoms with stable in SGEC cell lines derived from patients with SjS
no (or limited) inflammatory histopathological features24. compared with cells from healthy controls25. In addition,
To date, numerous studies have investigated this theory in SGECs of patients with SjS, high MYC (a regulatory
of autoimmune epithelitis, by examining affected SGECs gene involved in cell proliferation) expression was found
and epithelial cells of the lacrimal glands. These studies compared with controls, and translocation of Ro/SSA
have shown that epithelial cells are able to o ­ rchestrate and La/SSB autoantigens to the cell membrane was
innate and acquired immune responses in SjS25,26. observed in the cytoplasm via histological examination
by in situ hybridization. These data indicate intrinsic
Immune activation in epithelia. SGECs have immune activation in SGECs of patients with SjS25. The
been shown to constitutively express a plethora of cause of the activation of immune responses in the epi­
immune-competent molecules that are implicated in thelial cells is currently unknown; epigenetic changes or
lymphoid cell recruitment, homing, activation, differ­ latent viral infections might have a role (see below)20.
entiation and proliferation, and immune cell expansion Histopathological studies have revealed that glandular
and organization20,25 (FIG. 2). These molecules include epithelial cells express molecules that are indicative of
human leukocyte antigen (HLA) class I molecules, both a type I interferon (IFN) signature (predominantly
tumour necrosis factor (TNF) receptor superfamily IFNβ) and a type II IFN signature (predominately IFNγ),
member 5 (also known as CD40; a co-stimulatory pro­ whereas immune cells express molecules that are typical
tein involved in antigen presentation), adhesion mol­ for a type II IFN signature (IFNγ)30,31. The IFN signature
ecules, the apoptosis-related molecules TNF receptor in salivary gland lesions seen in patients with SjS and the
superfamily member 6 (also known as FAS receptor) and high constitutive Toll-like receptor 3 (TLR3) expression

Discovery of Use of the term ‘sicca syndrome’ Use of antimalarials in SjS Evaluation of Increased Diagnosis
filamentary keratitis for oral, ocular and vaginal dryness xerostomia risk of Clinical
(strands of (dry mouth) lymphoma presentation
degenerated Use of rose bengal dye for Salivary flow by salivary in patients Treatment
epithelial cells on the the assessment of ocular rates used for scintigraphy with SjS
corneal surface) damage in patients with SjS SjS diagnosis

1882 1903 1925 1933 1951 1954 1959 1964 1967 1968 1971 1975 1978 1982

Labial salivary gland


First use of Use of cortisone in SjS biopsy Identification Detection of
lacrimal blotting Association of the SjS- anti-Ro/SSA
paper test for ‘About keratoconjunctivitis sicca’ thesis between Lymphocytic sialadenitis precipitating and anti-La/SSB
detection of showing the association between sicca SjS and observed in oral tissue antibodies antibodies in
dry eyes syndrome and systemic features lymphoma of patients with SjS A, B and C patients with SjS

Figure 1 | Milestones in the history of SjS compressed in 100 years the mid‑twentieth century confirmed the association
Nature Reviewsof| Disease
sicca syndrome
Primers
(1882–1982). Patients affected by isolated sicca symptoms were first with other rheumatic and autoimmune diseases and with an unusually high
reported at the end of the nineteenth century and symptoms were frequency of haematological malignancies. The autoimmune
integrated into a ‘sicca syndrome’ by Gougerot in 1925. However, the first aetiopathogenetic basis of Sjögren syndrome (SjS) was confirmed in the
to suggest they could be attributed to a systemic disease was the Swedish 1960s, and the presence of autoantibodies (Ro/SSA and La/SSB) and
ophthalmologist Henrik Sjögren, who, in 1933, established that the disease organ-specific lymphocytic infiltration (focal lymphocytic sialadenitis)
spectrum extended beyond glandular involvement. Subsequent studies in become central for the pathobiology and diagnosis of the disease.

NATURE REVIEWS | DISEASE PRIMERS VOLUME 2 | 2016 | 3


© 2016 Macmillan Publishers Limited. All rights reserved
PRIMER

in SGECs and responsiveness to TLR3 signalling by Cytokines such as IL‑7 that drive effector T cells are
SGECs further support the implication of viral infection overexpressed by SGECs of patients with SjS33. Innate
in SjS pathogenesis. immune signalling via TLR activation, such as that
Immune activation in SjS can be triggered by sentinel elicited by viral infections, might induce IL‑7 produc­
cells, such as dendritic cells and plasmacytoid dendritic tion by SGECs, dendritic cells or endothelial cells33,34.
cells, as evidenced by experimental models21. The robust IL‑7 can drive effector T cells to induce sialoadenitis in
genetic association between HLA class I and class II mol­ mice35. Other cytokines that are overexpressed in the
ecules and SjS pathogenesis (see below) strongly suggests salivary glands of patients with SjS (such as IL‑7, IL‑17,
the involvement of T cells. In accordance, T cells that IL‑21, IL‑22 and IL‑23) have been shown to contribute
secrete IFNy and IL‑17 have been detected in inflamed to the abnormal T cell and B cell response and might
salivary glands of patients with SjS and are associated play a crucial role in SjS, and can increase the risk of
with tissue damage32,33. lymphoid neogenesis21,26,36.

Mucolytics Mucus changes Dry surfaces Increased Eyes: topical


chance of corticosteroids
infection and antibiotics
Epithelial cell
of the salivary or Mouth: good
lacrimal glands Eyes: eye drops and gels oral health
Mouth: saliva substitutes and fluorides
Ro/SSA Secretagogues Gland hypofunction
and La/SSB
autoantigens

Surface
expression of Apoptosis
autoantigens Autoantigen-
containing
vesicle

TLR Tissue damage

Autoantibodies

Cytokines and chemokines CD4+ IL-17


(type I IFN signature) T cell IFNγ

Plasma
IL-7 cell
IL-17

Dendritic
cell IL-6 BAFF
IL-12
IL-21 IL-12 TH1 cell IL-12
IL-6
IL-23 B cell
TFH cell IL-6 B cell depletion
IL-17 therapy
IL-21
TH17 cell IL-22
IL-7
BAFF

BAFF

Germinal centre
formation

BAFF

Figure 2 | The pathogenesis of autoimmune epitelitis as a potential explanation for SjS. Immune-competent
Nature Reviews | Disease Primers
molecules, such as Toll-like receptors (TLRs) are constitutively expressed by salivary gland epithelial cells (SGECs).
Activation of TLR signalling in gland epithelium causes the production of autoantigens, the upregulation of
immune-competent molecules (chemokines and cytokines), apoptosis and epithelial hypofunction. Autoantigens can be
released from SGECs and presented to immune cells. CD4+ T cells differentiate into follicular helper T (TFH) cells, which
increase B cell survival. Interaction between SGECs and B cells promotes B cell differentiation. Potential treatments are
highlighted in green boxes. BAFF, B cell-activating factor; IFN, interferon; TH, T helper.

4 | 2016 | VOLUME 2 www.nature.com/nrdp


© 2016 Macmillan Publishers Limited. All rights reserved
PRIMER

TLR signalling. SGECs, via TLRs, can also sense innate In Mediterranean countries, patients with SjS have
immunity signals, leading to the upregulation of the been reported to have a higher frequency (14%) of
previously mentioned immune-competent molecules chronic hepatitis C virus (HCV) infections than healthy
(including HLA class I molecules, FAS receptor, FAS individuals47. Histological analysis of salivary gland
ligand and pro-inflammatory cytokines) in SGECs biopsies revealed a higher frequency of FLS in patients
(FIG. 2); this mechanism links the innate and adaptive with SjS who are infected with HCV (57%) than in
immune responses 20,25. Furthermore, TLR3 signal­ patients with SjS who are not infected with HCV (5%)48.
ling in SGECs leads to hypofunction of the salivary In a separate study, HCV infection was identified in
glands, SGEC apoptosis and upregulation of Ro/SSA 13% of a cohort of Spanish patients with SjS. In these
and La/SSB autoantigens; the increase in the levels of patients, the autoimmune response was characterized
autoantigens suggests that TLR3 signalling in SGECs by an abnormal predominance of anti-La/SSB, over
might also be implicated in autoantigen presenta­ anti-Ro/SSA, autoantibodies compared with patients
tion20,25,30. Finally, SGECs might also mediate the expo­ with SjS in the absence of HCV infection49.
sure and presentation of intracellular ribonucleoproteins Lymphotropic viruses have also been associated with
(such as Ro/SSA and La/SSB) to the immune system by the development of autoimmune diseases. Some viral
the release of autoantigen-containing vesicles, such as domains of EBV, HHV6 and human immunodeficiency
exosomes from healthy SGECs or apoptotic blebs from virus 1 have molecular similarities to some antigenic
apoptotic SGECs20,25. epitopes on the La/SSB protein50. These viral domains
are referred to as ‘self-determinants’, and viral infections
Apoptosis. Apoptosis of SGECs has been proposed as a can induce the translocation of these viral domains to
potential aetiopathogenetic mechanism that is associ­ the cell surface and could promote the induction of
ated with the impairment of secretory glandular func­ autoantibodies. Owing to the increased understand­
tion37. The importance of epithelial cell apoptosis in ing of the genetic complexity and the central role of
the pathogenesis of SjS is supported by data obtained type I and type II IFNs in systemic autoimmunity 51, the
from an experimental mouse model. In these mice, emphasis is now on how viruses trigger autoimmunity
silencing the expression of nuclear factor-κB (NF‑κB) and specifically on the role of interactions of viruses
inhibitor-ζ in epithelial cells of the lacrimal gland of with pattern-recognition receptors, such as TLRs. This
the eye led to increased apoptosis of epithelial cells and concept is also highly relevant to the emerging impor­
SjS-like inflammatory lesion in the lacrimal glands. tance of gene–environment inter­actions in systemic
These features were associated with high titres of serum auto­immunity. However, viruses might trigger auto­
anti-Ro/SSA and anti-La/SSB antibodies and could be immunity long before the manifestation of disease, and
reversed by treating the mice with apoptosis-blocking auto­antibodies have been found in patients for up to
caspase inhibitors38. decades before the diagnosis of SjS52,53.

Environmental factors Smoking. Among other environmental factors, ­smoking


Viral infections. Viral infections are considered one of has emerged as a risk factor for SjS and other auto­
the key aetiological factors involved in SjS, as tissue of immune disorders, such as SLE and myositis54. Some
the salivary glands are sites of latent viral infections39. studies have reported a lower frequency of smoking
The list of viruses that can be involved in SjS is long and among patients with SjS than control individuals55,56, and
includes members of the Herpesviridae family, such as other studies have linked cigarette smoking with a higher
Epstein–Barr virus (EBV) and human herpesvirus 6 ­frequency of ANAs57 and a lower frequency of FLS58.
(HHV6). The potential causative role of EBV and HHV6
infections in SjS has been studied, but establishing a Genetic factors
causative relationship is difficult because of the high The genetic basis of SjS is expected to be as complex as
prevalence of these viruses in the healthy population40. that of other autoimmune diseases including SLE and
Immune responses to EBV (EBV-specific IgG) have been rheumatoid arthritis, each of which have >100 estab­
demonstrated in patients with SjS41. A higher prevalence lished genetic associations. However, compared with
of HHV6‑specific antibodies has been detected in the SLE and rheumatoid arthritis, the genetic architecture
serum of patients with SjS (in 36% of patients) than in of SjS remains relatively unexplored. Familial clustering
control individuals (in 10% of controls)42. However, studies show that approximately 35% of patients with
other studies could not confirm this finding 43,44. SjS have relatives with SjS or a related auto­immune
Retroviruses can infect cells of the immune sys­ disease59. Moreover, having a twin sibling with SjS is
tem and can cause aberrant antiretroviral responses associated with a 662‑fold higher risk of develop­
that could lead to autoimmunity. High serum titres of ing this disease than individuals without an affected
human T cell lymphotropic virus 1 (HTLV1)-specific twin sibling60.
antibodies and the presence of salivary IgA antibodies Genome-wide association studies have shown
against HTLV1 was reported in patients with SjS in strong associations between SjS and the HLA class I,
areas of Japan that are endemic for HTLV1, more so class II and class III regions, with multiple associations
than in individuals who do not have SjS45. In addition observed for HLA class II molecules, HLA-antigen D
to its effect in autoimmunity, HTLV1 might directly related (HLA‑DR) and HLA‑DQ loci61–63. Some HLA
infect SGECs46. class II alleles are associated with the production of

NATURE REVIEWS | DISEASE PRIMERS VOLUME 2 | 2016 | 5


© 2016 Macmillan Publishers Limited. All rights reserved
PRIMER

autoantibodies in SjS but not with other clinical features promote lymphomagenesis. For example, in patients
of the disease63. By integrating genetic and transcrip­ with SjS, a predictive risk factor for the development of
tional data, associations expression quantitative trait lymphoma is the presence of germinal centre-like struc­
loci were identified, for example, five HLA class I and tures within the mucosal sites of exocrine glands (where
class II loci (HLA‑A, HLA‑C, HLA‑DRB6, HLA‑DPB1 they usually are not present)67. Interestingly, the levels
and HLA‑DQA1)64. These results suggest that HLA risk of CXC-chemokine ligand 13 (CXCL13), a chemokine
alleles could not only affect HLA function (including that attracts memory B cells within germinal centres,
peptide binding and antigen presentation) but could have been found to be increased in patients with SjS and
also influence the expression levels of HLA class I and lymphoma compared with patients with SjS who do not
class II molecules. have lymphoma68. Two recent studies have demonstrated
Other loci involved in a wide variety of innate and that the levels of TNF ligand superfamily member 13B
adaptive immune processes have also been shown to be (also called B cell-activating factor (BAFF); a protein
substantially associated with SjS. Examples are genes involved in B cell survival, activation and differenti­
encoding molecules involved in type I and type II IFN ation) are increased in patients with SjS with lymphoma
signalling and target processes (such as IRF5, IL12A compared with those without 69,70. Polymorphisms in
and STAT4), NF-κB signalling (for example, TNIP1 and TNFSF13B and TNFRSF13C have been found to be
TNFAIP3), lymphocyte trafficking (such as CXCR5), associated with lymphoma develop­ment in patients with
and activation and differentiation of antibody-producing SjS71. FMS-like tyrosine kinase 3 (FLT3) is a cytokine
cells (such as BLK)61. related to the differentiation and proliferation of early
Very little is known about how these SjS-associated B cell progenitors, and increased serum levels of FLT3
genetic variants change normal biological activity or have been found in patients with SjS and lymphoma
how they are involved in disease. However, expression compared with healthy individuals72.
quantitative trait loci were also identified in IL12A, BLK
and TNIP1 alleles, suggesting that these variants might Diagnosis, screening, and prevention
act by influencing the expression levels of the associated Clinical presentation
proteins61. In addition, the precise subsets of immune SjS is a slowly progressing chronic autoimmune disease.
cells in which these variants cause pathogenetic effects Clinical manifestations are extremely variable; the pres­
are unclear. The expression of BLK is relatively limited entation of this disease can range from an organ-specific
to B cells, but the expression of all the other associated autoimmune disease to a systemic autoimmune disease
genes have been shown in multiple subtypes, such as and a lymphoproliferative condition.
natural killer cells, monocytes, dendritic cells, as well as
T cells and B cells61. Understanding these genetic effects Sicca symptoms. Dryness of the eyes and mouth are
and the consequences of these genetic risk variants in the classic symptoms associated with SjS and are
the function of the immune system are important for present in the vast majority of patients. Patients with
determining causal disease mechanisms in SjS. dry mouth usually complain of the inability to swal­
low dry food without fluid intake, changes in their
Lymphoma development sense of taste, burning mouth, difficulties in speak­
Patients with SjS are at increased risk of developing lym­ ing continuously and the need for waking during the
phoma65. Compared with other autoimmune diseases, night to have a drink of water 73. Physical examination
SjS has the highest risk of lymphoma development than ­usually reveals a dry, erythematous (red skin), sticky
the general population, with a standardized incidence oral mucosa, a dry fissured tongue and an accelerated
ratio of 9–44 in studies before 2000 and 5–15 in stud­ ­dental caries73. Dental caries in SjS is usually found in
ies after 2000 (REF. 65). Marginal zone lymphomas and a specific pattern, with damage commonly observed to
especially mucosa-associated lymphoid tissue (MALT) the cervical region and/or smooth surface of the teeth
lymphomas are the most common histological subtype (regions that are usually rather resistant to caries owing
of lymphoma observed in patients with SjS. to the continuous flow of saliva over these surfaces)73.
In patients with SjS, lymphomagenesis is a multi­ Approximately one-third of patients complain of fre­
step aetiopathogenetic process65. The first step is the quent swelling of the parotid glands74. Parotid swell­
chronic stimulation of polyclonal B cells, especially in ing is commonly seen in relatively young patients75.
patients in whom the autoantibody rheumatoid factor Eye symptoms include eyes that feel sandy and itchy,
is detected. Activation of polyclonal B cells might be and that might be red in appearance and might pres­
associated with an increased risk of oncogenic muta­ ent with dilatation of the bulbar conjunctival vessels.
tions and mono­clonal selection. A dysfunction in one of Furthermore, patients with SjS report an inability to
the checkpoints of the activation of autoimmune B cells wear contact lenses and the need for use of tear sub­
could speed up the development of a haematological stitutes. In addition to the salivary and lacrimal glands,
malignancy as the last step in the process. It was recently other exocrine glands might be affected in patients
found that functional germinal mutations in TNFAIP3, with SjS. Dryness can also affect the nose, the upper
which controls NF‑κB activation, were associated with respir­atory tract and the oro­pharynx, which can cause
SjS-related lymphomas66. rhinitis sicca, persistent cough and hoarseness1. Many
Two of the main features of SjS — chronic expo­ patients also experience xerosis cutis (abnormally dry
sure to autoantigens and immune activation — might skin) and, in women, vaginal dryness76,77.

6 | 2016 | VOLUME 2 www.nature.com/nrdp


© 2016 Macmillan Publishers Limited. All rights reserved
PRIMER

General symptoms and organ-specific complications. immune complex-mediated membranous or mem­


SjS can also be associated with systemic symptoms branoproliferative glomerulonephritis88. Some renal
(BOX 1), which can be seen in approximately 50–60% of complications of SjS, such as distal renal tubular acido­
patients19,69. Severe clinical manifestations have been sis might ­compromise renal function, but might also be
described in 15–20% of patients with SjS, whereas the clinically silent.
prevalence of lymphoma has been estimated at around Complications of SjS can affect both the peripheral
5% 78. Low levels of complement C3 and comple­ and the central nervous systems (CNS). Approximately
ment C4, hypergammaglobulinaemia or the presence 10% of patients with SjS are also diagnosed with periph­
of cryo­globulins, rheumatoid factor or anti-Ro/SSA eral neuropathies89, of which sensory neuropathy is the
auto­antibodes can be used to identify patients with a most commonly reported. The frequency of CNS involve­
potentially more-severe clinical expression of SjS that ment in patients with SjS is approximated at 3% of causes
might merit closer follow‑up over time. Several of these using the ESSDAI definitions84 and can include myelitis,
markers (such as complement C4, rheumatoid factor and aseptic meningitis and cerebral lesions that might mimic
cryoglobulins) are included in the current classification those observed in multiple sclerosis90 (FIG. 3e).
criteria of cryoglobulinaemic vasculitis79, and one study Other complications of SjS include cytopaenia
found that patients with SjS who fulfilled these criteria (in approximately one-third of patients with SjS), Raynaud
at diagnosis are at higher risk of death80. phenomenon (FIG. 3c) , serositis and p ­ rotein-losing
Systemic symptoms might develop either at the enteropathy, among others90.
presen­tation of SjS or later. When systemic symptoms
appear before the onset of the characteristic symptoms of Lymphoma. Diffuse large B cell lymphoma is often
SjS or when the typical symptoms are mild, this disease formed as a consequence of the progression of a low-
is referred to as occult SjS or non-sicca onset of SjS74. grade lymphoma65. The salivary glands, primarily the
According to the European League Against Rheumatism parotid glands, are the most frequent site of MALT lym­
(EULAR) SjS Disease Activity Index (ESSDAI; disease phomas in patients with SjS (FIG. 3f), but lymphomas can
score ranging from 0 to 123)81, systemic features of SjS also occur in other mucosal sites, such as the stomach,
can be described by recognizing various organ domains the lungs and the eyes. The most frequent presentation
at different levels of disease activity. of lymphoma is persistent parotid enlargement, which
General symptoms of SjS include fever, weight can be difficult to differentiate from benign parotid
loss and fatigue, and are common presenting features enlargement. Some clinical findings might be useful in
of this disease and are observed in seropositive and differentiating between parotid enlargement and lym­
sero­negative patients for both anti-Ro/SSA and anti- phoma, for example, benign parotid enlargement is
La/SSB antibodies. However, functional impairment often bilateral and the swelling fluctuates in size, whereas
related to general symptoms seems to be greater in lymphoma is more often unilateral, swelling is fixed
seronegative patients82,83. and sometimes hard. Ultrasonography of the salivary
The systemic symptoms and complications of SjS glands (including Doppler ultrasonography) and MRI
can be widespread, involving most systems of the body, might also aid the differentiation between benign and
including the cutaneous, articular, pulmonary, cardio­ m
­ alignant parotid enlargement
vascular, nephro-urological, nervous and haematological The main clinical predictors of lymphoma are per­
systems, among others (BOX 1; FIG. 3). sistent swelling of the salivary glands, lymphadenopathy
Synovitis or arthritis is common in patients with and palpable purpura (overwhelmingly related to cryo­
SjS84 (FIG. 3a), but causes less joint erosion and is more globulinaemia) (BOX 2). These predisposing factors are
relapsing and remitting than in rheumatoid arthri­ easily checked in clinical practice and should be included
tis84. Subclinical synovitis is frequently observed by in the management of patients with SjS. Moderate
­musculoskeletal ultrasonography in patients with SjS85. (an ESSDAI score of 5–14) and, especially, high dis­
Skin involvements of disease include cutaneous ease activity (an ESSDAI score of ≥14) is predictive of
vasculitis84, which can include various lesion subtypes, ­lymphoma development in patients with SjS91,92.
predominantly flat purpura, observed in patients
with hypergammaglobulinaemia and palpable pur­ Diagnostic tests
pura, which are associated with cryoglobulinaemic Sicca symptoms. Diagnostic tests to determine sicca
v­ asculitis86 (FIG. 3b). symptoms try to measure glandular dysfunction and
Involvement of the pulmonary system is c­ ommon quantify the degree of involvement of the main glands
in SjS and is observed in 16% of patients 84, but is affected (that is, the major salivary and lacrimal
rarely clinically relevant. Patients with SjS are at risk glands) (TABLE 1). The two main oral tests to assess
of develop­ing interstitial lung diseases (FIG. 3d) and are ­salivary gland function are the measurement of salivary
more likely to have rheumatoid factor, anti-Ro/SSA and flow rates and salivary gland scintigraphy 93. The main
anti-La/SSB autoantibodies and lymphopaenia than ocular tests include Schirmer’s tests and analysis of the
patients with SjS with no lung involvement of disease87. corneal surface using dyes (fluorescein and lissamine
The kidneys are potentially targeted by two dis­ green) that stain degenerated or dead cells (­corneal
tinct immunopathological pathways in patients with stainings)94. Ultrasonography and MRI are mainly used
SjS: infiltration of the tubular epithelium by activated to evalu­ate the most common complications of SjS,
lymphocytes, resulting in interstitial nephritis, and such as i­ nfections and lymphoma95.

NATURE REVIEWS | DISEASE PRIMERS VOLUME 2 | 2016 | 7


© 2016 Macmillan Publishers Limited. All rights reserved
PRIMER

Box 1 | Organ‑by‑organ systemic manifestations


Oral symptoms Pancreatic complications
• Hyposalivation • Recurrent acute pancreatitis
• Soreness
Nephro-urological complications
• Adherence of food to the mucosa
• Renal tubular acidosis (9% of patients)
• Dysphagia
• Glomerulonephritis (4% of patients)*
• Difficulties in speaking or eating
• Interstitial cystitis (in the absence of bacterial
• Dental caries infection)
• Oral candidiasis • Osteomalacia
Ocular symptoms • Recurrent renal colic due to renal stones
• Insufficient tears • Hypokalaemic paralysis*
• Inability to tear
Peripheral nervous system complications
• Foreign-body sensation
• Mixed polyneuropathy
• Conjunctival inflammation (keratoconjunctivitis sicca)
• Axon sensory polyneuropathy
• Eye fatigue
• Sensory ataxic neuronopathy*
• Decreased visual acuity
• Blepharitis • Axon sensorimotor polyneuropathy
• Bacterial keratitis • Trigeminal or other cranial neuropathies
• Demyelinating polyradiculoneuropathy
General symptoms
• Autonomic neuropathy
• Fatigue
• Pure sensory neuronopathy
• Chronic pain
• Low-grade fever • Mononeuritis multiplex*
• Weight loss • Small-fibre neuropathy (painful paresthesias)

Lymph node complications Central nervous system complications


• Reactive multiple lymphadenopathy (swelling of the • White matter lesions
lymph nodes) (multiple sclerosis‑like disease)*
• Lymphoproliferative complications • Neuromyelitis optica spectrum disorder*
Cutaneous complications • Recurrent aseptic meningitis
• Cutaneous vasculitis (10% of patients) Haematological complications
• Purpura • Haemolytic anaemia*,‡
• Cutaneous ulcers* • Unexplained leukopaenia (lymphopaenia and
• Annular erythema (9% of patients) neutropaenia)
• Xerosis cutis (abnormally dry skin; 23–68% of patients) • Unexplained thrombocytopaenia*
Articular and muscle complications • Evans syndrome‡
• Arthralgias (joint pain; 60–70% of patients) • Unexplained monoclonal gammopathy
• Non-erosive symmetric arthritis • Thrombotic thrombocytopaenic purpura*,‡
• Subclinical synovitis (20–30% of patients) • B cell lymphoma*
• Jaccoud arthropathy (non-erosive joint disorder)‡
Obstetrics
• Myalgias (20–40% of patients)
• Autoimmune congenital heart block*
• Myositis‡
• Cardiac fibroelastosis
Pulmonary complications • Unexplained fetal valvular disease
• Chronic obstructive lung disease
• Neonatal lupus
• Bronchiectasis
• Interstitial lung diseases* Ear, nose and throat complications
• Pleuritis‡ • Recurrent parotid enlargement
• Bilateral multicystic parotid masses
Cardiovascular complications
• Sensorineural hearing loss
• Raynaud phenomenon (13% of patients)
• Parotid lymphoma*
• Pericarditis‡
• Pulmonary arterial hypertension*,‡ *Denotes severe systemic manifestations74,84,90. ‡Denotes rarely
reported manifestations (<1% of patients) or suggesting
• Dysautonomia polyautoimmunity (that is, the coexistence of other systemic
• Cryoglobulinaemic vasculitis* autoimmune diseases).

8 | 2016 | VOLUME 2 www.nature.com/nrdp


© 2016 Macmillan Publishers Limited. All rights reserved
PRIMER

a b c

d e f

Figure 3 | Systemic involvement in patients with SjS. a | Synovitis (swelling of the extensor tendon sheath; black arrows)
Nature Reviews | Disease Primers
and arthritis (fusiform swelling of the proximal interphalangeal joints; white arrows). b | Cutaneous vasculitis or raised
reddish-purple spots on the legs. c | Raynaud phenomenon: white fingers (inadequate blood flow; white arrows) and red
fingers (return of blood flow; black arrows). d | Biopsy-proven lymphocytic interstitial pneumonia: multiple areas of
ground-glass attenuation on pulmonary CT (arrows). e | Cerebral multiple sclerosis‑like involvement: bright spots (arrows)
represent demyelinating lesions on cerebral MRI. f | Parotid mucosa-associated lymphoid tissue (MALT) lymphoma:
enlarged right parotid gland with diffuse cystic replacement (arrows). SjS, Sjögren syndrome.

Autoantibodies and immunological abnormalities. specific antibodies against Ro52 and Ro60 (REF. 97). Recent
Autoantibodies are the key serological markers of auto­ studies suggest that anti-Ro52 and anti-Ro60 autoantibod­
immune diseases and can be present up to 20 years before ies might be associated with different clinical phenotypes
the diagnosis of SjS52,55. of SjS and that separate detection is desirable when a
ANAs are a range of autoantibodies that are specific diagnosis of SjS is suspected98. Anti‑Ro52 antibodies are
for nuclear and cytoplasmic components of human cells. closely associated with the main clinical, immunological
ANAs are the most frequently detected autoantibodies and histopathological features of SjS99. In addition, Ro52
in patients with SjS (found in >80% of patients) and, as has been detected on the surface of apoptotic cardiac
such, are one of the better ways to identify SjS in patients cells from patients with congenital heart block100, which
presenting with sicca features in non-specialized clinical is an acquired heart disease that can develop in the fetus
settings, such as in primary care. of mothers with SjS and is associated with the transfer of
Rheumatoid factor antibodies are specific for the Fc maternal Ro52 antibodies across the placenta to the fetus.
fragment of IgG. Rheumatoid factor is present in 50% of Anti-La/SSB antibodies target the La/SSB protein,
patients with SjS and has been associated with the main which is involved in variable aspects of RNA metabolism,
clinical, histopathological and laboratory features of including the binding of viral-related RNAs. Anti-La/SSB
this disease90. antibodies are detected in up to 50% of patients with SjS
The detection of ANA and rheumatoid factor auto­ and are frequently associated with the presence of anti-Ro/
antibodies is useful in patients who are referred for fur­ SSA antibodies. The detection of both anti-La/SSB and
ther diagnostic assessment from primary care centres to anti-Ro/SSA autoantibodies is associated with a higher
indicate the presence of autoimmune disease. However, chance of testing positive for ANAs101 and with systemic
the detection of ANA and rheumatoid factor antibodies activity of SjS82,102. Only between 2.3% and 7% of patients
is not included in the current classification criteria for SjS, with SjS have isolated anti-La/SSB antibodies without con­
as they are common in a range of autoimmune diseases comitant anti-Ro/SSA antibodies49,103,104. Patients with SjS
(as such, they have low specificity for SjS). and anti-Ro/SSA antibodies reportedly have a higher fre­
Anti-Ro/SSA antibodies are ANAs that target two pro­ quency of the main clinical and immunological features
teins — Ro52 and Ro60 — that are associated with small of SjS than patients with SjS and anti-La/SSB antibodies
molecules of RNA96. Anti-Ro/SSA antibodies are detected only 103. However, patients with SjS and anti-La/SSB anti­
in up to 70% of patients with SjS, although the proportion bodies have a higher frequency of some clinical features of
of patients with these antibodies might vary according to SjS, such as dry mouth and the presence of ANA-specific
the detection method; 12–20% of patients who were found antibodies, than patients who are ­negative for both
to be negative for anti-Ro/SSA antibodies might still have anti-Ro/SSA and anti-La/SSB antibodies103.

NATURE REVIEWS | DISEASE PRIMERS VOLUME 2 | 2016 | 9


© 2016 Macmillan Publishers Limited. All rights reserved
PRIMER

Cryoglobulins are immunoglobulins that precipi­ Salivary biopsy. Histopathological analysis of minor
tate at <37 °C in vitro105. Nearly 10% of patients with SjS salivary gland biopsy is a very specific test for the diag­
present with circulating cryoglobulins, which are over­ nosis of SjS, together with the detection of anti-Ro/SSA
whelmingly mixed (that is, they consist of both IgG and antibodies109. The biopsy technique with the best ­balance
IgM components)105. Cryoglobulins are the principal between efficacy in removing the salivary glands and
immunological markers that are suggestive of a poor safety is using a linear incision of the buccal side of
prognosis in SjS, including a higher prevalence of sys­ the lower lip, rather than the use of a punch biopsy 110;
temic disease, increased risk of B cell lymphoma and parotid gland biopsy has also been used by some111. The
poor survival19,78,89,106. The detection of cryoglobulins in key requirements for an accurate histological evaluation
patients with SjS is essential, both at initial diagnosis of are an adequate number of glands (3–5 in total), with
the disease and during the follow‑up; some data obtained the evaluation of a sufficient glandular surface and the
from laboratory analysis can point to the presence of an determination of an average focus score (the n ­ umber
underlying mixed cryoglobulinaemia, such as low levels of lymphocytic aggregates per 4 mm 2 of glandular
of complement C4, increased levels of rheumatoid factor surface analysed)110.
and serum monoclonal IgM gammopathy 105. FLS detection is considered the gold-standard diag­
Hypocomplementaemia, which is characterized by nostic test for SjS, but results should be interpreted with
low levels of complement C3 and/or complement C4, caution in the absence of a suggestive clinical manifesta­
are found in 10–25% of patients with SjS. Like cryo­ tions. The presence of FLS has been reported in individ­
globulins, hypocomplementaemia is associated with the uals without any signs of SjS, for example, in 2.5% of
adverse outcomes of SjS, such as lymphoma and death19. autopsy subjects112 and 17% of young volunteers113, FLS
Circulating monoclonal immunoglobulins (also is defined as one or more dense aggregate (or focus)
known as monoclonal gammopathy of undetermined of ≥50 lymphocytes in the perivascular or periductal
significance) are found in up to 20% of patients with areas of the salivary glands and is considered the key
SjS107, with mIgGκ being the most frequent type19,108. histopathological feature of SjS110 (FIG. 4). The finding
Monoclonal gammopathy is related to a high preva­ of one or more germinal centre-like structure in the
lence of parotid gland enlargement and extraglandular ­salivary glands has been associated with a higher risk of
features of SjS, among others, and is associated with a ­developing lymphoma114.
poor prognosis, owing to the development of neoplasia
and eventual death107,108. Laboratory abnormalities. Patients with SjS often
have a triad of abnormalities that can be determined
using routine laboratory testing. Approximately one-
Box 2 | Risk indicators for lymphoma development in SjS third of patients with SjS have cytopaenia19,115, includ­
ing normocytic anaemia, leukopaenia (related to
Clinical, laboratory, immunological, histopathological and genetic findings associated lymphopaenia and/or neutropaenia) and thrombocyto­
with increased risk for the development of lymphoma. paenia, especially in patients carrying anti-Ro/SSA or
Clinical features anti-La/SSB antibodies. Cytopaenias are predominantly
• Persistent parotid gland enlargement asymptomatic but might be clinically overt in some
• Purpura cases74, and are closely associated with systemic disease
• European League Against Rheumatism Sjögren syndrome (SjS) Disease Activity Index and poor outcomes19,89,116,117. The detection of polyclonal
(ESSDAI) score of ≥5 hypergammaglobulinaemia points to polyclonal B cell
activation, and the erythrocyte sedimentation rate
Laboratory abnormalities
often correlates with the quantity of circulating gamma
• CD4+ lymphopaenia
immunoglobulins. Serum levels of C‑reactive protein are
Immunological findings usually normal in patients with SjS, so increased ­levels
• Low complement C3 or C4 levels indicate the presence of an infection. Raised serum
• Mixed cryoglobulinaemia ­levels of β2‑microglobulin have been reported in one-
• Monoclonal gammopathy of undetermined significance third of patients with SjS69. Other laboratory abnormal­
• Increased lymphocyte-related cytokine levels (including B cell-activating factor, ities might suggest SjS-related renal involvement (such
FMS-like tyrosine kinase 3 ligand, CXC-chemokine ligand 13 and CXC-chemokine as increased levels of creatinine, proteinuria, hypokal­
ligand 11) aemia and a urine pH of >5.5 despite severe acidaemia)
• Increased β2‑microglobulin levels or associated liver disease (indicated by increased levels
• Presence of rheumatoid factor of ­transaminases or alkaline phosphatases).
Histopathological features
Classification criteria
• Presence of germinal centres in the lymph nodes The AECG classification criteria118 have been used
• Focus score of >3 worldwide for the past 10 years and are still the criteria
Genetic polymorphisms used in clinical practice for the diagnosis of SjS. This
• TNFSF13B criteria require evidence of a SjS-specific autoimmune
• TNFRSF13C process (either the detection of anti-Ro/SSA and/or
• TNFAIP3
anti-La/SSB antibodies or the presence of FLS in ­salivary
gland biopsy), and has a sensitivity of 93.5% and a

10 | 2016 | VOLUME 2 www.nature.com/nrdp


© 2016 Macmillan Publishers Limited. All rights reserved
PRIMER

Table 1 | Diagnostic tests to evaluate sicca symptoms in patients with SjS


Diagnostic Technical details Comments Refs
test
Evaluating the main salivary glands (parotid and submandibular glands)
Unstimulated Quantification of salivary volume (weight) produced Results can be influenced by age, 1
salivary flow in a defined time length of disease, comorbidities,
rates temperature or medications
Stimulated Measurement of the salivary flow rate after Better correlation with 1
salivary flow stimulation with chewing gum or lemon juice histopathological results or structural
rates glandular damage compared with the
unstimulated salivary flow rate test
Salivary Non-invasive quantification of uptake and secretion Severe involvement (grade IV) at 93
scintigraphy of technetium 99 by the salivary glands diagnosis associated with higher
systemic activity and poor outcomes
Evaluating the lacrimal glands
Schirmer’s test I Measurement of lacrimal gland output (measured on Evaluates baseline secretion 94
filter paper under local anaesthesia)
Schirmer’s test II Measurement of lacrimal gland output (measured on Measures baseline plus reflex 94
filter paper without anaesthesia) secretion
Corneal Evaluation of corneal damage (degenerated or dead Nowadays, rose bengal (a non-vital 94
stainings cells) through the use of dyes (such as fluorescein, dye) has been substituted by
rose bengal and lissamine green) and a slit lamp lissamine green (a vital dye)
SjS, Sjögren syndrome.

specificity of 94%. In 2012, Shiboski et al.119 proposed a Screening and prevention


new set of classification criteria for SjS, on behalf of the SjS is a chronic disease that cannot be prevented.
American College of Rheumatology (known as the ACR However, screening approaches that target specific sub­
criteria), which is similar to the AECG classification in sets of patients with SjS might either prevent or ensure
both sensitivity (92.5%) and specificity (95.4%). timely treatment of the main complications. Oral and
ocular dryness are the principal symptoms, together with
Differential diagnosis episodic parotid gland swelling, that are indicative of SjS.
The most frequent cause of sicca symptoms is the However, current evidence suggests that the diagnosis of
chronic use of drugs that can cause dryness of mucosal SjS should not be delayed until the development of sicca
surfaces, mainly antihypertensive, antihistamine and symptoms. A large number of non-sicca features might
anti­depressant medications, and these medications are appear up to 20 years before the development of sicca
commonly used in the elderly. In addition, sicca symp­ symptoms52,53, including the development of annular
toms can be caused by a range of external or environ­ erythema, autoimmune cytopaenias, or congential heart
mental factors that might cause mucosal dryness: for block or neonatal lupus erythematosus in babies carried
example, allergy and/or atopy, local infections, dehydra­ by mothers with anti‑Ro/SSA antibodies123. The presence
tion or irradiation, chronic viral infections such as of autoantibodies (especially anti-Ro/SSA antibodies) can
HCV, human immunodeficiency virus or HTLV1, and be used for the early diagnosis of SjS in these patients.
systemic diseases that mimic the symptomology of SjS By contrast, the frequency of anti-Ro/SSA or anti-La/SSB
through the infiltration of the exocrine glands by granu­ autoantibodies is often lower in patients with SjS who
lomas (sarcoidosis and tuberculosis), amyloid proteins only have neurological involvement than in patients with
(amyloidosis) or malignant cells (haematological neo­ SjS who have no neurological involvement, and these
plasia). A more recently recognized cause of s­ alivary patients often require a salivary gland biopsy showing
gland infiltration by immune cells is IgG4‑related the presence of FLS for the early diagnosis of SjS74. Some
disease (IgG4‑RD). The involvement of organs infre­ studies suggest that SjS is now being diagnosed earlier
quently affected by SjS (such as the pancreas, biliary in life; in a Spanish cohort, the mean age at SjS diagno­
tract, gallbladder, mediastinum, retroperitoneum, sis was 62 years in 1997 (REF. 124) and was reported as
hypophysis and gonads) can be the first clinical sign of 54 years in 2015 (REF. 19).
IgG4‑RD120. Raised levels of IgG4 have been reported
in 7% of patients with SjS121, and IgG4‑RD should be Management
confirmed in these patients by histopathological and Monitoring and follow‑up
­immunohistochemical study of the salivary glands Given the heterogeneity of SjS, the frequency and tools
and other organs120. used for monitoring and follow‑up will differ from
As SjS affects men less often than women, other dis­ patient to patient. Studies have shown that, in many
eases with similar symptoms to SjS, such as HCV infec­ cases, the progression of the disease can be successfully
tion, IgG4‑RD and sarcoidosis, should be specifically determined from diagnosis, whereas the disease might
investigated in men who present with sicca syndrome49,122. follow an unexpected path in others. Thus, a general

NATURE REVIEWS | DISEASE PRIMERS VOLUME 2 | 2016 | 11


© 2016 Macmillan Publishers Limited. All rights reserved
PRIMER

However, with sufficient self-monitoring, patients can


a b be seen yearly or when required, for example, when
pre-defined symptoms or situations that mirror potential
lymphoma development appear. The physician in charge
should monitor the ESSDAI score. Two important risk
factors not included in the ESSDAI are high focus scores
(≥3) or the presence of germinal centres in the salivary
glands67,127. The more of these factors that are present, the
higher the risk of development of systemic complications
or lymphoma, so more-frequent follow-ups are required.
If prevailing lymphoma is suspected, further steps should
be determined in collaboration with onco­logists128.
c d With regard to eye and mouth dryness, monitor­ing and
­follow‑­up of SjS aim to prevent irreversible tooth dam­
age, which is mainly caused by the lack of saliva. At least
bi‑annual dental visits for prophylactic treatment and
for the early recognition of tooth damage are necessary
for patients with SjS129. The ocular surface is rarely sub­
jected to irreversible damage as a consequence of SjS, and
regu­lar ophthalmologist visits are mandatory in the most
severe and treatment-resistant cases.
Age at disease onset or diagnosis can be important
for patient monitoring 11. Younger patients (<40 years
of age), who become positive for anti-Ro/SSA or
Figure 4 | Histopathological features of the salivary gland inReviews
Nature SjS scenarios. Although
| Disease Primers anti-La/SSB autoantibodies, less often develop sicca
focal lymphocytic sialadenitis (FLS) is the key histopathological feature of Sjögren
symptoms but are prone to more-severe and systemic
syndrome (SjS), salivary glands within normal limits (meaning the glandular tissue is
non-pathological but is different to that observed in healthy individuals), in addition to disease than older patients (>40 years of age) with
other morphological patterns of chronic inflammation can be observed in salivary gland these autoantibodies53. The presence of well-preserved
tissue from patients with SjS. a | Salivary glands within normal limits showing the general ­g landular ­t issue with a high functional capacity in
architecture with scattered plasma cells (circles), without acinar atrophy. b | Nonspecific younger patients might provide an explanation.
chronic sialadenitis showing scattered or focal infiltration of lymphocytes, macrophages A subgroup of patients with SjS (almost 25% of
and plasma cells (circles) into the salivary gland, and are not adjacent to normal looking patients) who fulfil the 2002 AECG criteria118 (BOX 4)
acini that are located in the gland lobules. c | FLS is distinguished by one or more dense present without detectable levels of anti-Ro/SSA or
aggregate of ≥50 lymphocytes (circles; usually more than several hundred), generally anti-La/SSB antibodies 19. These patients are often
located in perivascular or periductal locations. The aggregates are adjacent to middle-­aged or older and have a milder disease course
normal-appearing acini, found in gland lobes or lobules without duct dilation or
in terms of systemic complications and disease-related
interstitial fibrosis and only contain low numbers of plasma cells. d | Sclerosing chronic
sialadenitis characterized by interstitial fibrosis (black arrows), in addition to several lymphomas. However, sicca symptoms and fatigue are
patterns of chronic inflammation (circles) and acinar atrophy (yellow arrows). Daniels frequent and severe, which makes the use of the ESSPRI
et al.110 suggest that sclerosing chronic sialadenitis could be an advanced stage of tool to capture the disease burden ideal126. The ESSDAI
nonspecific chronic sialadenitis. Images courtesy of L. A. Hernández, Hospital Clinic, would typically be very low (<3). After an initial assess­
Barcelona, Spain. ment, ESSDAI measurements should be repeated after
1 or 2 years, which can be done by a well-informed
general practitioner. Management will generally focus
monitoring and follow‑up plan that is suitable for all on supportive measures for fatigue and arthralgias and
patients cannot be provided and will depend on the risk monitoring of unexpected organ complications.
of complications.
The ESSDAI125 is a very important tool for evaluating Management of sicca symptoms
systemic disease activity in patients with SjS and includes Symptomatic treatment of SjS is usually limited to the
the evaluation of a range of laboratory tests (BOX 3), which treatment of sicca symptoms (FIG. 2). For the treatment
should all be fully assessed at diagnosis of SjS and yearly of dry eyes, tear substitution therapy consisting of
during the first 5–10 years thereafter 19. The EULAR SjS high-viscosity eye drops and gels, in combination with
Patient-Reported Index (ESSPRI) instrument seems less oral mucolytic agents to dissolve mucus, is the mainstay
useful than the ESSDAI for evaluating systemic activity, treatment. Patients with SjS-associated severe or refrac­
although fatigue might be bothersome from early on126. tory keratoconjunctivitis sicca might require a short-
Patients with SjS should be subdivided into two term course of topical anti-inflammatory agents (for
groups: those with risk factors for systemic complications example, twice daily topical cyclosporine). Autologous
or lymphoma and those with little risk of these compli­ serum eye drops (eye drops containing biochemical
cations. Between 25% and 30% of patients with SjS are at components that mimic natural tears more closely
high risk for the development of systemic complications than regular eye drops) and temporary occlusion of the
and merit closer follow‑up; in these patients, a clinical puncta (a procedure to reduce the rate of tear drainage
assessment every 3–6 months should be mandatory. from the eye) have been used in most refractory cases130.

12 | 2016 | VOLUME 2 www.nature.com/nrdp


© 2016 Macmillan Publishers Limited. All rights reserved
PRIMER

Local management of the oral component of SjS is not superior to placebo in patients with SjS132, although
focused on moistening of oral surfaces, in addition to the low disease activity of patients who were enrolled in
the use of preventive measures to preserve or obtain this study (a mean ESSDAI score of 2) does not allow
optimal oral health. Correct oral hygiene combined with ap­ otential benefit of the drug in systemic disease to
frequent application of fluorides in patients with dental be excluded.
complications are important to prevent the occurrence of The usefulness of other immunosuppressive agents
new dental caries and to slow the progression of existing (such as cyclosporine A, azathioprine, methotrexate,
caries129. Several saliva substitutes are available but are not leflunomide and mycophenolic acid) has been evalu­
used over time by many patients. ated in a small cohort of patients with SjS130. The results
Drugs with a systemic effect, for example, secreta­ from these studies are homogeneous, and limited benefit
gogues, such as pilocarpine and cevimeline, can also be was seen on the sicca symptoms of SjS; the benefits on
used for the symptomatic management of sicca symp­ systemic disease were not evaluated. In addition, high
toms. Both drugs are muscarinic receptor agonists and rates of adverse events were observed (seen in 41–100%
induce a transient increase in salivary and lacrimal gland of patients)130.
outputs in patients with some residual functional
­glandular tissue. Both drugs are approved by the US FDA Biological therapies. Promising results have been
in the United States for the treatment of dry eyes and reported with several biological agents, especially those
mouth in patients with SjS, but cevimeline is not avail­ targeting B cells. B cell depletion therapy with rituximab
able worldwide. Common adverse effects of these drugs (a CD20‑specific antibody) reduced some symptoms
include sweating, flushing, the urgent need for urination of SjS, including fatigue, and improved salivary flow in
and gastrointestinal discomfort, which might limit their two small RCTs and also in uncontrolled studies134–139.
clinical application130,131. Serological and histological analysis showed complete
depletion of B cells in serum, a partial depletion of B cells
Management of systemic disease in the s­ alivary glands140 and a reduction in the absolute
Only a few clinical trials have investigated the effect of numbers of follicular helper T cells and their hallmark
systemic treatments for the extraglandular manifesta­ cytokine IL‑21. Despite pretreatment with corticosteroids,
tions of SjS for numerous reasons. First, extraglandular antihistamines and paracetamol, some patients developed
symptoms are heterogeneous (BOX 1). Second, clinical a serum sickness-like syndrome, which can be associ­
­trials generally include patients with low systemic disease ated with hypergammaglobulinaemia, making patients
activity, for example, thereby excluding several patient with SjS prone to the formation of antichimeric anti­
subgroups in SjS. Third, there was previously a lack of bodies against rituximab135. A multicentre RCT recruited
validated outcome measures to evaluate the systemic 122 patients in a consecutive manner and assigned them to
manifestations in patients with SjS130. However, treat­ rituximab infusions (1 g) or placebo treatments at week 0
ments for ­extraglandular manifestations of SjS need to and week 2. This trial did not show significant differences
be developed. in the primary outcome (a composite score that evalu­
ated dryness, pain, fatigue and general health at 24 weeks),
Non-biological therapies. Non-biological therapies are objective diagnostic tests (for example, Schirmer’s test and
still used for SjS treatment, even though strong evidence salivary gland biopsy) or the ESSDAI score. Significant
is lacking. The main non-biological drugs that are used differences were found for some other end points (that is,
for the treatment of SjS are corticosteroids and hydroxy­ sicca symptoms, fatigue, visual analogue scale and salivary
chloroquine. The use of prednisone (a corticosteroid) to flow rate)136. Post hoc analysis applying the SjS response
treat SjS is based on its effectiveness in the management index also showed a significant difference in response rate
of other autoimmune diseases. Based on clinical experi­ between rituximab and placebo groups141. Despite the
ence, low-dose prednisone is used for the treatment of findings of this trial, rituximab is prescribed off-label for
arthritis and cutaneous symptoms. High doses of predni­ the management of severe ­extraglandular manifestations
sone have shown efficacy for the treatment of kidney, of SjS, including lymphoma131.
lung and CNS features in uncontrolled studies on SjS130.
The long-term use of corticosteroids is associated with
adverse effects, including osteoporosis, diabetes, weight Box 3 | The ESSDAI
gain and dyslipidaemia130. The European League Against Rheumatism Sjögren
The therapeutic effect of hydroxychloroquine is syndrome (SjS) Disease Activity Index (ESSDAI) is used to
based on the inhibition of TLR signalling, which affects assess the severity of SjS and includes assessments of the
the innate immune response by reducing the production following laboratory parameters.
of pro-inflammatory cytokines, such as type I IFNs, and • Complement levels
other serological parameters, such as IL‑6, IgG, erythro­ • Blood cell counts
cyte sedimentation rate and rheumatoid factor 132,133. • Cryoglobulin levels
Hydroxychloroquine is used as maintenance therapy in • Serum gammaglobulin levels
SjS, based on the reduction of symptoms and improved • IgG levels
serological parameters in patients with SLE and rheuma­ • Serum monoclonal components
toid arthritis133. However, a randomized controlled trial
• Muscle enzyme levels
(RCT) found that hydroxychloroquine treatment was

NATURE REVIEWS | DISEASE PRIMERS VOLUME 2 | 2016 | 13


© 2016 Macmillan Publishers Limited. All rights reserved
PRIMER

Other biological therapies that target B cells include was not evaluated142. An open-label trial of belimumab
epratuzumab and belimumab. Epratuzumab is a B cell (a BAFF-blocking drug) showed a decrease in ESSDAI
receptor CD22‑specific monoclonal antibody that acts scores in 60% of patients treated with this drug 143.
via B cell antigen receptor signalling and leads to partial With respect to T cell targeting, abatacept (a fusion
B cell depletion. A small prospective study in patients molecule) prevents CD28 co‑stimulation between
with SjS showed a beneficial effect of epratuzumab on antigen-presenting cells, including B cells and T cells.
fatigue, but the effect on extraglandular manifestations Abatacept is also presumed to inhibit the full activa­
tion of T cells and T cell-dependent B cell activation.
In two open-label trials, abatacept reduced the glandular
Box 4 | 2002 AECG criteria for the diagnosis of SjS and extraglandular manifestations of SjS, reflected by a
Revised international classification criteria for SjS* signifi­cant improvement in ESSDAI scores, and a reduc­
I. Ocular symptoms: a positive response to at least one of the following questions: tion in the levels of IgG and rheumatoid factor and in the
1. Have you had daily, persistent, troublesome dry eyes for more than 3 months? number of follicular helper T cells144,145. A phase III RCT
2. Do you have a recurrent sensation of sand or gravel in the eyes? (ASAP III trial) is ongoing 146 (TABLE 2).
3. Do you use tear substitutes more than 3 times a day?
II. Oral symptoms: a positive response to at least one of the following questions:
Management of lymphomas
1. Have you had a daily feeling of dry mouth for more than 3 months?
2. Have you had recurrently or persistently swollen salivary glands as an adult?
Following the diagnosis of lymphoma, the therapeutic
3. Do you frequently drink liquids to aid in swallowing dry food? approach must be planned according to the subtype of
III. Ocular signs — that is, objective evidence of ocular involvement defined as a lymphoma present. Some clinicians recommend a watch­
positive result for at least one of the following two tests: ful waiting approach in patients with low-grade lympho­
1. Schirmer’s test I, performed without anaesthesia (≤5 mm in 5 minutes) mas that affects the exocrine glands only 65. By contrast,
2. Rose bengal score or other ocular dye score (≥4 according to van Bijsterveld’s patients with disseminated MALT lymphoma or those
scoring system) with concomitant high disease activity have an increased
IV. Histopathology: in minor salivary glands (obtained through normal-appearing mucosa) risk of lymphoma progression, so could benefit from
focal lymphocytic sialoadenitis, evaluated by an expert histopathologist, with a focus score early treatment to stop progression into more-aggressive
>1, defined as a number of lymphocytic foci (which are adjacent to normal-appearing
types of lymphomas65. The best therapeutic regimen for
mucous acini and contain more than 50 lymphocytes) per 4 mm2 of glandular tissue
V. Salivary gland involvement: objective evidence of salivary gland involvement defined
the treatment of lymphoma is the combination of rituxi­
by a positive result for at least one of the following diagnostic tests: mab with alkylating agents, such as cyclophosphamide,
1. Unstimulated whole salivary flow (≤1.5 ml in 15 minutes) chlorambucil, fludarabine and/or bendamustine128,147.
2. Parotid sialography showing the presence of diffuse sialectasias (punctate, cavitary The combination of rituximab and bendamustine has
or destructive pattern), without evidence of obstruction in the major ducts been shown to be effective and safe for marginal zone
3. Salivary scintigraphy showing delayed uptake, reduced concentration and/or and MALT lymphomas147. In patients with diffuse large
delayed excretion of tracer B cell lymphoma, treatment should be adapted according
VI. Autoantibodies: presence in the serum of the following autoantibodies: to the histological grade of the lymphoma and is based
1. Antibodies to Ro/SSA or La/SSB antigens, or both on the association of rituximab with cyclophosphamide,
Revised rules for classification doxorubicin, vincristine and prednisone148.
For primary SjS. In patients without any potentially associated disease, primary SjS may
be defined as follows: Quality of life
• a. The presence of any four of the six items is indicative of primary SjS, as long as The main symptoms associated with SjS are unpleasant
either item IV (histopathology) or VI (serology) is positive and can be debilitating. Thus, many patients report a
• b. The presence of any three of the four objective criteria items (that is, items III, IV, reduction in their quality of life (QOL) or, more specifi­
V and VI) cally, a reduction in their health-related QOL (HRQOL).
• c. The classification tree procedure represents a valid alternative method for QOL is defined as the overall well-being of individuals
classification, although it should be more properly used in clinical–epidemiological and societies and covers a broad range of concepts, includ­
surveys ing personal well-being, the environment, happiness levels
For secondary SjS. In patients with a potentially associated disease (for instance, and personal freedom, some of which are not necessarily
another well-defined connective tissue disease), the presence of item I or item II plus relevant to SjS. In a study evaluating QOL and HRQOL in
any two from among items III, IV and V may be considered as indicative of secondary SjS, patients demonstrated reduced QOL and HRQOL to
SjS. Exclusion criteria: a comparable level as patients with rheumatoid arthri­
• Past head and neck radiation treatment tis or SLE149. Other studies have found similar results
• Hepatitis C virus infection with reduced QOL measured by the WHOQOL-BREF,
• Acquired immunodeficiency syndrome (AIDS) which is linked to psychological disorders such as anxiety
• Pre-existing lymphoma and somatization150,151. In addition, the 36‑Item Short-
• Sarcoidosis Form Health Survey (SF‑36) shows a reduced HRQOL
• Graft-versus-host disease
in patients with SjS similar to that seen in patients with
rheumatoid arthritis or SLE; compared with SjS, the physi­
• Use of anticholinergic drugs (as a time shorter than fourfold the half-life of the drug)
cal and pain domains of patients are more profoundly
AECG, American–European Consensus Group; SjS, Sjögren syndrome. *See REF. 118. reduced in rheumatoid arthritis149,152–154. Generally, a
Reproduced from Classification criteria for Sjögren’s syndrome: a revised version of the
European criteria proposed by the American–European Consensus Group, Vitali, C. et al., 61,
reduction in the HRQOL-assessed SF‑36 scores is more
554–558, © 2002, with permission from BMJ Publishing Group Ltd. dependent on fatigue, pain and low mood than on the
severity of dryness symptoms in patients with SjS154,155.

14 | 2016 | VOLUME 2 www.nature.com/nrdp


© 2016 Macmillan Publishers Limited. All rights reserved
PRIMER

Table 2 | Ongoing diagnostic and therapeutic studies in primary SjS


ClinicalTrials.gov Patients Diagnostic approach or procedure Type of study Study phase Funding
identifier
Diagnosis
NCT02132585 SjS Speckle-tracking echocardiography Observational N/A Other
NCT01470638 SjS FLT3 ligand levels Observational N/A Other
NCT01807689 SjS Salivary biomarkers Observational N/A Other
NCT00074373 Congenital heart block Research registry Observational N/A Other
New drug interventions
NCT02004067 SjS Ocular cyclosporine Interventional Phase IV Industry
NCT01316770 SjS Dexamethasone irrigation Interventional Phase II US NIH
NCT00543166 SjS Dehydroepiandrosterone Interventional Phase IV Other
NCT01759108 SjS Rebamipide Interventional NP Other
NCT01782235 SjS Tocilizumab Interventional Phase II/III Other
NCT02067910 SjS Abatacept Interventional Phase III Industry
NCT02464319 SjS Human IL‑2 Interventional Phase II Other
NCT01552681 SjS Baminercept Interventional Phase II NIH
NCT02610543 SjS UCB5857 Interventional Phase II Industry
NCT02334306 SjS AMG 557 or MEDI5872 Interventional Phase II Industry
NCT02149420 SjS VAY736 Interventional Phase II Industry
NCT02291029 SjS CFZ533 Interventional Phase II Industry
NCT01379573 Congenital heart block Hydroxychloroquine Interventional Phase II Other
NCT02503163 Dry eyes with SjS KCT‑0809 ophthalmic solution Interventional Phase III Industry
NCT02503176 Dry eyes with SjS KCT‑0809 ophthalmic solution Interventional Phase III Industry
NCT02503189 Dry eyes with SjS KCT‑0809 ophthalmic solution Interventional Phase III Industry
Other therapeutic approaches
NCT02370225 SjS Aerobic exercise Interventional NP Other
NCT01501019 SjS Exercise training Interventional NP Other
NCT02031172 SjS Effects of sustained reading on the ocular surface Observational NP Other
NCT02110446 SjS Chinese herbal formula SS1 Interventional Phase II Other
NCT00953485 SjS Allogeneic mesenchymal stem cells Interventional Phase I/II Other
NCT02112019 SjS Sialoendoscopy Interventional NP Other
NCT01174329 SjS Automatic neuro-electrostimulation Interventional Phase II/III Other
NCT02153515 Dry eyes Fingerprick autologous blood Interventional Phase III Other
NCT02193490 Dry eyes DNase Interventional Phase I/II Industry
NCT02148497 Dry eyes Multicoloured Placido disk imaging Interventional NP Other
NCT02066896 Keratoconjunctivitis sicca Laser 3R 790 nm infrared GaAIAs Interventional Phase II Other
Data from ClinicalTrials.gov, accessed December 16 2015. FLT3, FMS-like tyrosine kinase 3; GaAIA, gallium-aluminium-arsenide; N/A, not applicable;
NP, not provided; SjS, Sjögren syndrome; SS1, Strengthen Earth 1 oz.

The EuroQol EQ‑5D tool has also confirmed a reduced Outlook


HRQOL in patients with SjS, and linked this to more-­ The clinical knowledge of SjS has rapidly evolved over
severe symptoms (particularly pain levels) and the pres­ recent years, but not enough to permit considerable
ence of systemic disease156. Although there are some changes in the daily management of patients. The areas
differences between these studies regarding the worst-­ with the biggest changes are discussed below.
affected domains of HRQOL in SjS and the associations
of HRQOL with other features of disease, they consistently Novel aetiopathogenetic pathways
show the reduction of HRQOL in patients with SjS and the Some studies are investigating mechanisms previously
association between systemic features of disease, such as identified in other diseases, such as IFN-related mech­
fatigue, pain and low mood. The presence of dryness was anisms that involve the expression of microRNA and IFN-
less closely associated with a reduced HRQOL, if at all. inducible proteins, such as IFNγ-inducible protein 16, and

NATURE REVIEWS | DISEASE PRIMERS VOLUME 2 | 2016 | 15


© 2016 Macmillan Publishers Limited. All rights reserved
PRIMER

cytokines, for example, IL‑17 and IL‑21 (REFS 157,158). Unfortunately, the heterogeneous diagnostic approaches
Other aetiopathogenetic pathways under investigation used to confirm SjS have halted the field. The need for
include autophagy, the role of aromatase and, especially, consensus is clear, and the EULAR-SjS Task Force Group
epigenetic dysregulation, which might be restricted to the is working on developing organ-specific recommenda­
main cellular components involved in the aetiopatho­ tions84 and new scores for better characterization and
genesis of SjS, such as epithelial cells and lymphocytes159,160. measurement of systemic disease173,174. The scoring of sys­
Epigenetic mechanisms including histone modifications, temic disease might provide better patient stratification,
non-coding RNAs and, particularly, DNA methylation, as patients with the greatest systemic activity at diagnosis
could have an aetiopathogenetic role in SjS161. With of SjS have been shown to have a poorer prognosis19,89.
respect to novel antigens, some new molecules have also
been proposed, for example, the poly(U)-­binding-splicing New diagnostic tests
factor PUF60 and carbamylated proteins162,163. Cell types Some imaging techniques, such as three-dimensional
under investigation include ­natural killer cells; in one MRI and 18F-fluorodeoxyglucose PET, are increasingly
study, patients with SjS with low numbers of n ­ atural killer used to improve the non-invasive diagnosis of SjS,
cells in salivary gland tissue and blood responded better to including parotid and dorsal root involvement of the
belimumab treatment than patients with higher numbers disease, in addition to lymphadenopathy or interstitial
of natural killer cells164. lung disease175–177.
Ultrasonography is increasingly used for the diagno­
Environmental factors sis of SjS. Several studies have found a potential role for
The classic debate on the influence of genetics versus ultrasonography in the early diagnosis of SjS, with a high
environment factors on the pathogenesis of human dis­ diagnostic accuracy 178,179.
ease also exists for SjS. One study has estimated the herit­
ability of SjS to be around 54%60, but other studies have Classification criteria
focused on the effect of environmental factors. Examples The existence of two sets of classification criteria (the
of novel environmental factors include the human o ­ cular AECG and the ACR criteria), both of which have simi­
and oral microbiota in SjS165,166. Other aetiopathogenetic lar diagnostic accuracies, has induced some confusion
investigations include the potential effect of exposure to among the clinical community. An international ACR–
environmental toxins, including a high risk of developing EULAR study group is trying to identify a consensual
SjS in people with occupational exposure to solvents167, set of criteria that maintains the same standards as pre­
in rescue and recovery workers exposed to aerosol­ vious sets, namely, the mandatory presence of either
ized dust in the 2001 World Trade Center site168 or an anti-Ro/SSA antibodies (anti-La/SSB antibodies are over­
increased risk of SjS associated with airborne pollution whelmingly associated with anti-Ro/SSA antibodies) or
(fine particulates)169. FLS. Future efforts should explore the usefulness of incor­
porating features other than the presence of anti-Ro/SSA
Diagnosing sicca symptoms autoantibodies and the glandular involvement of disease,
For ocular tests, studies are now using double staining including organ-specific ESSDAI involvements (clinical,
(the use of both lissamine green and fluorescein), to cal­ laboratory and immunological) and the use of novel
culate the ocular staining score170 and have investigated imaging techniques (especially ultrasonography, but also
specific damage in some corneal areas, such as the central CT or MRI) into the diagnostic criteria for SjS.
optical zone171 or the influence of a desiccating environ­
ment on corneal staining 172. Technical developments Complexity and chronicity
are promoting more-accurate quantitative evaluation SjS is associated with a considerable number of comorbid­
in salivary gland scintigraphic studies with enhanced ities, principally, the higher risk of lymphomas, but also
accuracy, although there remains no clear consensus on other systemic or organ-specific autoimmune diseases180,
their use in clinical practice93. With respect to salivary non-haematological cancer 181 and cardio­vascular 56,182 and
gland biopsy, an international consensus on updating infectious19 complications. This constellation of associ­
the histopathological classification that has been used ated disorders often provokes considerable concern and
with minimal variations for >40 years is needed. A close doubt in both patients and physicians, and is respon­
cooperation between clinicians and pathologists should sible for the high degree of complexity and chronicity
also be promoted to facilitate the integration of patho­ of SjS, which leads to increased hospitaliza­tion and out­
logical results with clinical manifestations of each patient. patient visits183. Studies focusing on the complexity and
Histopathological findings that do not fit the current chronicity of SjS are required for better understanding of
criteria, such as focal aggregates of <50 lymphocytes, disease outcomes.
chronic atrophic sialadenitis and the presence of germi­
nal centre-like structures, could have some diagnostic Drug-free management
value if integrated into the appropriate clinical scenario. Drug-free therapeutic interventions often receive little
attention and resources, and data from a systematic
Diagnosing systemic disease review showed that all drug-free therapeutic studies
The increasing use of the ESSDAI and the recent pub­ in SjS were of low quality and had a high risk of bias,
lication of data from large European cohorts using the measured according to the Cochrane Risk of Bias Tool184.
ESSDAI19,78 have confirmed SjS as a systemic disease. Future studies should evaluate the disease from a holistic

16 | 2016 | VOLUME 2 www.nature.com/nrdp


© 2016 Macmillan Publishers Limited. All rights reserved
PRIMER

perspective, focusing on the influence of determinants Disruptive innovations


involving personal life and work on the phenotypic Disruptive innovations that displace earlier technologies
expression of SjS. This should include the effects of diet or working methods are rapidly emerging in this cen­
(Mediterranean, Japanese, high-sugar and processed tury, including health-related areas, which are among
food), regular aerobic exercise (principally for the the areas with the greatest potential for dramatic change.
manage­ment of non-sicca general symptoms) and stress In SjS, the most innovative therapeutic approaches are
(which acts not only to worsen the intensity of sicca those investigating the use of gene therapy to modulate
­features but also as a potential trigger of systemic flares). aquaporin or IL‑17 expression192, the role of robotic or
bioartificial techniques, including tissue engineering-­
Biological therapies based organ regeneration of lacrimal or salivary
Direct and indirect B cell blockade seems to be the glands193, and the proteomic and genomic analysis of
most-promising approach in terms of biological thera­ saliva to search for SjS-specific diagnostic soluble bio­
pies for the management of SjS, although other therapeu­ markers194,195. Enhanced understanding of the influence
tic targets are under investigation (TABLE 2). IL‑6 blockade of the disease on factors external to the patient and the
is another promising option, as IL‑6 induces the polar­ aetiopathogenetic mechanisms of extraglandular dam­
ization of follicular helper T cells and is also important age could lead to the generation of new treatments for
for the induction of IL‑21 production185. IL‑6 blockade is SjS. In addition, a more holistic approach to individual
currently being investigated in a phase III RCT186. Other patient management, the formation of active inter­
co‑stimulatory pathways, such as blocking CD40–CD40 national collaborations promoting multicentre patient
ligand, which crosslinks innate and adaptive immune registries and a consensus on diagnostic and therapeutic
cells and pathways, are also promising therapeutic approaches, and application of new disruptive technolo­
options187. Isolated studies are exploring other drugs, gies have the potential to be gamechangers in our under­
such as fingolimod (an immunomodulatory drug) in standing of the disease and provide solid improvements
patients with multiple sclerosis-like CNS involvement in the QOL and the prognosis of patients with SjS.
and bortezomib188,189. However, the disappointing results
of the two largest RCTs carried out in SjS, which both Quality of life
evaluated drugs that were considered to be effective in Most studies have reported nonspecific general com­
daily practice (hydroxychloroquine and rituximab), have plaints as the main contributors to the poor HRQOL
opened a debate about RCTs in SjS132,136. These trials used in patients with SjS. Ongoing studies are investigating
a composite outcome based on the subjective evaluation other features that could contribute to poor HRQOL,
of dryness, fatigue and pain. The strong influence of per­ for example, inadequate dental and periodontal health196,
sonal and environmental factors on the intensity of this the effect of gynaecological involvement on the sex lives
triad of symptoms could explain the lack of significant of women197,198 and the effect of voice disorders and
differences. In addition, inadequate patient selection, the other ear, nose and throat symptoms on occupational
influence of concomitant drugs and the hetero­geneity and social activities199. In terms of moving forwards,
of diagnostic tests could have contributed to the lack of one key question that remains unanswered is whether
significant differences between placebo and therapeutic therapeutic interventions can lead to improved HRQOL.
groups. The EULAR-SjS Task Force and other inter­ Whether therapies that lead to clinical and symptomatic
national groups are working on finding more solid, improvements are also associated with an enhanced
­objective outcomes for future RCTs in SjS173,190,191. HRQOL is of considerable interest 145,200.

1. Ramos-Casals, M., Brito-Zerón, P., Sisó-Almirall, A. Arthritis Care Res. (Hoboken) 66, 454–463 15. Yao, Q., Altman, R. D. & Wang, X. Systemic
& Bosch, X. Primary Sjögren syndrome. BMJ 344, (2014). lupus erythematosus with Sjögren syndrome
e3821 (2012). 8. Kabasakal, Y. et al. The prevalence of Sjögren’s compared to systemic lupus erythematosus alone:
2. Manoussakis, M. N. et al. Sjögren’s syndrome syndrome in adult women. Scand. J. Rheumatol. 35, a meta-analysis. J. Clin. Rheumatol. 18, 28–32
associated with systemic lupus erythematosus: 379–383 (2006). (2012).
clinical and laboratory profiles and comparison with 9. Ramos-Casals, M. et al. Google-driven search for big 16. Young, A. et al. How does functional disability in early
primary Sjögren’s syndrome. Arthritis Rheum. 50, data in autoimmune geoepidemiology: analysis of rheumatoid arthritis (RA) affect patients and their
882–891 (2004). 394,827 patients with systemic autoimmune lives? Results of 5 years of follow‑up in 732 patients
3. Vitali, C. et al. Preliminary criteria for the diseases. Autoimmun. Rev. 14, 670–679 (2015). from the Early RA Study (ERAS). Rheumatology
classification of Sjögren’s syndrome. Results 10. Mathews, P. M. et al. Ocular complications of primary (Oxford) 39, 603–611 (2000).
of a prospective concerted action supported by Sjögren syndrome in men. Am. J. Ophthalmol. 160, 17. Carmona, L. et al. Rheumatoid arthritis in Spain:
the European Commununity. Arthritis Rheum. 36, 447–452.e1 (2015). occurrence of extra-articular manifestations and
340–347 (1993). 11. Ramos-Casals, M. et al. Primary Sjögren syndrome estimates of disease severity. Ann. Rheum. Dis. 62,
4. Qin, B. et al. Epidemiology of primary Sjögren’s in Spain: clinical and immunologic expression in 897–900 (2003).
syndrome: a systematic review and meta-analysis. 1010 patients. Medicine (Baltimore) 87, 210–219 18. Avouac, J. et al. Associated autoimmune diseases in
Ann. Rheum. Dis. 74, 1983–1989 (2015). (2008). systemic sclerosis define a subset of patients with
5. Kvarnstrom, M., Ottosson, V., Nordmark, B. 12. Civilibal, M. et al. A child with primary Sjögren milder disease: results from 2 large cohorts of
& Wahren-Herlenius, M. Incident cases of primary syndrome and a review of the literature. Clin. Pediatr. European Caucasian patients. J. Rheumatol. 37,
Sjögren’s syndrome during a 5‑year period in (Phila.) 46, 738–742 (2007). 608–614 (2010).
Stockholm County: a descriptive study of the patients 13. Yokogawa, N. et al. Comparison of labial minor 19. Brito-Zerón, P. et al. Systemic activity and mortality
and their characteristics. Scand. J. Rheumatol. 44, salivary gland biopsies from childhood Sjögren in primary Sjögren syndrome: predicting survival
135–142 (2015). syndrome and age-matched controls. J. Rheumatol. using the EULAR‑SS Disease Activity Index (ESSDAI)
6. Weng, M.‑Y., Huang, Y.‑T., Liu, M.‑F. & Lu, T.‑H. 41, 1178–1182 (2014). in 1045 patients. Ann. Rheum. Dis. 75, 348–355
Incidence and mortality of treated primary Sjögren’s 14. Baer, A. N., Maynard, J. W., Shaikh, F., Magder, L. S. (2016).
syndrome in Taiwan: a population-based study. & Petri, M. Secondary Sjögren’s syndrome in 20. Tzioufas, A. G., Kapsogeorgou, E. K.
J. Rheumatol. 38, 706–708 (2011). systemic lupus erythematosus defines a distinct & Moutsopoulos, H. M. Pathogenesis of Sjögren’s
7. Maldini, C. et al. Epidemiology of primary Sjögren’s disease subset. J. Rheumatol. 37, 1143–1149 syndrome: what we know and what we should learn.
syndrome in a French multiracial/multiethnic area. (2010). J. Autoimmun. 39, 4–8 (2012).

NATURE REVIEWS | DISEASE PRIMERS VOLUME 2 | 2016 | 17


© 2016 Macmillan Publishers Limited. All rights reserved
PRIMER

21. Hillen, M. R., Ververs, F. A., Kruize, A. A. 44. Iakimtchouk, K., Myrmel, H. & Jonsson, R. Serological and management. Br. J. Haematol. 168, 317–327
& Van Roon, J. A. Dendritic cells, T‑cells and epithelial screening for hepatitis B and C and human (2015).
cells: a crucial interplay in immunopathology herpesvirus 6 in Norwegian patients with primary This article reviews the aetiopathogenetic
of primary Sjögren’s syndrome. Expert Rev. Clin. Sjögren’s syndrome. J. Rheumatol. 26, 2065–2066 mechanisms and clinical management of
Immunol. 10, 521–531 (2014). (1999). SjS-related lymphoma.
22. Moutsopoulos, H. M. Sjögren’s syndrome: 45. Terada, K. et al. Prevalence of serum and salivary 66. Nocturne, G. et al. Germline and somatic genetic
autoimmune epithelitis. Clin. Immunol. Immunopathol. antibodies to HTLV‑1 in Sjögren’s syndrome. Lancet variations of TNFAIP3 in lymphoma complicating
72, 162–165 (1994). 344, 1116–1119 (1994). primary Sjögren’s syndrome. Blood 122, 4068–4076
This article dissects the main mechanisms on which 46. Nakamura, H. et al. Direct infection of primary salivary (2013).
the previously most-accepted hypothesis of SjS gland epithelial cells by human T lymphotropic virus 67. Theander, E. et al. Lymphoid organisation in labial
aetiopathogenesis was formed. type I in patients with Sjögren’s syndrome. salivary gland biopsies is a possible predictor for the
23. Tzioufas, A. G., Tsonis, J. & Moutsopoulos, H. M. Arthritis Rheumatol. 67, 1096–1106 (2015). development of malignant lymphoma in primary
Neuroendocrine dysfunction in Sjögren’s syndrome. 47. García-Carrasco, M. et al. Hepatitis C virus infection Sjögren’s syndrome. Ann. Rheum. Dis. 70,
Neuroimmunomodulation 15, 37–45 (2008). in primary Sjögren’s syndrome: prevalence and clinical 1363–1368 (2011).
24. Fox, R. I. & Stern, M. Sjögren’s syndrome: significance in a series of 90 patients. Ann. Rheum. 68. Nocturne, G. et al. CXCL13 and CCL11 serum levels
mechanisms of pathogenesis involve interaction Dis. 56, 173–175 (1997). and lymphoma and disease activity in primary
of immune and neurosecretory systems. 48. Haddad, J. et al. Lymphocytic sialadenitis of Sjögren’s Sjögren’s syndrome. Arthritis Rheumatol. 67,
Scand. J. Rheumatol. Suppl. 116, 3–13 (2002). syndrome associated with chronic hepatitis C virus 3226–3233 (2015).
25. Manoussakis, M. N. & Kapsogeorgou, E. K. The role liver disease. Lancet 339, 321–323 (1992). 69. Gottenberg, J. E. et al. Serum levels of
of intrinsic epithelial activation in the pathogenesis 49. Brito-Zerón, P. et al. How hepatitis C virus modifies the beta2‑microglobulin and free light chains of
of Sjögren’s syndrome. J. Autoimmun. 35, 219–224 immunological profile of Sjögren syndrome: analysis immunoglobulins are associated with systemic disease
(2010). of 783 patients. Arthritis Res. Ther. 17, 250 (2015). activity in primary Sjögren’s syndrome. Data at
26. Nocturne, G. & Mariette, X. Advances in 50. Haaheim, L. R. et al. Serum antibodies from patients enrollment in the prospective ASSESS cohort.
understanding the pathogenesis of primary Sjögren’s with primary Sjögren’s syndrome and systemic lupus PLoS ONE 8, e59868 (2013).
syndrome. Nat. Rev. Rheumatol. 9, 544–556 (2013). erythematosus recognize multiple epitopes on 70. Quartuccio, L. et al. BLyS upregulation in Sjögren’s
27. Gong, Y.‑Z. et al. Differentiation of follicular helper the La(SS‑B) autoantigen resembling viral protein syndrome associated with lymphoproliferative
T cells by salivary gland epithelial cells in primary sequences. Scand. J. Immunol. 43, 115–121 (1996). disorders, higher ESSDAI score and B‑cell clonal
Sjögren’s syndrome. J. Autoimmun. 51, 57–66 (2014). 51. Hall, J. C. et al. Molecular subsetting of interferon expansion in the salivary glands. Rheumatology
28. Youinou, P., Saraux, A. & Pers, J.‑O. B‑lymphocytes pathways in Sjögren’s syndrome. Arthritis Rheumatol. (Oxford) 52, 276–281 (2013).
govern the pathogenesis of Sjögren’s syndrome. 67, 2437–2446 (2015). This is an elegant study that supported the efficacy
Curr. Pharm. Biotechnol. 13, 2071–2077 (2012). 52. Jonsson, R. Autoantibodies present before symptom of blocking BAFF in patients with SjS.
29. Morva, A. et al. Salivary gland epithelial cells (SGECs) onset in primary Sjögren syndrome. JAMA 310, 71. Nezos, A. et al. B‑cell activating factor genetic variants
promote the differentiation of B cells. Presented 1854–1855 (2013). in lymphomagenesis associated with primary Sjögren’s
at the12th International Symposium on Sjögren’s This study provides the first evidence showing that syndrome. J. Autoimmun. 51, 89–98 (2014).
Syndrome (2013). SjS-related autoantibodies might be detected 72. Tobón, G. J. et al. The Fms-like tyrosine kinase 3
30. Kyriakidis, N. C. et al. Toll-like receptor 3 stimulation several years before the diagnosis of the disease. ligand, a mediator of B cell survival, is also a marker
promotes Ro52/TRIM21 synthesis and nuclear 53. Theander, E. et al. Prediction of Sjögren’s syndrome of lymphoma in primary Sjögren’s syndrome.
redistribution in salivary gland epithelial cells, partially years before diagnosis and identification of patients Arthritis Rheum. 62, 3447–3456 (2010).
via type I interferon pathway. Clin. Exp. Immunol. 178, with early onset and severe disease course by 73. Carr, A. J. et al. Sjögren’s syndrome — an update
548–560 (2014). autoantibody profiling. Arthritis Rheumatol. 67, for dental practitioners. Br. Dent. J. 213, 353–357
31. Hall, J. C. et al. Precise probes of type II interferon 2427–2436 (2015). (2012).
activity define the origin of interferon signatures in 54. Klareskog, L., Gregersen, P. K. & Huizinga, T. W. J. 74. Brito-Zerón, P. et al. Early diagnosis of primary
target tissues in rheumatic diseases. Proc. Natl Acad. Prevention of autoimmune rheumatic disease: state Sjögren’s syndrome: EULAR‑SS Task Force clinical
Sci. USA 109, 17609–17614 (2012). of the art and future perspectives. Ann. Rheum. Dis. recommendations. Expert Rev. Clin. Immunol. 12,
32. Nguyen, C. Q., Hu, M. H., Li, Y., Stewart, C. 69, 2062–2066 (2010). 137–156 (2016).
& Peck, A. B. Salivary gland tissue expression of 55. Bartoloni, E. et al. Cardiovascular disease risk burden This study notes that patients with SjS can present
interleukin‑23 and interleukin‑17 in Sjögren’s in primary Sjögren’s syndrome: results of a population- with systemic manifestations of disease and are
syndrome: findings in humans and mice. based multicentre cohort study. J. Intern. Med. 278, at high risk of developing poor outcomes.
Arthritis Rheum. 58, 734–743 (2008). 185–192 (2015). 75. Aframian, D. J., Konttinen, Y. T., Carrozzo, M.
33. Bikker, A. et al. Increased expression of interleukin‑7 56. Pérez-De‑Lis, M. et al. Cardiovascular risk factors in & Tzioufas, A. G. Urban legends series: Sjögren’s
in labial salivary glands of patients with primary primary Sjögren’s syndrome: a case–control study syndrome. Oral Dis. 19, 46–58 (2013).
Sjögren’s syndrome correlates with increased in 624 patients. Lupus 19, 941–948 (2010). 76. Akpek, E. K. et al. Ocular and systemic morbidity
inflammation. Arthritis Rheum. 62, 969–977 (2010). 57. Karabulut, G. et al. Cigarette smoking in primary in a longitudinal cohort of Sjögren’s syndrome.
34. Jin, J.‑O., Shinohara, Y. & Yu, Q. Innate immune Sjögren’s syndrome: positive association only with Ophthalmology 122, 56–61 (2015).
signaling induces interleukin‑7 production from ANA positivity. Mod. Rheumatol. 21, 602–607 77. Albrecht, J. et al. Skin involvement and outcome
salivary gland cells and accelerates the development (2011). measures in systemic autoimmune diseases. Clin. Exp.
of primary Sjögren’s syndrome in a mouse model. 58. Manthorpe, R. et al. Lower frequency of focal lip Rheumatol. 24, S52–S59 (2006).
PLoS ONE 8, e77605 (2013). sialadenitis (focus score) in smoking patients. Can 78. Baldini, C. et al. Primary Sjögren’s syndrome as a
35. Jin, J.‑O., Kawai, T., Cha, S. & Yu, Q. Interleukin‑7 tobacco diminish the salivary gland involvement as multi-organ disease: impact of the serological profile
enhances the Th1 response to promote the judged by histological examination and anti-SSA/Ro on the clinical presentation of the disease in a large
development of Sjögren’s syndrome-like autoimmune and anti-SSB/La antibodies in Sjögren’s syndrome? cohort of Italian patients. Rheumatology 53,
exocrinopathy in mice. Arthritis Rheum. 65, Ann. Rheum. Dis. 59, 54–60 (2000). 839–844 (2013).
2132–2142 (2013). 59. Reveille, J. D., Wilson, R. W., Provost, T. T., Bias, W. B. 79. Quartuccio, L. et al. Validation of the classification
36. Alunno, A. et al. T regulatory and T helper 17 cells in & Arnett, F. C. Primary Sjögren’s syndrome and other criteria for cryoglobulinaemic vasculitis. Rheumatology
primary Sjögren’s syndrome: facts and perspectives. autoimmune diseases in families. Prevalence and (Oxford) 53, 2209–2213 (2014).
Mediators Inflamm. 2015, 243723 (2015). immunogenetic studies in six kindreds. Ann. Intern. 80. Retamozo, S. et al. Cryoglobulinaemic vasculitis at
37. Manganelli, P. & Fietta, P. Apoptosis and Sjögren Med. 101, 748–756 (1984). diagnosis predicts mortality in primary Sjögren
syndrome. Semin. Arthritis Rheum. 33, 49–65 60. Kuo, C.‑F. et al. Familial risk of Sjögren’s syndrome syndrome: analysis of 515 patients. Rheumatology
(2003). and co‑aggregation of autoimmune diseases in (Oxford) http://dx.doi.org/10.1093/rheumatology/
38. Okuma, A. et al. Enhanced apoptosis by disruption of affected families: a nationwide population study. kew194 (2016).
the STAT3–IκB‑ζ signaling pathway in epithelial cells Arthritis Rheumatol. 67, 1904–1912 (2015). 81. Seror, R. et al. EULAR Sjögren’s syndrome disease
induces Sjögren’s syndrome-like autoimmune disease. 61. Lessard, C. J. et al. Variants at multiple loci implicated activity index: development of a consensus systemic
Immunity 38, 450–460 (2013). in both innate and adaptive immune responses are disease activity index for primary Sjögren’s syndrome.
39. Lucchesi, D., Pitzalis, C. & Bombardieri, M. EBV and associated with Sjögren’s syndrome. Nat. Genet. 45, Ann. Rheum. Dis. 69, 1103–1109 (2010).
other viruses as triggers of tertiary lymphoid 1284–1292 (2013). This study has led to a step forwards in the
structures in primary Sjögren’s syndrome. Expert Rev. This is a key study about the complex genetic evaluation of systemic complications of SjS.
Clin. Immunol. 10, 445–455 (2014). landscape of SjS. 82. ter Borg, E. J. & Kelder, J. C. Lower prevalence
40. Wen, S. et al. Association of Epstein–Barr virus (EBV) 62. Li, Y. et al. A genome-wide association study in Han of extra-glandular manifestations and anti-SSB
with Sjögren’s syndrome: differential EBV expression Chinese identifies a susceptibility locus for primary antibodies in patients with primary Sjögren’s
between epithelial cells and lymphocytes in salivary Sjögren’s syndrome at 7q11.23. Nat. Genet. 45, syndrome and widespread pain: evidence for
glands. Am. J. Pathol. 149, 1511–1517 (1996). 1361–1365 (2013). a relatively benign subset. Clin. Exp. Rheumatol. 32,
41. Kivity, S. et al. Infection and autoimmunity in Sjögren’s 63. Gottenberg, J.‑E. et al. In primary Sjögren’s syndrome, 349–353 (2014).
syndrome: a clinical study and comprehensive review. HLA class II is associated exclusively with 83. Karageorgas, T. et al. Fatigue in primary Sjögren’s
J. Autoimmun. 51, 17–22 (2014). autoantibody production and spreading of the syndrome: clinical, laboratory, psychometric,
42. Biberfeld, P. et al. Human herpesvirus‑6 (HHV‑6, autoimmune response. Arthritis Rheumatol. 48, and biologic associations. Arthritis Care Res.
HBLV) in sarcoidosis and lymphoproliferative 2240–2245 (2003). (Hoboken) 68, 123–131 (2016).
disorders. J. Virol. Methods 21, 49–59 (1988). 64. Williams, P. H. et al. Horizons in Sjögren’s syndrome 84. Ramos-Casals, M. et al. Characterization of systemic
43. Baboonian, C., Venables, P. J., Maini, R. N., genetics. Clin. Rev. Allergy Immunol. 32, 201–209 disease in primary Sjögren’s syndrome: EULAR‑SS
Kangro, H. O. & Osman, H. K. Antibodies to human (2007). Task Force recommendations for articular, cutaneous,
herpesvirus‑6 in Sjögren’s syndrome. Arthritis Rheum. 65. Nocturne, G. & Mariette, X. Sjögren syndrome- pulmonary and renal involvements. Rheumatology
33, 1749–1750 (1990). associated lymphomas: an update on pathogenesis (Oxford) 54, 2230–2238 (2015).

18 | 2016 | VOLUME 2 www.nature.com/nrdp


© 2016 Macmillan Publishers Limited. All rights reserved
PRIMER

85. Riente, L. et al. Ultrasound imaging for the 107. Brito-Zerón, P. et al. Circulating monoclonal patient index for primary Sjögren’s syndrome.
rheumatologist. XXIII. Sonographic evaluation of hand immunoglobulins in Sjögren syndrome. Medicine Ann. Rheum. Dis. 70, 968–972 (2011).
joint involvement in primary Sjögren’s syndrome. (Baltimore) 84, 90–97 (2005). 127. Johnsen, S. J. et al. Evaluation of germinal center-like
Clin. Exp. Rheumatol. 27, 747–750 (2009). 108. Brito-Zerón, P. et al. Monoclonal gammopathy related structures and B cell clonality in patients with primary
86. Quartuccio, L. et al. Clinical and biological to Sjögren syndrome: a key marker of disease Sjögren syndrome with and without lymphoma.
differences between cryoglobulinaemic and prognosis and outcomes. J. Autoimmun. 39, 43–48 J. Rheumatol. 41, 2214–2222 (2014).
hypergammaglobulinaemic purpura in primary (2012). 128. Pollard, R. P. E. et al. Treatment of mucosa-associated
Sjögren’s syndrome: results of a large multicentre 109. Guellec, D. et al. Diagnostic value of labial minor lymphoid tissue lymphoma in Sjögren’s syndrome:
study. Scand. J. Rheumatol. 44, 36–41 (2015). salivary gland biopsy for Sjögren’s syndrome: a retrospective clinical study. J. Rheumatol. 38,
87. Chen, M. H. et al. Lung involvement in primary a systematic review. Autoimmun. Rev. 12, 416–420 2198–2208 (2011).
Sjögren’s syndrome: correlation between high- (2013). 129. Zero, D. T. et al. Clinical practice guidelines for oral
resolution computed tomography score and mortality. 110. Daniels, T. E. et al. Associations between salivary management of Sjögren disease: dental caries
J. Chin. Med. Assoc. 77, 75–82 (2014). gland histopathologic diagnoses and phenotypic prevention. J. Am. Dent. Assoc. 147, 295–305
88. Goules, A. V., Tatouli, I. P., Moutsopoulos, H. M. features of Sjögren’s syndrome among 1,726 registry (2016).
& Tzioufas, A. G. Clinically significant renal participants. Arthritis Rheum. 63, 2021–2030 130. Ramos-Casals, M., Brito-Zerón, P., Sisó-Almirall, A.,
involvement in primary Sjögren’s syndrome: clinical (2011). Bosch, X. & Tzioufas, A. G. Topical and systemic
presentation and outcome. Arthritis Rheum. 65, This is the largest study describing the different medications for the treatment of primary Sjögren’s
2945–2953 (2013). histopathological scenarios that are found in the syndrome. Nat. Rev. Rheumatol. 8, 399–411 (2012).
89. Ramos-Casals, M. et al. Systemic involvement in minor salivary glands of patients with SjS. 131. Ramos-Casals, M., Tzioufas, A. G., Stone, J. H.,
primary Sjögren’s syndrome evaluated by the 111. Pijpe, J. et al. Parotid gland biopsy compared with Siso, A. & Bosch, X. Treatment of primary Sjögren
EULAR‑SS Disease Activity Index: analysis of 921 labial biopsy in the diagnosis of patients with primary syndrome: a systematic review. JAMA 304, 452–460
Spanish patients (GEAS‑SS registry). Rheumatology Sjögren’s syndrome. Rheumatology (Oxford) 46, (2010).
(Oxford) 53, 321–331 (2014). 335–341 (2007). This is the first published systematic review that
90. Ramos-Casals, M., Tzioufas, A. G. & Font, J. Primary 112. Vered, M., Buchner, A., Haimovici, E., Hiss, Y. evaluated the use of the main topical and systemic
Sjögren’s syndrome: new clinical and therapeutic & Dayan, D. Focal lymphocytic infiltration in aging drugs for the treatment of SjS.
concepts. Ann. Rheum. Dis. 64, 347–354 (2005). human palatal salivary glands: a comparative study 132. Gottenberg, J.‑E. et al. Effects of hydroxychloroquine
91. Nocturne, G. et al. Rheumatoid factor and disease with labial salivary glands. J. Oral Pathol. Med. 30, on symptomatic improvement in primary Sjögren
activity are independent predictors of lymphoma in 7–11 (2001). syndrome: the JOQUER randomized clinical trial.
primary Sjögren’s syndrome. Arthritis Rheumatol. 68, 113. Radfar, L., Kleiner, D. E., Fox, P. C. & Pillemer, S. R. JAMA 312, 249–258 (2014).
977–985 (2016). Prevalence and clinical significance of lymphocytic foci 133. Kruize, A. A. et al. Hydroxychloroquine treatment for
92. Risselada, A. P. et al. The prognostic value of routinely in minor salivary glands of healthy volunteers. primary Sjögren’s syndrome: a two year double blind
performed minor salivary gland assessments in Arthritis Rheum. 47, 520–524 (2002). crossover trial. Ann. Rheum. Dis. 52, 360–364
primary Sjögren’s syndrome. Ann. Rheum. Dis. 73, 114. Risselada, A. P., Looije, M. F., Kruize, A. A., (1993).
1537–1540 (2014). Bijlsma, J. W. & van Roon, J. A. The role of ectopic 134. Dass, S. et al. Reduction of fatigue in Sjögren
93. Vinagre, F., Santos, M. J., Prata, A., da Silva, J. C. germinal centers in the immunopathology of primary syndrome with rituximab: results of a randomised,
& Santos, A. I. Assessment of salivary gland function Sjögren’s syndrome: a systematic review. double-blind, placebo-controlled pilot study.
in Sjögren’s syndrome: the role of salivary gland Semin. Arthritis Rheum. 42, 368–376 (2013). Ann. Rheum. Dis. 67, 1541–1544 (2008).
scintigraphy. Autoimmun. Rev. 8, 672–676 (2009). 115. Koh, J. H., Lee, J., Chung, S.‑H., Kwok, S.‑K. 135. Meijer, J. M. et al. Effectiveness of rituximab
94. Paugh, J. R. in Clinical Ocular Pharmacology & Park, S.‑H. Relation of autoimmune cytopenia treatment in primary Sjögren’s syndrome:
(eds Barlett, J. D. & Jaanus, S. D.) 283–294 to glandular and systemic manifestations in primary a randomized, double-blind, placebo-controlled trial.
(Butterworth Heinemann, 2007). Sjögren syndrome: analysis of 113 Korean patients. Arthritis Rheum. 62, 960–968 (2010).
95. Theander, E. & Mandl, T. Primary Sjögren’s syndrome: J. Rheumatol. 42, 1817–1824 (2015). 136. Devauchelle-Pensec, V. et al. Treatment of primary
diagnostic and prognostic value of salivary gland 116. Mandl, T., Bredberg, A., Jacobsson, L. T. H., Sjögren syndrome with rituximab: a randomized trial.
ultrasonography using a simplified scoring system. Manthorpe, R. & Henriksson, G. CD4+ Ann. Intern. Med. 160, 233–242 (2014).
Arthritis Care Res. (Hoboken) 66, 1102–1107 (2014). T‑lymphocytopenia — a frequent finding in anti-SSA 137. Pijpe, J. et al. Rituximab treatment in patients with
96. Fabini, G., Rutjes, S. A., Zimmermann, C., Pruijn, G. J. antibody seropositive patients with primary Sjögren’s primary Sjögren’s syndrome: an open-label phase II
& Steiner, G. Analysis of the molecular composition of syndrome. J. Rheumatol. 31, 726–728 (2004). study. Arthritis Rheum. 52, 2740–2750 (2005).
Ro ribonucleoprotein complexes. Identification of 117. Baimpa, E., Dahabreh, I. J., Voulgarelis, M. 138. Gottenberg, J.‑E. et al. Efficacy of rituximab in
novel Y RNA-binding proteins. Eur. J. Biochem. 267, & Moutsopoulos, H. M. Hematologic manifestations systemic manifestations of primary Sjögren’s
2778–2789 (2000). and predictors of lymphoma development in primary syndrome: results in 78 patients of the autoimmune
97. Mekinian, A., Nicaise-Roland, P., Chollet-Martin, S., Sjögren syndrome: clinical and pathophysiologic and rituximab registry. Ann. Rheum. Dis. 72,
Fain, O. & Crestani, B. Anti-SSA Ro52/Ro60 antibody aspects. Medicine (Baltimore) 88, 284–293 (2009). 1026–1031 (2013).
testing by immunodot could help the diagnosis of 118. Vitali, C. et al. Classification criteria for Sjögren’s 139. Carubbi, F. et al. Efficacy and safety of rituximab
Sjögren’s syndrome in the absence of anti-SSA/SSB syndrome: a revised version of the European criteria treatment in early primary Sjögren’s syndrome:
antibodies by ELISA. Rheumatology (Oxford) 52, proposed by the American–European Consensus a prospective, multi-center, follow‑up study.
2223–2228 (2013). Group. Ann. Rheum. Dis. 61, 554–558 (2002). Arthritis Res. Ther. 15, R172 (2013).
98. Schulte-Pelkum, J., Fritzler, M. & Mahler, M. This is the ‘gold-standard’ criteria used worldwide 140. Pijpe, J. et al. Clinical and histologic evidence of
Latest update on the Ro/SS‑A autoantibody system. for the diagnosis of SjS. salivary gland restoration supports the efficacy
Autoimmun. Rev. 8, 632–637 (2009). 119. Shiboski, S. C. et al. American College of of rituximab treatment in Sjögren’s syndrome.
99. Retamozo, S. et al. Anti‑Ro52 antibody testing Rheumatology classification criteria for Sjögren’s Arthritis Rheum. 60, 3251–3256 (2009).
influences the classification and clinical syndrome: a data-driven, expert consensus approach 141. Cornec, D. et al. Development of the Sjögren’s
characterisation of primary Sjögren’s syndrome. in the Sjögren’s International Collaborative Clinical Syndrome Responder Index, a data-driven composite
Clin. Exp. Rheumatol. 30, 686–692 (2012). Alliance cohort. Arthritis Care Res. (Hoboken) 64, endpoint for assessing treatment efficacy.
100. Ambrosi, A. & Wahren-Herlenius, M. Congenital heart 475–487 (2012). Rheumatology (Oxford) 54, 1699–1708 (2015).
block: evidence for a pathogenic role of maternal 120. Stone, J. H., Brito-Zerón, P., Bosch, X. 142. Steinfeld, S. D. et al. Epratuzumab (humanised
autoantibodies. Arthritis Res. Ther. 14, 208 (2012). & Ramos-Casals, M. Diagnostic approach to the anti‑CD22 antibody) in primary Sjögren’s syndrome:
101. Yang, Z., Liang, Y. & Zhong, R. Is identification of complexity of IgG4‑related disease. Mayo Clin. Proc. an open-label phase I/II study. Arthritis Res. Ther.
anti‑SSA and/or -SSB antibodies necessary in serum 90, 927–939 (2015). 8, R129 (2006).
samples referred for antinuclear antibodies testing? 121. Mavragani, C. P., Fragoulis, G. E., Rontogianni, D., 143. Mariette, X. et al. Efficacy and safety of belimumab
J. Clin. Lab. Anal. 26, 447–451 (2012). Kanariou, M. & Moutsopoulos, H. M. Elevated IgG4 in primary Sjögren’s syndrome: results of the BELISS
102. Quartuccio, L. et al. Anti-SSA/SSB-negative Sjögren’s serum levels among primary Sjögren’s syndrome open-label phase II study. Ann. Rheum. Dis. 74,
syndrome shows a lower prevalence of patients: do they unmask underlying IgG4‑related 526–531 (2015).
lymphoproliferative manifestations, and a lower risk disease? Arthritis Care Res. (Hoboken) 66, 773–777 144. Adler, S. et al. Evaluation of histological, serological
of lymphoma evolution. Autoimmun. Rev. 14, (2014). and clinical changes in response to abatacept
1019–1022 (2015). 122. Nocturne, G. et al. IgG4 multiorgan treatment of primary Sjögren’s syndrome: a pilot
103. Baer, A. N. et al. The SSB-positive/SSA-negative lymphoproliferative syndrome as a differential study. Arthritis Care Res. (Hoboken) 65, 1862–1868
antibody profile is not associated with key phenotypic diagnosis of primary Sjögren’s syndrome in men? (2013).
features of Sjögren’s syndrome. Ann. Rheum. Dis. 74, Ann. Rheum. Dis. 70, 2234–2235 (2011). 145. Meiners, P. M. et al. Abatacept treatment reduces
1557–1561 (2015). 123. Brito-Zerón, P., Izmirly, P. M., Ramos-Casals, M., disease activity in early primary Sjögren’s syndrome
104. Danda, D. et al. La positive, Ro60 negative subset Buyon, J. P. & Khamashta, M. A. The clinical spectrum (open-label proof of concept ASAP study).
of primary Sjögren’s syndrome is a reality. ACR http:// of autoimmune congenital heart block. Nat. Rev. Ann. Rheum. Dis. 73, 1393–1396 (2014).
acrabstracts.org/abstract/la-positive-ro60-negative- Rheumatol. 11, 301–312 (2015). 146. US National Library of Medicine. Efficacy and safety of
subset-of-primary-sjogrens-syndrome-is-a-reality/ 124. Ramos, M. et al. [Primary Sjögren’s syndrome: clinical abatacept in patients with primary Sjögren’s syndrome
(2014). and immunologic study of 80 patients]. Med. Clin. (ASAPIII). ClinicalTrials.gov. https://clinicaltrials.gov/
105. Ramos-Casals, M., Stone, J. H., Cid, M. C. & Bosch, X. (Barc.) 108, 652–657 (in Spanish) (1997). ct2/show/NCT02067910 (2016).
The cryoglobulinaemias. Lancet 379, 348–360 125. Seror, R. et al. Validation of EULAR primary Sjögren’s 147. Rummel, M. J. et al. Bendamustine plus rituximab
(2012). syndrome disease activity (ESSDAI) and patient versus CHOP plus rituximab as first-line treatment
106. Quartuccio, L. et al. Biomarkers of lymphoma in indexes (ESSPRI). Ann. Rheum. Dis. 74, 859–866 for patients with indolent and mantle-cell
Sjögren’s syndrome and evaluation of the lymphoma (2015). lymphomas: an open-label, multicentre, randomised,
risk in prelymphomatous conditions: results of a 126. Seror, R. et al. EULAR Sjögren’s Syndrome Patient phase 3 non‑inferiority trial. Lancet 381,
multicenter study. J. Autoimmun. 51, 75–80 (2014). Reported Index (ESSPRI): development of a consensus 1203–1210 (2013).

NATURE REVIEWS | DISEASE PRIMERS VOLUME 2 | 2016 | 19


© 2016 Macmillan Publishers Limited. All rights reserved
PRIMER

148. Voulgarelis, M., Giannouli, S., Tzioufas, A. G. 166. Szymula, A. et al. T cell epitope mimicry between 184. Hackett, K. L. et al. A systematic review of non-
& Moutsopoulos, H. M. Long term remission of Sjögren’s syndrome antigen A (SSA)/Ro60 and oral, pharmacological interventions for primary Sjögren’s
Sjögren’s syndrome associated aggressive B cell gut, skin and vaginal bacteria. Clin. Immunol. 152, syndrome. Rheumatology (Oxford) 54, 2025–2032
non‑Hodgkin’s lymphomas following combined B cell 1–9 (2014). (2015).
depletion therapy and CHOP (cyclophosphamide, 167. Chaigne, B. et al. Primary Sjögren’s syndrome and 185. Ma, C. S., Deenick, E. K., Batten, M. & Tangye, S. G.
doxorubicin, vincristine, prednisone). Ann. Rheum. Dis. occupational risk factors: a case–control study. The origins, function, and regulation of T follicular
65, 1033–1037 (2006). J. Autoimmun. 60, 80–85 (2015). helper cells. J. Exp. Med. 209, 1241–1253 (2012).
149. Bowman, S. J. et al. Measurement of fatigue and 168. Webber, M. P. et al. Nested case–control study of 186. US National Library of Medicine. Efficacy of
discomfort in primary Sjögren’s syndrome using a new selected systemic autoimmune diseases in World Trade tocilizumab in primary Sjögren’s syndrome. (ETAP).
questionnaire tool. Rheumatology 43, 758–764 Center rescue/recovery workers. Arthritis Rheumatol. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/
(2004). 67, 1369–1376 (2015). NCT01782235 (2014).
150. Inal, V., Kitapcioglu, G., Karabulut, G., Keser, G. 169. Bernatsky, S. et al. Fine particulate air pollution, 187. Xie, J. H. et al. Engineering of a novel anti‑CD40L
& Kabasakal, Y. Evaluation of quality of life in relation nitrogen dioxide, and systemic autoimmune rheumatic domain antibody for treatment of autoimmune
to anxiety and depression in primary Sjögren’s disease in Calgary, Alberta. Environ. Res. 140, diseases. J. Immunol. 192, 4083–4092 (2014).
syndrome. Mod. Rheumatol. 20, 588–597 (2010). 474–478 (2015). 188. Signoriello, E., Sagliocchi, A., Fratta, M. & Lus, G.
151. Hyphantis, T., Mantis, D., Voulgari, P. V., Tsifetaki, N. 170. Rose-Nussbaumer, J. et al. Inter-grader agreement Fingolimod efficacy in multiple sclerosis associated
& Drosos, A. A. The psychological defensive profile of of the Ocular Staining Score in the Sjögren’s with Sjögren syndrome. Acta Neurol. Scand. 131,
primary Sjögren’s syndrome patients and its International Clinical Collaborative Alliance (SICCA) 140–143 (2015).
relationship to health-related quality of life. Clin. Exp. Registry. Am. J. Ophthalmol. 160, 1150–1153.e3 189. Jakez-Ocampo, J., Atisha-Fregoso, Y. & Llorente, L.
Rheumatol. 29, 485–493 (2011). (2015). Refractory primary Sjögren syndrome successfully
152. Thomas, E., Hay, E. M., Hajeer, A. & Silman, A. J. 171. Kaido, M. et al. Corneal fluorescein staining correlates treated with bortezomib. J. Clin. Rheumatol. 21,
Sjögren’s syndrome: a community-based study with visual function in dry eye patients. 31–32 (2015).
of prevalence and impact. Br. J. Rheumatol. 37, Invest. Ophthalmol. Vis. Sci. 52, 9516–9522 (2011). 190. Oni, C. et al. Eligibility for clinical trials in primary
1069–1076 (1998). 172. López-Miguel, A. et al. Clinical and molecular Sjögren’s syndrome: lessons from the UK Primary
153. Segal, B. et al. Primary Sjögren’s syndrome: health inflammatory response in Sjögren syndrome- Sjögren’s Syndrome Registry. Rheumatology (Oxford)
experiences and predictors of health quality among associated dry eye patients under desiccating stress. 55, 544–552 (2016).
patients in the United States. Health Qual. Life Am. J. Ophthalmol. 161, 133–141.e2 (2016). 191. Devauchelle-Pensec, V. et al. Which and how many
Outcomes 7, 46 (2009). 173. Seror, R. et al. Defining disease activity states and patients should be included in randomised controlled
154. Meijer, J. M. et al. Health-related quality of life, clinically meaningful improvement in primary trials to demonstrate the efficacy of biologics in primary
employment and disability in patients with Sjögren’s Sjögren’s syndrome with EULAR primary Sjögren’s Sjögren’s syndrome? PLoS ONE 10, e0133907 (2015).
syndrome. Rheumatology (Oxford) 48, 1077–1082 syndrome disease activity (ESSDAI) and patient- 192. Baum, B. J., Alevizos, I., Chiorini, J. A., Cotrim, A. P.
(2009). reported indexes (ESSPRI). Ann. Rheum. Dis. 75, & Zheng, C. Advances in salivary gland gene therapy
155. Champey, J. et al. Quality of life and psychological 382–389 (2016). — oral and systemic implications. Expert Opin. Biol.
status in patients with primary Sjögren’s syndrome 174. Seror, R. et al. Development of clinESSDAI score Ther. 15, 1443–1454 (2015).
and sicca symptoms without autoimmune features. (clinical EULAR Sjögren’s Syndrome Disease Activity 193. Gao, Z. et al. Generation of bioartificial salivary gland
Arthritis Rheum. 55, 451–457 (2006). Index) without the biological domain: a tool for using whole-organ decellularized bioscaffold. Cells
156. Lendrem, D. et al. Do the European League Against biological studies. Ann. Rheum. Dis. http://dx.doi.org/ Tissues Organs 200, 171–180 (2014).
Rheumatism Sjögren’s syndrome outcome measures 10.1136/annrheumdis-2015-208504 (2016). 194. Tzioufas, A. G. & Kapsogeorgou, E. K. Biomarkers.
correlate with health status in primary Sjögren’s 175. Ren, Y.‑D. et al. Conventional MRI techniques Saliva proteomics is a promising tool to study Sjögren
syndrome? Rheumatology (Oxford) 54, 655–659 combined with MR sialography on T2‑3D‑DRIVE syndrome. Nat. Rev. Rheumatol. 11, 202–203 (2015).
(2015). in Sjögren syndrome. Int. J. Clin. Exp. Med. 8, 195. Delaleu, N. et al. High fidelity between saliva
157. Baer, A. N., Petri, M., Sohn, J., Rosen, A. 3974–3982 (2015). proteomics and the biologic state of salivary glands
& Casciola-Rosen, L. Association of antibodies 176. Birnbaum, J. et al. Use of a novel high-resolution defines biomarker signatures for primary Sjögren’s
to interferon-inducible protein‑16 with markers magnetic resonance neurography protocol to detect syndrome. Arthritis Rheumatol. 67, 1084–1095
of more severe disease in primary Sjögren’s syndrome. abnormal dorsal root ganglia in Sjögren patients with (2015).
Arthritis Care Res. (Hoboken) 68, 254–260 (2016). neuropathic pain: case series of 10 patients and 196. Le Gall, M. et al. A prospective evaluation of dental
158. Lim, S. A. et al. Association of IL‑21 cytokine with review of the literature. Medicine (Baltimore) 93, and periodontal status in patients with suspected
severity of primary Sjögren syndrome dry eye. 121–134 (2014). Sjögren’s syndrome. Joint Bone Spine 83, 235–236
Cornea 34, 248–252 (2015). 177. Cohen, C. et al. 18F‑fluorodeoxyglucose positron (2016).
159. Iwasa, A. et al. Aromatase controls Sjögren syndrome- emission tomography/computer tomography as an 197. Priori, R. et al. Quality of sexual life in women
like lesions through monocyte chemotactic protein‑1 objective tool for assessing disease activity in Sjögren’s with primary Sjögren syndrome. J. Rheumatol. 42,
in target organ and adipose tissue-associated syndrome. Autoimmun. Rev. 12, 1109–1114 (2013). 1427–1431 (2015).
macrophages. Am. J. Pathol. 185, 151–161 (2015). 178. Hammenfors, D. S., Brun, J. G., Jonsson, R. 198. van Nimwegen, J. F. et al. The impact of primary
160. Konsta, O. D. et al. The contribution of epigenetics & Jonsson, M. V. Diagnostic utility of major salivary Sjögren’s syndrome on female sexual function.
in Sjögren’s syndrome. Front. Genet. 5, 71 (2014). gland ultrasonography in primary Sjögren’s syndrome. Rheumatology 54, 1286–1293 (2015).
161. Miceli-Richard, C. et al. Overlap between differentially Clin. Exp. Rheumatol. 33, 56–62 (2015). 199. Tanner, K. et al. The quality of life burden associated
methylated DNA regions in blood B lymphocytes and 179. Baldini, C. et al. Salivary gland ultrasonography: with voice disorders in Sjögren’s syndrome. Ann. Otol.
genetic at‑risk loci in primary Sjögren’s syndrome. a highly specific tool for the early diagnosis of primary Rhinol. Laryngol. 124, 721–727 (2015).
Ann. Rheum. Dis. 75, 933–940 (2016). Sjögren’s syndrome. Arthritis Res. Ther. 17, 146 200. Devauchelle-Pensec, V. et al. Effects of rituximab
162. Fiorentino, D. F. et al. PUF60: a prominent new target (2015). therapy on quality of life in patients with primary
of the autoimmune response in dermatomyositis and 180. Ramos-Casals, M., Brito-Zerón, P. & Font, J. The Sjögren’s syndrome. Clin. Exp. Rheumatol. 29, 6–12
Sjögren’s syndrome. Ann. Rheum. Dis. 75, 1145–1151 overlap of Sjögren’s syndrome with other systemic (2011).
(2016). autoimmune diseases. Semin. Arthritis Rheum. 36,
163. Bergum, B. et al. Antibodies against carbamylated 246–255 (2007). Acknowledgements
proteins are present in primary Sjögren’s syndrome 181. Xu, Y. et al. The prevalence and clinical characteristics The authors thank L. Alós, Pathology Department, Hospital
and are associated with disease severity. of primary Sjögren’s syndrome patients with lung Clínic, IDIBAPS, Barcelona, Spain, for her assistance in
Ann. Rheum. Dis. http://dx.doi.org/10.1136/ cancer: an analysis of ten cases in China and literature preparing the histopathological images.
annrheumdis-2015-207751 (2015). review. Thorac Cancer 6, 475–479 (2015).
164. Seror, R. et al. Low numbers of blood and salivary 182. Juarez, M. et al. Cardiovascular risk factors in women Author contributions
natural killer cells are associated with a better with primary Sjögren’s syndrome: United Kingdom Introduction (P.B.‑Z.); Epidemiology (M.R.‑C.); Mechanisms/
response to belimumab in primary Sjögren’s primary Sjögren’s syndrome registry results. pathophysiology (A.T., K.S. and R.J.); Diagnosis, screening
syndrome: results of the BELISS study. Arthritis Res. Arthritis Care Res. (Hoboken) 66, 757–764 (2014). and prevention (C.B. and P.B.‑Z.); Management (E.T., H.B. and
Ther. 17, 241 (2015). 183. Atisha-Fregoso, Y., Rivera-Vicencio, Y., X.M.); Quality of life (S.J.B.); Outlook (M.R.‑C.); Overview of
165. Terzulli, M., Ruiz, L. C., Kugadas, A., Masli, S. & Baños-Pelaez, M. & Hernández-Molina, G. Main Primer (M.R.‑C.).
Gadjeva, M. TSP‑1 deficiency alters ocular microbiota: causes and risk factors for hospitalisation in patients
implications for Sjögren’s syndrome pathogenesis. with primary Sjögren’s syndrome. Clin. Exp. Competing interests statement
J. Ocul. Pharmacol. Ther. 31, 413–418 (2015). Rheumatol. 33, 721–725 (2015). The authors declare no competing interests.

20 | 2016 | VOLUME 2 www.nature.com/nrdp


© 2016 Macmillan Publishers Limited. All rights reserved

You might also like