Download as pdf or txt
Download as pdf or txt
You are on page 1of 16

746319

research-article2018
TAB0010.1177/1759720X17746319Therapeutic Advances in Musculoskeletal DiseaseN Del Papa and C Vitali

Therapeutic Advances in Musculoskeletal Disease Review

Management of primary Sjögren’s


Ther Adv Musculoskel Dis

2018, Vol. 10(2) 39­–54

syndrome: recent developments and DOI: 10.1177/


https://doi.org/10.1177/1759720X17746319
https://doi.org/10.1177/1759720X17746319
1759720X17746319

new classification criteria


© The Author(s), 2018.
Reprints and permissions:
http://www.sagepub.co.uk/
journalsPermissions.nav

Nicoletta Del Papa and Claudio Vitali

Abstract:  For many years primary Sjögren’s syndrome (pSS) has been considered an orphan
disease, since no specific therapies were recognized as being capable of contrasting the
development and progression of this disorder. The treatment of oral and ocular features,
as well as of the systemic organ involvement, has been entrusted to the joint management
of different subspecialty physicians, like ophthalmologists, otolaryngologists, dentists
and rheumatologists. These latter subspecialty doctors are usually more involved in the
treatment of systemic extraglandular involvement and, to do it, they have long been using
the conventional therapies borrowed by the treatment schedules adopted in other systemic
autoimmune diseases. The increasing knowledge of the biological pathways that are operative
in patients with pSS, and the parallel development of molecular biology technology, have
allowed the production and availability of a number of biological agents able to positively
act on different disease mechanisms, and thus are candidates for testing in therapeutic
trials. Meanwhile, the scientific community has made a great effort to develop new accurate
and validated classification criteria and outcome measures to be applied in the selection of
patients to be included and monitored in therapeutic studies.
Some of the new-generation biotechnological agents have been tested in a number of open-
label and randomized controlled trials that have produced in many cases inconclusive or
contradictory results. Behind the differences in trial protocols, adopted outcome measures
and predefined endpoints, reasons for such unsatisfactory results can be found in the
large heterogeneity of clinical subtypes in the examined cohorts. The future challenge for a
substantial advancement in the therapeutic approach to pSS could be to identify the pathologic
mechanisms, outcome tools and biomarkers that characterize the different subsets of the
disease in order to test carefully selected target therapies with the highest probability of
success in each different clinical phenotype.

Keywords:  Sjögren’s syndrome, clinical subsetting, classification criteria, outcome measures,


pathogenetic mechanisms, biological therapies

Received: 12 September 2017; revised manuscript accepted: 12 November 2017

Correspondence to:
Nicoletta Del Papa
Introduction from signs and symptoms arising from the iso- Day Hospital of
Sjögren’s syndrome (SS) is a chronic autoim- lated involvement of salivary and lachrymal Rheumatology,
Department of
mune disorder whose typical manifestations are glands, or more rarely of other exocrine glands, Rheumatology, ASST G.
oral and ocular dryness. SS is defined as primary to manifestations derived from the involvement Pini-CTO, via Pini 3, 20122
Milan, Italy
(p) when it presents alone and secondary when of different organs or systems. The ‘so-called’ delpapa@gpini.it
associated with other diseases, such as rheuma- systemic extraglandular involvement is present in Claudio Vitali
toid arthritis (RA) and systemic lupus erythema- approximately 60% of patients with pSS. The Villa San Giuseppe, Istituto
S. Stefano, Como, Italy
tosus (SLE).1,2 In both variants of the disease, extraglandular features can be classified as perie-
the sicca complaints are the result of mononu- pithelial when driven by lymphocytic infiltration
clear infiltration of the salivary and lacrimal around epithelial tissues of parenchymal organs
glands. The clinical spectrum of pSS may vary such as lung, kidney and liver, or mediated by the

journals.sagepub.com/home/tab 39
Therapeutic Advances in Musculoskeletal Disease 10(2)

Figure 1.  Schematic representation of the heterogeneity of the clinical, serological and histopathological
features in patients with Sjögren’s syndrome (pSS). The pattern of expression in the subgroups with limited
glandular manifestations (on the left) and immune-complex-mediated extra-glandular features (on the
right) are certainly better known than those of patients with periepithelial lesions (in the middle). It may
be supposed that this latter subgroup may have intermediate clinical and pathologic features and present
specific serological markers.2 BAFF, B-cell activating factor; GC, germinal centre; IC, immune complex; IFN,
interferon; Th17, T helper 17; TI, tubulo-interstitial; Abs, antibodies.

microvascular deposition of immune complexes subsets of patients with this disorder. The patho-
that may cause purpura, glomerulonephritis and genetic mechanisms underlying this feature are
peripheral neuropathy.2 Similarly to what hap- still far from being completely understood.3
pens in other autoimmune systemic disorders, in Severe fatigue has been described in approxi-
a relevant number of cases, pSS is characterized mately one third of the patients and found to be
by B-cell hyperactivity and consequently by closely associated with arthralgias/myalgias, wide-
hypergammaglobulinaemia and production of spread pain assuming the clinical aspect of fibro-
autoantibodies, namely rheumatoid factor (RF) myalgia, anxiety, depression and impaired sleep
and anti-Ro and anti-La antibodies (see Figure patterns.3 Unexpectedly, the levels of proinflam-
1).1,2 matory cytokines are inversely related to patient-
reported levels of fatigue.4 These data may help to
The fact that SS can present such a wide spec- explain why treating inflammation does not nec-
trum of clinical features suggests that variable essarily improve fatigue in patients with pSS and
pathogenetic mechanisms may be operative in the may raise doubts about the previously formulated
different subsets of patients. This hypothesis is hypothesis that proinflammatory cytokines
supported by the findings that mononuclear cell directly mediate fatigue in chronic immunological
amount and organization as well as T/B-cell ratio conditions. In addition, some data suggest that
in the infiltrates of target tissues, gammaglobulin widespread pain, which is closely related to
and antibody levels in the serum, cytokine expres- fatigue, is predominant in seronegative patients
sion in both peripheral blood and glands, can be without extraglandular involvement.5
completely different in patients with the disease
limited to exocrine gland aggression or character- On the opposite site, the presence of immune-
ized by extraglandular features (see Figure 1). complex-mediated features, together with salivary
Even the impact of fatigue, which represents one gland enlargement, cryoglobulinemia, hypocom-
of the major contributors to the impaired quality plementemia, lymphopenia, germinal centre-like
of life in patients with pSS, is variable in different organization of mononuclear ­ infiltrates in the

40 journals.sagepub.com/home/tab
N Del Papa and C Vitali

salivary glands, RF positivity, hypergammaglob- Proinflammatory cytokines. It has been widely


ulinemia with monoclonal component charac- documented that some proinflammatory cyto-
terize the patients with more severe extraglandular kines, such as interleukin (IL)-1, tumour necrosis
manifestations, highest levels of disease activity factor (TNF)-α and IL-6 are upregulated in sali-
and high risk to develop lymphoma in the course vary gland tissue obtained from patients with
of pSS.6–8 pSS.14 IL-6 certainly plays an important role in the
differentiation of extrafollicular Th cells, germinal
Substantial progress has been made in recent years centre B cells and plasma cells. IL-6 levels were
in the knowledge of the main biological pathways also found to be increased in serum, saliva and
that are activated in the disease course. Additionally, tears of patients with pSS. Salivary gland epithelial
the advances in molecular biology have opened up cells from patients with this disorder have been
the possibility of developing specific target thera- shown to produce IL-6.15 The mechanisms of
pies that are potentially able to interfere with or action of IL-6 are now better known and may open
block the main cells or biological molecules that up new therapeutic possibilities for the treatment
are believed to play fundamental roles in SS patho- of pSS. IL-6 binding to its specific receptor leads
genesis. This has enhanced international efforts to to homodimerization of the receptor component
conduct randomized controlled studies in the set- gp130, which mainly results in the activation of the
ting of pSS. To do so, reliable and accurate classi- Janus kinase 1 (JAK1), and the subsequent phos-
fication criteria have been developed to select phorylation of the signal transducer and activator
homogeneous groups of patient to be enrolled in of transcription 3 (STAT3).16 STAT3 plays a cen-
clinical trials. Moreover, validated and sensitive tral role in transmitting cytokine signals to the
outcome measures have been constructed with the nucleus and promoting cell proliferation and anti-
purpose of accurately assessing the results of any apoptotic signals.17,18 Furthermore, the JAK1/
innovative therapeutic intervention. STAT3 pathway might be involved in other func-
tions, such as increased expression of IFNγ in Th1
cells, and of IL-17 in Th17 cells, respectively.19
Main pathogenetic mechanisms of SS

Innate immunity mechanisms Adaptive immunity mechanisms


Interferons. Several studies in both peripheral Antigen presentation.  Cathepsin S is an endopro-
blood9 and minor salivary gland (MSG) tis- tease located in the lysosomes of antigen-present-
sues10,11 from patients with pSS have shown an ing cells. It is involved in controlling autoantigen
upregulation of genes induced by type I interferon presentation to CD4+ T cells by interfering with
(IFNα and β), type II IFN (IFNγ) or both. This the binding of major histocompatibility complex
phenomenon has been defined as the IFN signa- II molecules.20 Increased levels of cathepsin S
ture and it is shared by other systemic autoim- have been found in the tears of patients with SS.21
mune disorders like SLE. Plasmacytoid dendritic
cells are considered the main source of IFNα,11 Costimulation.  Costimulation is a central event in
while T helper 1 (Th1) CD4+ cells and natural the perpetuation and amplification of autoim-
killer (NK) cells are believed to be the main IFNγ mune response; B7 (CD80 and CD86) molecules
producers in pSS salivary gland tissues.10 Type I expressed on classic antigen-presenting cells play
IFN signature seems to be prevalent in the periph- a critical role in enhancing immune responses by
eral blood of patients with SS whereas type II providing activation signals to T cells, through
IFN signature appears to be predominant in SS their link to the CD28 receptor. The cytotoxic T
salivary gland biopsies.12 lymphocyte antigen 4 (CTLA4) molecule is the
counterpart of this system, competing with CD28
Once produced, probably after a local infection of for the link with B7 molecules, and thus has an
still unidentified viruses and the consequent acti- inhibitory effect on the costimulatory system. The
vation of some toll-like receptors, type I IFNs second type of costimulatory molecule is repre-
induce apoptosis of salivary epithelial cells, expo- sented by the interaction of the CD40 molecule
sure of endogenous autoantigens to the immune on B cells with the CD40 ligand on antigen-pre-
system, upregulation of B-cell activating factor senting cells, T cells, endothelial and epithelial
(BAFF) expression, increased B-cell survival and cells, and B cells. In pSS, CD80, CD86 and
differentiation, and ultimately autoantibody pro- CD40 molecules were found to be expressed on
duction and immune complex formation.13 salivary gland epithelial cells, indicating that these

journals.sagepub.com/home/tab 41
Therapeutic Advances in Musculoskeletal Disease 10(2)

costimulatory systems are locally activated.22 The these kinases has recently been licensed for the
inhibition of the costimulatory signals is consid- treatment of chronic lymphocytic leukemia.28
ered crucial for the treatment of autoimmune Recent data support activation of the PI3Kδ
diseases. pathway in affected salivary gland tissues from an
animal model of SS.29 Moreover, BTK levels were
B-cell activation.  B-cell activation is considered shown to be increased in circulating B cells of a
a key pathogenetic mechanism of SS. Hyper- significant percentage of patients with pSS, in
gammaglobulinemia, formation of germinal cen- association with high serum RF levels.30 Even
tre-like structures in the infiltrated salivary gland spleen tyrosine kinase (Syk) seems to have an
tissue, production of RF and autoantibody important role in inducing B-cell differentiation
directed against ribonucleoproteins are the bio- and proliferation after the engagement of BCR in
logical phenomena related to the increased lymphoma and autoimmune disease such as sys-
B-cell presence and activation.23 Although the temic lupus and pSS.31
pathways influencing B-cell activation in pSS
remain partially unknown, many studies have CD22 is a type I transmembrane protein
indicated that a number of genes are involved in expressed on most mature B lymphocytes. It acts
B-cell survival, proliferation and signalling.24 as a coreceptor of BCR and its engagement by
The increased number of B cells among the specific reactants, such as high-affinity monoclo-
mononuclear cells infiltrating salivary gland tis- nal antibodies, may induce an intensification of
sue, namely in the germinal centre-like struc- the normal inhibitory role of CD22 on the BCR,
tures, suggests the antigen-driven nature of the leading to reduced signalling and diminished acti-
local immune response. vation of B cells.32

BAFF/BAFF receptor interaction. The B-cell acti- Ectopic germinal centre formation.  The formation of
vating factor (BAFF) axis comprises two ligands, ectopic germinal centres is increasingly recognized
which are BAFF itself (named also Blys) and the as a crucial mechanism in the pathogenesis of SS
‘a proliferating-inducing ligand’ (APRIL), and and other systemic autoimmune diseases. Germinal
three related receptors (BCMA, TACI, BR3) with centre formation in the salivary glands of patients
different affinity for the two ligands.25 BAFF is a with pSS is associated with higher focus scores, RF
member of the TNF family that acts by prolong- positivity and anti-Ro/La positivity, the presence of
ing the survival of circulating B cells. Its binding extraglandular manifestations, and it is considered
to the corresponding receptors inhibits intracel- as a marker of lymphoma development.33,34 An
lular apoptotic pathways and provides survival increased number of CD4-T follicular helper cells
signals for B cells.26 High BAFF levels have been and follicular dendritic cells have been found in the
found in patients with pSS, and have been shown salivary glands of patients with SS. These cells prob-
to be associated with the specific immunological ably participate in the germinal-like centre organiza-
markers of the disease, systemic disease activity tion and may drive B-cell differentiation towards
and lymphoproliferative features, such as mono- memory B cells and plasma cells.35,36
clonal lymphocytic selection, myoepithelial sial-
adenitis and lymphoma. In addition, it has been Finally, it has recently been demonstrated that a
suggested that the abnormal BAFF signal found large number of biological effector molecules may
in B cells infiltrating the salivary glands can play a take part in the formation and maintenance of
key role in the formation of ectopic germinal cen- germinal-centre-like structures. These biological
tres,14 survival of self-reactive B cells, and their mediators include some cytokines, such as IL-21
localization to follicle/marginal zone niches.27 and IL-22, chemokines CCL19 and CCL21, and
chemokine receptors CXCL12 and CXCL13.37,38
B-cell receptor (BCR) signalling. BCR activation The fact that Th17 cells (a CD4+ Th subtype
is another mechanism that is able to induce B-cell implicated in the pathogenesis of SS) are a major
survival, proliferation, functional differentiation source of IL-21 and IL-22 seems to provide a
and migration. These findings may suggest new rationale for targeting these cells in future thera-
rationale ways for targeting B-cell signalling in peutic interventions.39 Some data suggest that a
pSS. Some kinases, such as phosphatydil-inositol synergistic role in this pathologic network may be
3-kinase delta isoform (PI3Kδ), and Bruton’s played by lymphotoxin β. Its receptor blockade in
tyrosine kinase (BTK), are crucial in B-cell recep- the NOD mouse model of SS led to reduction of
tor (BCR) signal transduction. The blockade of B-cell accumulation in lacrimal glands.40

42 journals.sagepub.com/home/tab
N Del Papa and C Vitali

Table 1.  The 2016 ACR/EULAR classification criteria for pSS.20

Consensus criteria items for the classification of pSS:


Abnormal unstimulated salivary flow rate* (⩽0.1 ml/min) (1 point)
Abnormal Schirmer’s test (<5 mm in 5 min) (1 point)
Abnormal findings with lissamine green or fluorescein staining  
⩾5 in Ocular Staining Score or ⩾4 in Van Bijsterveld Score (1 point)
Presence of anti-Ro/SSA antibodies (3 points)
Histological evidence of focal lymphocytic sialadenitis,$ with a focus score ⩾1 focus/4 mm2 (3 points)
(1 focus = 50 lymphocytes/4 mm2)
Diagnosis is considered established if score ⩾4 points, after application of inclusion and
exclusion criteria
Inclusion criteria:
 Dryness of eyes or mouth for at least 3 months, not explained otherwise (e.g. medications,  
infection)
Exclusion criteria:
  Status post head/neck radiation  
 HIV/AIDS  
 Sarcoidosis  
  Active infection with hepatitis C virus (PCR replication rate)  
  Amyloidosis, graft versus host disease, IgG4-related disease  
The lack of any other potentially associated disease is the key requirement for classification
as pSS
*The patient is asked to sit still, not to speak or to chew for 5–15 min; the saliva produced during this time is transferred
to a test tube and weighed.
$Biopsy: removal of 3–5 labial minor salivary glands from the lower lip under local anaesthesia; fixation of biopsy

material in formalin and HE staining.


ACR/EULAR, American College of Rheumatology/European League Against Rheumatism; AIDS, acquired
immunodeficiency syndrome; HE, hematoxylin and eosin stain; HIV, human immunodeficiency virus; IgG,
immunoglobulin G; PCR, polymerase chain reaction; pSS, primary Sjögren’s syndrome.

Classification criteria and outcome measure [verified by using the validated questionnaire
for SS previously included in the American European
Given the advances in the knowledge of the Consensus Group (AECG) classification crite-
pathogenetic mechanisms of SS, and the increas- ria],44 or to patients with at least one of the sys-
ing availability of biological agents that are able to temic features listed in ESSDAI (EULAR SS
target the activated pathways in the disorder, a Disease Activity Index).45 The criteria are based
growing interest has arisen in assessing the most on five objective items and the individuals are
effective way of conducting clinical trials. The classified as having pSS if they have a total score
2016 American College of Rheumatology/ of at least 4, derived from the weighted sum of
European League Against Rheumatism (ACR/ the five items. Anti-SSA/Ro antibody positivity
EULAR) classification criteria for pSS represent and focal lymphocytic sialadenitis with a focus
an important advancement in reaching an agree- score of at least 1 focus/4 mm2 of tissue have the
ment on the way to select patients to be recruited highest weight in the criteria set, each scoring
for clinical studies.41,42 three points (Table 1).

These criteria are the last step of a lengthy scien- The inclusion of the salivary gland ultrasono-
tific process that started a long time ago and has graphic (US) examination has been proposed
produced a plethora of different classification among the classification items. However, consen-
criteria sets.43 The new ACR/EULAR classifica- sual US procedures and validated US scoring sys-
tion criteria are applicable either to patients with tems are needed before considering this
at least one symptom of ocular or oral dryness possibility.46

journals.sagepub.com/home/tab 43
Therapeutic Advances in Musculoskeletal Disease 10(2)

With respect to the 2002 AECG criteria that have with at least moderate activity (ESSDAI ⩾5) are
represented the most used classification criteria in the best candidates to be included.49 However, it
the last 15 years,47 the exclusion criteria of the has been pointed out that patients with an
ACR/EULAR criteria have also been updated. ESSDAI greater than 13, and so with a well
Immunoglobulin G4 (IgG4)-related disease has known increased mortality risk, should be
been added and it has been established that hepa- excluded. A ESSDAI greater than 13 has also
titis C infection has to be confirmed by evidence been suggested as an additional risk factor for the
of viral genome in peripheral blood. Finally, pre- development of lymphoma in the following course
existing lymphoma has been removed from the of the disease.49,51
exclusion criteria.41,42
Similarly, in trials devoted to evaluating the effi-
The 2016 ACR/EULAR classification criteria cacy of any drug on subjective symptoms, it has
have again pointed out the central role of MSG been suggested that patients who have an unsatis-
biopsy and specific autoantibodies for pSS classi- factory symptom state corresponding to a ESSPRI
fication. Recently, consensus guidelines on how of at least 5 should be selected.49
to correctly perform a salivary gland biopsy and
the related histopathologic examination have A modified ESSDAI scoring system, the so-called
been released by an expert group for application ClinESSDAI, has been derived from the ESSDAI
in clinical research and in clinical trials.48 by exclusion of the biological domain.52 This new
scale for systemic disease activity was built to
In the last 5 years the EULAR task force on pSS eliminate problems of collinearity in trials in
has proposed and validated two new tools for which biomarkers included in the biological
assessing disease activity and patient-reported domain of the original ESSDAI were analysed as
outcomes. The ESSDAI45 is a multidomain scor- separate outcome measures. The psychometric
ing system in which each domain (constitutional, properties of ClinESSDAI, including reliability
lymphadenopathy, glandular, articular, cutane- and sensitivity to change, are similar to those of
ous, respiratory, renal, muscular, peripheral nerv- ESSDAI.52
ous system, central nervous system, haematological
and biological) can be separately scored. Each Other disease-specific indexes have been devel-
domain score is also stratified according to the oped for evaluation of subjective features of the
severity of the manifestations. The ESSDAI has patients, such as the Profile of Fatigue and
been validated and the minimal clinically signifi- Discomfort (PROFAD) and Sicca Symptoms
cant difference was defined as a reduction of the Inventory (SSI). In addition, nondisease-specific
activity score of at least three points.49 indices have been proposed and used in clinical
trials to assess the health-related quality of life
A second tool, the EULAR Sjögren’s syndrome and functional impairment (Short Form 36), as
patient-reported index (ESSPRI), has been cre- well as other aspects of disease burden such as
ated with the purpose of assessing the subjective anxiety, depression or sleep disorder.53
symptomatology of patients. The total score of
ESSPRI is defined as the mean score of three
Lykert’s numerical scales for dryness, pain and Management of sicca symptoms
fatigue.50 The minimal clinically significant dif- Therapy of sicca complaints is crucial to improve
ference has also been defined as an improvement the quality of life of patients with SS (see Table
of at least one point or 15% of the baseline 2). At the same time, this is a very difficult chal-
value.49 lenge for physicians since evidence-based treat-
ment options are few and most therapeutic
The ESSDAI and ESSPRI are increasingly being procedures are empirically based.
used as inclusion criteria and endpoints in clinical
trials evaluating innovative therapies for pSS. General measures such as air humidification of
Moreover, their use in clinical practice could also the environment, namely of the bedroom, caries
be recommended. prevention, and smoking cessation play an impor-
tant role. Treatment of dry eye requires a close
It has also been suggested that, in trials aimed at and constant collaboration with an ophthalmolo-
evaluating the effectiveness of any experimental gist. Various tear substitutes are available to treat
drug on patients with systemic features, those keratoconjunctivitis sicca. The composition of

44 journals.sagepub.com/home/tab
N Del Papa and C Vitali

Table 2.  Some recommended treatments for sicca symptoms in patients with primary Sjögren’s syndrome.

Sicca manifestation Therapeutic measure (grade of recommendation)


Keratoconjunctivitis sicca Tear substitutes (A)
Secretagogues: pilocarpine, cevimeline (A)
Cyclosporine A eye drops 0.1% (B)
Short-term topical corticosteroids (C)
Punctal plugs (C)
Oral dryness Patient education
Avoid drugs that promote xerostomia (A)
Topical fluorides for caries prevention (A)
Secretagogues: pilocarpine, cevimeline (A)
Saliva substitutes, sugar-free chewing gum and electrostimulation of
salivary glands (C)
Grades of Recommendation according to Centre for Evidence-based Medicine in Oxford:
(A) Evidence obtained from meta-analyses or at least one randomized controlled trial.
(B) Evidence from at least one well designed experimental study.
(C) Evidence from at least one well designed descriptive study or case–control studies.

the tear substitutes varies in accordance with the to prevent caries are also strongly recommended.
complex physiology of the pathogenesis of dry eye A multicentre RCT has shown that mild intraoral
which can be limited to lachrymal glands or may electrostimulation can alleviate the oral dryness
sometimes even compromise the meibomian and had no adverse effects.60
glands. Generally, the use of preservative-free eye
drops is preferred to avoid the development of In patients with moderate to severe oral dryness
local hypersensitivity phenomena. A randomized and with residual salivary-gland function, oral
controlled trial (RCT) has shown that local anti- muscarinic agonists, such as pilocarpine or
inflammatory treatment with cyclosporine A eye cevimeline, are the treatment of choice in the
drops can be effective.54 Pilocarpine and cemive- absence of contraindications.55,56 Commonly
line, which are effective in stimulating salivary reported adverse effects include sweating, warmth
flow, have also been proven to improve subjective and flushing sensation, increased urinary fre-
complaints and objective signs of dry eye.55,56 The quency, headache and abdominal discomfort.
use of punctal plugs to block the drainage system
is effective in increasing the residence time of
both residual and instilled tears on the ocular Systemic conventional treatment
surface.57 The decision to adopt systemic treatment, and
the choice of the specific treatment in pSS, is
Treatment of oral dryness and related manifesta- often driven by the level of disease activity and by
tions is usually conducted with the essential con- the specifically involved organ system. The few
tribution of dentists and otolaryngologists. In RCTs evaluating the use of conventional disease-
patients with pSS the decrease in saliva produc- modifying antirheumatic drugs (DMARDs) in
tion causes difficulty in swallowing dry foods, patients with pSS did not provide conclusive evi-
chewing and speaking, and increases susceptibil- dence supporting their efficacy.61,62 Thus, treat-
ity to multiple caries and oral infections such as ment strategies are frequently based on
dental caries, periodontal disease and oral can- experiences acquired in other autoimmune rheu-
didiasis.58 Patients with pSS should be educated matic diseases, such as SLE or RA. Following the
about the need for regular oral-health monitoring indications given for other connective tissue dis-
and care to prevent these infective processes. orders and in view of its favourable side-effect
profile, hydroxychloroquine (HCQ) is the drug of
Saliva substitutes, lubricating agents and mechan- choice for various mild to moderate systemic
ical stimulation by chewing sugar-free gum are manifestations, such as arthralgia, arthritis and
usually employed in patients with mild hypo- cutaneous lesions. Unfortunately, a RCT on pSS
sialia.59 Topical fluoride and fluoride toothpaste failed to demonstrate any difference between

journals.sagepub.com/home/tab 45
Therapeutic Advances in Musculoskeletal Disease 10(2)

Table 3.  Some recommended systemic traditional treatment in patients with primary Sjögren’s syndrome.

Parotid swelling Short-term oral corticosteroids (D)


Antibiotic treatment, if required (D)
Arthritis Hydroxychloroquine (C)
NSAIDs
Short-term oral/intraarticular corticosteroids (C)
Other DMARDs as with rheumatoid arthritis (C)
Interstitial lung disease Corticosteroids, oral or intravenous (C)
Cyclophosphamide for active alveolitis (C)
Pirfenidone, nintedanib (C)
Tubulointerstitial nephritis Potassium and bicarbonate replacement (D)
Glomerulonephritis Corticosteroids, oral or intravenous (C)
Cyclophosphamide
Mycofenolate mofetil
(according to specific nephrologic guidelines)
Peripheral neuropathy Gabapentinoids (D)
Corticosteroids
IVIg (D)
Cryoglobulinemic vasculitis Corticosteroids, plasmapheresis (C)
Grades of Recommendation according to Centre for Evidence-based Medicine in Oxford:
(A) Evidence obtained from meta-analyses or at least one randomized controlled trial.
(B) Evidence from at least one well designed experimental study.
(C) Evidence from at least one well designed descriptive study or case–control studies.
(D) Evidence from expert opinion.
DMARD, disease-modifying antirheumatic drug; IVIg, intravenous immunoglobulins; NSAID, nonsteroidal anti-
inflammatory drug.

HCQ and placebo treated patients regarding sicca hyperactivity is believed to be an essential patho-
complaints, pain and fatigue.63 However, this genetic moment. Rituximab was the first anti-B-
study had significant limitations, such as the low cell receptor monoclonal antibody approved to
disease activity of patients, the shortness of fol- treat B-cell lymphomas. It acts by blocking the
low-up time, and the use of nonvalidated end- CD20 B-lymphocytic receptor leading to B-cell
points. Treatment recommendations concerning depletion in peripheral blood by antibody- and
corticosteroid use and immunosuppressive drugs complement-dependent cytotoxicity, and by the
are summarized in Table 3. Briefly, arthralgia and activation of apoptotic pathways.65 In pSS B
arthritis are usually treated, as happens in other cells infiltrate the glandular tissue around the
inflammatory arthritides, with methotrexate or ductal epithelium with the cooperation of stro-
leflunomide, whereas severe organ manifestations mal cells and follicular dendritic cells that are
can be successfully treated with high-dose meth- essential for the organization of the germinal-
ylprednisolone and cyclophosphamide. Severe centre-like structures, mono-oligoclonal selec-
cryoglobulinaemic vasculitis may favourably tion and possibly lymphoproliferation. It has
respond to plasmapheresis. Finally, patients with been clearly shown that rituximab therapy
B-cell lymphoma should be referred to experi- induces a partial depletion of B cells in the sali-
enced haematologists for a correct therapeutic vary glands.66 Contemporaneous reduction of
approach that is usually based on lymphoma sub- T-follicular helper cells and salivary gland
type and stage, according to current guidelines on expression of IL-17 has also been reported.67
the treatment of hemato-oncological disorders.64 This could be ascribed to the fact that B-cell
reduction in the glands induces a reduction of
the B-cell production of some cytokines that
Biological target therapies: present data drive the differentiation and proliferation of
and future perspectives Th17 cell subtype.

B-cell targeted agents A number of open-label prospective studies have


Targeting B cells is certainly a rationale thera- evaluated rituximab effectiveness in patients with
peutic approach in diseases like SS, where B-cell pSS, while only a few RCTs have been completed

46 journals.sagepub.com/home/tab
N Del Papa and C Vitali

so far. A wide range of outcomes were assessed in B-cell activation signals was done in many of
these studies, including sicca features, fatigue, these studies, namely in the large RCTs per-
pain, systemic features, quality of life and lym- formed so far. Finally, the often existing discrep-
phoma development (Table 4). ancies between the severity of subjective dryness
complaints and the corresponding objective
The preliminary open-label studies and two pilot measurement of lachrymal and salivary flows can
controlled trials on rituximab therapy for pSS explain why rituximab was ineffective when only
demonstrated some improvement in some sub- sicca symptoms were chosen to be assessed as
jective or objective parameters, or both.68–73,76 In outcome measures.
spite of these encouraging results, two more
recent large RCTs74,75 failed to reach the estab- Other anti-CD20 monoclonal antibodies such as
lished primary endpoints, and thus did not con- ocrelizumab and obinutuzumab (humanized
firm the promising previous results, although B-cell-depleting agent), and ofatumumab (fully
some secondary endpoints were satisfied. A com- human B-cell-depleting agent) are now available
posite scoring system which included dryness, and have demonstrated an acceptable safety pro-
pain, fatigue and global health evaluation, some file in patients with B-cell lymphoma.79
objective diagnostic tests and ESSDAI score
developed in a post hoc analysis of one of the RCT Anti-BAFF therapy. Belimumab is a monoclonal
studies74 was found to be improved. In contrast to antibody interfering with soluble BAFF that has
the poor clinical results obtained in the open- been licensed for patients with SLE. A prospec-
label and controlled studies, it has been clearly tive 1-year open-label bicentric study (BELISS)
shown that rituximab therapy is effective in reduc- on the effectiveness of belimumab has been car-
ing the level of some biological markers of the dis- ried out in 30 patients with pSS.80 The primary
ease as IgM-RF, gammaglobulins and IgG, endpoint was a composite score which included
autoantibody levels and B-cell number in glandu- five items: at least a 30% reduction of VAS for
lar infiltrates.77 dryness, fatigue and pain, plus a 30% reduction of
VAS of the global physician evaluation on sys-
A number of reasons can be postulated to explain temic activity, plus a 25% reduction of selected
why rituximab treatment was ineffective in the biological markers. Two of the five considered
majority of the performed studies. The heteroge- items were improved in 60% of patients at the
neity of the populations in the studies, and of the 28-week follow up. Almost 90% of the 15 patients
outcome measures that were used, certainly rep- who were responders at week 28 also responded
resent critical points. Some partially successful at week 52.81 A post hoc analysis of the BELISS
studies were carried out in patients with a short population revealed that high type I IFN scores at
disease duration and in patients in whom a resid- baseline were associated with better response to
ual glandular function was preliminarily demon- treatment.82
strated.68,70 To recruit patients with a long disease
duration, or having a large variability of this CD20 and BAFF blockade.  Despite the demonstra-
parameter, may certainly lead to unsatisfactory tion that the BAFF/BAFF receptor axis is related
results. The same may happen when patients are to lymphoma development, some preliminary
selected with exhausted gland function in the case observations seem to indicate that lymphoprolif-
that the experimental therapy is administered to eration is resistant to belimumab alone.83 These
demonstrate improvement of salivary flow. The data, together with the evidence of an upregula-
effect of B-cell depletion therapy on some biologi- tion of BAFF levels following rituximab treat-
cal markers of B-cell activation and on the ment, have suggested the potential usefulness of
improvement of the ESSDAI score observed in combination therapy of belimumab with ritux-
the French registry study,78 where the choice of imab.84 A randomized trial is currently under way
rituximab treatment was preliminarily condi- in which the two target therapies are sequentially
tioned by the presence of clear signs of systemic administered.
involvement and high disease activity at baseline,
clearly indicates how this therapy can really be Anti-BCR signalling therapies. Small molecules
effective in this particular subgroup of patients. capable of inhibiting transduction signals after the
Unfortunately, the ESSDAI was not available activation of BCR, by blocking PI3TK, BTK and
when the oldest studies were made and no prese- Syk, are now available.28–30 RCTs have been
lection of patients according to the presence of started with these molecules in patients with pSS.

journals.sagepub.com/home/tab 47
Table 4.  Open-label and randomized controlled trials with rituximab in pSS.

Authors, years Study Pts RTX Follow- Salivary Ocular Dryness Fatigue Pain ESSDAI ESSPRI SF36
(Pts features) design (No.) regimen up flow tests VAS oral/ VAS/ VAS domains
RTX/C weeks USF/SSF Sch/ODT° ocular Scale
Pijpe68 (early Open label 8 A 12 =/+ =/+ +/= NA/+* NA NA NA + (BP/
group) 8/0 PF/Vit)
Pijpe68 (with Open label 7 A 12 =/= =/= =/= NA/= NA NA NA + (all)
MALT-L) 7/0
Devauchelle- Open label 16 B 36 =/NA =/NA + +/NA + NA NA + (all)
Pensec69 16/0 (general but PF=
Therapeutic Advances in Musculoskeletal Disease 10(2)

dryness)
Meiners70 Open label 28 C 60 NA/+ NA/NA +/NA NA/+* NA + + + (PF)
28/0
Carubbi71 Open label 41 C 120 +/NA +/NA +/NA +/NA + + + NA
19/22 +DMARDS
St Clair71 Open label 12 C 52 =/= =/NA +/= +/NA + NA NA + (Vit)
12/0
Dass72 RCT (pilot) 30 C 26 =/NA =/NA NA +/+$ = NA NA +
20/10 (SF)
Meijer73 RCT 30 C 48 =/+ =/+ +/+ NA/+$ NA NA NA +
20/10
Devauchelle- RCT 120 C 24 =/NA =/NA + + = NA NA =
Pensec74 63/57 (general
dryness)
Bowman75 RCT 133 C 48 =/NA =/NA = (general =/=$ = = = =
67/66 dryness)
Regimen A, 375 mg/m2 at day 0, 7, 14, 21; B, 375 mg/m2 at day 0, 15; C, 1 g at day 0, 15.
*MFI, Multidimensional Fatigue Index.
$PROFAD-SSI, Profile of Fatigue and Discomfort, Sicca Symptoms Inventory.

=, unchanged; +, significantly improved; BP, bodily pain domain; C, controls; ESSDAI, EULAR SS disease activity index; ESSPRI, EULAR SS patient reported index; EULAR, European
League Against Rheumatism; NA, not available; ODT, Ocular dye test; PF, physical functioning domain; pSS, primary Sjögren’s syndrome; Pts, patients; RTX, rituximab; Sch, Schirmer’s
test; SF, social functioning domain; SF36, Short Form 36; SSF, stimulated salivary flow; USF, unstimulated whole salivary flow; VAS, visual analogue scale; Vit, vitality domain.

48 journals.sagepub.com/home/tab
N Del Papa and C Vitali

Epratuzumab is a B-cell-directed nondepleting tocilizumab, a monoclonal antibody targeting


monoclonal antibody that targets CD22.32 It is IL-6, has been considered potentially active in
currently being evaluated in two phase III clinical this disorder. RCTs on the effectiveness of tocili-
trials in patients with SLE, a disease associated zumab in patients presenting with systemic
with abnormalities in B-cell function and involvement and specific autoantibodies are
activation. ongoing and the results are expected relatively
soon.
In a past phase II study, the administration of
epratuzumab led to a significant improvement of Because IFNs and IL-6 produce their multiple
fatigue, decrease in patient and physician global biological effects by activating JAK/STAT path-
assessments, and in composite score that also ways, the increasing availability of small mole-
included the Schirmer-I test, unstimulated whole cules able to modulate these transducer signals
salivary flow, erythrocyte sedimentation rate may open up new scenarios in the treatment of
(ESR) and IgG levels.85 pSS. Phase II studies with filgotinib, a JAK1
inhibitor,91 are in progress.
Therapies against antigen-presenting cell mecha-
nism and costimulatory molecules.  Given the role Therapy against molecules favouring the lymphoid
of cathepsin S in antigen presentation to T cells, infiltrates organization and germinal-centre-like
an inhibitor of this molecule is presently being formation. Baminercept is a fusion protein that
tested in a phase II study. includes the lymphotoxin-β receptor. This molec-
ular construct has been shown to be able to
Abatacept is a fusion protein composed of the Fc reduce B-cell infiltration and organization in the
region of IgG1 combined with the extracellular NOD mouse model.40 An RCT with baminercept
domain of CTL4. As previously mentioned in a series of patients with SS has been completed
CTLA4, competing with CD28, is the natural and has shown an improvement in ESSDAI score,
inhibitor of the B7-CD28 costimulatory system, but no effect on sicca features.92 The clinical
and so is active in negatively modulating the anti- application of such a treatment is presently under
gen presentation mechanism.22 Preliminary data evaluation because of the high risk of liver dam-
obtained in open-label studies showed that admin- age observed in some cases.
istration of abatacept has favourable effect on sys-
temic features, extent of glandular infiltrates and
reduction of RF title and IgG levels.86,87 Starting Conclusion
from these data, an RCT is currently ongoing. In the last few years a great effort has been made
Other monoclonal antibodies binding the CD40 by the rheumatologic scientific community to
molecule and interfering with CD40/CD40L link- reach an agreement on classification criteria and
age are presently under investigation. develop validated outcome measures for pSS.
The 2016 ACR/EULAR classification criteria are
Anti-cytokine therapies. In view of the demon- the result of this effort.41,42 This criteria set does
strated over expression of TNFα and IL-1 in not introduce new substantial elements with
pSS,14 and the good results obtained by anti- respect to the previous largely used AECG crite-
TNFα biological agents in RA, two explorative tri- ria, as demonstrated by the strong agreement
als have been conducted in patients with pSS with between the two criteria sets.41,42 Some criticisms
two different anti-TNFα agents, and both have have been made on the new classification criteria,
given negative results.88,89 Similar negative results namely the fact these criteria appear to be less
were also obtained by using an IL-1 inhibitor.90 stringent with respect to previously proposed
ones. This excess of sensitivity, with the conse-
Considering that pSS is a disease characterized by quent loss of specificity, may lead to possible
IFN type I signature, anti-IFN therapies appear erroneous selection of false-positive patients in
to be promising and some studies on this specific clinical therapeutic trials.93 This risk is certainly
target therapy are ongoing. relatively low when one considers the revised
comprehensive exclusion criteria that are detailed
Since levels of IL-6 have been found to be in the criteria formulation (see Table 1).
increased in both the blood and saliva of patients
with pSS, and overproduction of this cytokine by Conversely, to have more ‘liberal’ criteria may
the epithelial glandular cells has been shown,15 allow the early selection of patients who may be

journals.sagepub.com/home/tab 49
Therapeutic Advances in Musculoskeletal Disease 10(2)

more likely to positively respond to the innovative Conflict of interest statement


therapeutic regimens that are presently or will be The authors declare that there is no conflict of
available.94 interest.

ESSDAI and ESSPRI have been defined and vali-


dated in the last few years.45,49 Both these out-
come measures are presently considered as References
mandatory in designing new therapeutic trials in 1. Both T, Dalm VASH, van Hagen PM, et al.
pSS, since these indices allow the correct selec- Reviewing primary Sjögren’s syndrome: beyond
tion of candidate patients with a relevant level of the dryness – from pathophysiology to diagnosis
activity and measure the effectiveness of experi- and treatment. Int J Med Sci 2017; 14: 191–200.
mental treatment. In contrast, both indices appear
2. Goules AV and Tzioufas AG. Primary Sjögren’s
poorly sensitive to changes in patients with mild syndrome: clinical phenotypes, outcome and
stable disease.95 ESSPRI can also be criticized the development of biomarkers. Autoimmun Rev
since its extreme simplicity does not allow the 2016; 15: 695–703.
capture of different components of pain and
fatigue in patients with the disorder. However, 3. Karageorgas T, Fragioudakis S, Nezos A,
et al. Fatigue in primary Sjögren’s syndrome:
more detailed validated indices are available if the
clinical, laboratory, psychometric, and biologic
achievement of more specific results is required.53 associations. Arthritis Care Res 2016; 68: 123–131.

Molecular events promoting antigen presenta- 4. Howard Tripp N, Tarn J, Natasari A, et al.
tion, costimulation, inflammatory cell homing Fatigue in primary Sjögren’s syndrome is
and function, B-cell proliferation, as well as the associated with lower levels of proinflammatory
cytokines. RMD Open 2016; 2: e000282.
formation of ectopic germinal centres have been
the object of extensive studies in animal models of 5. Segal BM, Pogatchnik B, Henn L, et al. Pain
SS and in patients with pSS. The advances in severity and neuropathic pain symptoms in
molecular biology and engineering have enabled primary Sjögren’s syndrome: a comparison
the development of promising target therapies study of seropositive and seronegative Sjögren’s
able to interfere with or block the different patho- syndrome patients. Arthritis Care Res 2013; 65:
1291–1298.
logical pathways that are active in this disorder.
As demonstrated by the often unsatisfactory 6. Skopouli FN, Dafni U, Ioannidis JP, et al.
results of the previously conducted therapeutic Clinical evolution, and morbidity and mortality
trials, the possibility of successful use of these of primary Sjögren’s syndrome. Semin Arthritis
new therapeutic agents remains a major chal- Rheum 2000; 29: 296–304.
lenge. These unexpectedly negative results could 7. Fragkioudaki S, Mavragani CP and
be the consequence of the remarkable heteroge- Moutsopoulos HM. Predicting the risk of
neity of clinical and biological features that may lymphoma among patients with Sjögren’s
characterize different subsets of patients with SS. syndrome: an easy to use clinical tool. Medicine
Better knowledge of the different pathways that 2016; 95: e3766.
are operative in diverse clinical phenotypes at the 8. Nocturne G, Virone A, Ng WF, et al.
systemic level and in salivary gland tissue, and Rheumatoid factor and disease activity are
selection of the target therapy with the highest independent predictors of lymphoma in primary
probability of success, appears to be the best way Sjögren’s syndrome. Arthritis Rheumatol 2016; 68:
to reach the goal of actually improving the clinical 977–985.
condition and the quality of life of patients with 9. Brkic Z, Maria NI, van Helden-Meeuwsen CG,
pSS. Moreover, the identification or development et al. Prevalence of interferon type I signature
of reliable and sensitive outcome measures and in CD14 monocytes of patients with Sjögren’s
biomarkers specific for each of the different dis- syndrome and association with disease activity
ease subsets is certainly the second very difficult and BAFF gene expression. Ann Rheum Dis 2013;
challenge to be tackled in the near feature. 72: 728–735.
10. Hall JC, Casciola-Rosen L, Berger AE, et al.
Funding Precise probes of type II interferon activity define
This research received no specific grant from any the origin of interferon signatures in target tissues
funding agency in the public, commercial, or not- in rheumatic diseases. PNAS 2012; 109:
for-profit sectors. 17609–17614.

50 journals.sagepub.com/home/tab
N Del Papa and C Vitali

11. Gottenberg JE, Cagnard N, Lucchesi C, et al. Sjögren’s syndrome related lymphomagenesis.
Activation of IFN pathways and plasmacytoid J Immunol Res 2015; 754825. DOI:
dendritic cell recruitment in target organs of 10.1155/2015/754825.
primary Sjögren’s syndrome. PNAS 2006; 103:
25. Ramos-Casals M. The B-lymphocyte stimulator
2770–2775.
connection in Sjögren’s syndrome. Rheumatology
12. Hall JC, Baer AN, Shah AA, et al. Molecular (Oxford) 2013; 52: 223–225.
subsetting of interferon pathways in Sjögren’s
26. Cancro MP, D’Cruz DP and Khamashta MA.
syndrome. Arthritis Rheumatol 2015; 67:
The role of B lymphocyte stimulator (BLyS) in
2437–2446.
systemic lupus erythematosus. J Clin Invest 2009;
13. Nezos A, Gravani F, Tassidou A, et al. Type 119: 1066–1073.
I and II interferon signatures in Sjögren’s
27. Quartuccio L, Salvin S, Fabris M, et al. BLyS
syndrome pathogenesis: contributions in distinct
upregulation in Sjögren’s syndrome associated
clinical phenotypes and Sjögren’s related
with lymphoproliferative disorders, higher
lymphomagenesis. J Autoimmun 2015; 63: 47–58.
ESSDAI score and B-cell clonal expansion in the
14. Youinou P and Pers JO. Disturbance of cytokine salivary glands. Rheumatology (Oxford) 2013; 52:
networks in Sjögren’s syndrome. Arthritis Res Ther 276–281.
2011; 13: 227–237.
28. Wiestner A. The role of B-cell receptor inhibitors
15. Pertovaara M, Silvennoinen O and Isomäki P. in the treatment of patients with chronic
Cytokine-induced STAT1 activation is increased lymphocytic leukemia. Haematologica 2015; 100:
in patients with primary Sjögren’s syndrome. Clin 1495–1507.
Immunol 2016; 165: 60–67.
29. Nayar S, Campos J, Buckley CD, et al.
16. Hirano T, Ishihara K and Hibi M. Roles Phosphatidylinositol 3-kinase delta pathway a
of STAT3 in mediating the cell growth, novel therapeutic target for Sjögren’s syndrome.
differentiation and survival signals relayed Ann Rheum Dis 2016; 75: A58.
through the IL-6 family of cytokine receptors.
30. Corneth OBJ, Verstappen GMP, Paulissen SMJ,
Oncogene 2000; 19: 2548–2556.
et al. Enhanced Bruton’s tyrosine kinase activity
17. Bromberg J. Stat proteins and oncogenesis. J Clin in peripheral blood B lymphocytes from patients
Invest 2002; 109: 1139–1142. with autoimmune disease. Arthritis Rheumatol
2017; 69: 1313–1324.
18. Song JI and Grandis JR. STAT signaling in head
and neck cancer. Oncogene 2000; 19: 2489–2495. 31. Mócsai A, Ruland J and Tybulewicz VL. The
SYK tyrosine kinase: a crucial player in diverse
19. Ghoreschi K, Laurence A and O’Shea JJ. Janus
biological functions. Nat Rev Immunol 2010; 10:
kinases in immune cell signaling. Immunol Rev
387–402.
2009; 228: 273–287.
32. Dörner T, Shock A, Goldenberg DM, et al.
20. Thurmond RL, Sun S, Karlsson L,
The mechanistic impact of CD22 engagement
et al. Cathepsin S inhibitors as novel
with epratuzumab on B cell function:
immunomodulators. Curr Opin Invest Drugs 2005;
implications for the treatment of systemic
6: 473–482.
lupus erythematosus. Autoimmun Rev 2015;
21. Hamm-Alvarez SF, Janga SR, Edman MC, et al. 14: 1079–1086.
Tear cathepsin S as a candidate biomarker for
33. Risselada AP, Looije MF, Kruize AA, et al.
Sjögren’s syndrome. Arthritis Rheumatol 2014; 66:
The role of ectopic germinal centers in the
1872–1881.
immunopathology of primary Sjögren’s
22. Kapsogeorgou EK, Moutsopoulos HM and syndrome: a systematic review. Semin Arthritis
Manoussakis MN. Functional expression of a Rheum 2013; 42: 368–376.
costimulatory B7.2 (CD86) protein on human
34. Theander E, Vasaitis L, Baecklund E, et al.
salivary gland epithelial cells that interacts with
Lymphoid organisation in labial salivary
the CD28 receptor, but has reduced binding to
gland biopsies is a possible predictor for the
CTLA4. J Immunol 2001; 166: 3107–3113.
development of malignant lymphoma in primary
23. Kroese FG, Abdulahad WH, Haacke E, et al. Sjögren’s syndrome. Ann Rheum Dis 2011; 70:
B-cell hyperactivity in primary Sjögren’s 1363–1368.
syndrome. Expert Rev Clin Immunol 2014; 10:
35. Szabo K, Papp G, Barath S, et al. Follicular
483–499.
helper T cells may play an important role in the
24. Nezos A and Mavragani CP. Contribution severity of primary Sjögren’s syndrome. Clin
of genetic factors to Sjögren’s syndrome and Immunol 2013; 147: 95–104.

journals.sagepub.com/home/tab 51
Therapeutic Advances in Musculoskeletal Disease 10(2)

36. Bombardieri M, Barone F, Humby F, et al. 46. Jousse-Joulin S, Milic V, Jonsson MV, et al.
Activation-induced cytidine deaminase expression Is salivary gland ultrasonography a useful tool
in follicular dendritic cell networks and in Sjögren’s syndrome? A systematic review.
interfollicular large B cells supports functionality Rheumatology (Oxford) 2016; 55: 789–800.
of ectopic lymphoid neogenesis in autoimmune
47. Vitali C, Bombardieri S, Jonsson R, et al.;
sialoadenitis and MALT lymphoma in Sjögren’s
European Study Group on Classification Criteria
syndrome. J Immunol 2007; 179: 4929–4938.
for Sjögren’s Syndrome. Classification criteria
37. Barone F, Nayar S, Campos J, et al. IL-22 for Sjögren’s syndrome: a revised version of the
regulates lymphoid chemokine production and European criteria proposed by the American-
assembly of tertiary lymphoid organs. PNAS European Consensus Group. Ann Rheum Dis
2015; 112: 11024–11029. 2002; 61: 554–558.
38. Pitzalis C, Jones GW, Bombardieri M, et al. 48. Fisher BA, Jonsson R, Daniels T,
Ectopic lymphoid-like structures in infection, et al. Standardisation of labial salivary
cancer and autoimmunity. Nat Rev Immunol glandhistopathology in clinical trials in primary
2014; 14: 447–462. Sjögren’s syndrome. Ann Rheum Dis 2017; 76:
1161–1168.
39. Alunno A, Carubbi F, Bistoni O, et al. T
regulatory and T helper 17 cells in primary 49. Seror R, Bootsma H, Saraux A, et al. Defining
Sjögren’s syndrome: facts and perspectives. disease activity states and clinically meaningful
Mediators Inflamm 2015; 2015: 243723. DOI: improvement in primary Sjögren’s syndrome with
10.1155/2015/243723. EULAR primary Sjögren’s syndrome disease
activity (ESSDAI) and patient-reported indexes
40. Fava RA, Kennedy SM, Wood SG, et al.
(ESSPRI). Ann Rheum Dis 2016; 75: 382–389.
Lymphotoxin-beta receptor blockade reduces
CXCL13 in lacrimal glands and improves 50. Seror R, Ravaud P, Mariette X, et al. EULAR
corneal integrity in the NOD model of Sjögren’s Sjögren’s Syndrome Patient Reported Index
syndrome. Arthritis Res Ther 2011; 13: R182. (ESSPRI): development of a consensus patient
index for primary Sjögren’s syndrome. Ann
41. Shiboski CH, Shiboski SC, Seror R, et al.;
Rheum Dis 2011; 70: 968–972.
International Sjögren’s Syndrome Criteria
Working Group. 2016 American College of 51. Brito-Zeron P, Kostov B, Solans R, et al.
Rheumatology/European League Against Systemic activity and mortality in primary
Rheumatism classification criteria for primary Sjögren’s syndrome: predicting survival using the
Sjögren’s syndrome: a consensus and data-driven EULAR-SS Disease Activity Index (ESSDAI) in
methodology involving three international patient 1045 patients. Ann Rheum Dis 2016; 75: 348–
cohorts. Ann Rheum Dis 2017; 76: 9–16. 555.
42. Shiboski CH, Shiboski SC, Seror R, et al.; 52. Seror R, Meiners P, Baron G, et al. Development
International Sjögren’s Syndrome Criteria of the ClinESSDAI: a clinical score without
Working Group. 2016 American College of biological domain. A tool for biological studies.
Rheumatology/European League Against Ann Rheum Dis 2016; 75: 1945–1950.
Rheumatism classification criteria for primary
53. Bowman S, Pillemer S, Jonsson R, et al.
Sjögren’s syndrome: a consensus and data-driven
Revisiting Sjögren’s syndrome in the new
methodology involving three international patient
millennium: perspectives on assessment and
cohorts. Arthritis Rheumatol 2017; 69: 35–45.
outcome measures. Report of a workshop held
43. Baldini C, Talarico R, Tzioufas AG, et al. on 23 March 2000 at Oxford, UK. Rheumatology
Classification criteria for Sjögren’s syndrome: a 2001; 40: 1180–1188.
critical review. J Autoimmun 2012; 39: 9–14.
54. Leonardi A, Van Setten G, Amrane M, et al.
44. Vitali C, Bombardieri S, Jonsson R, et al. Efficacy and safety of 0.1% cyclosporine A
Classification criteria for Sjögren’s syndrome: a cationic emulsion in the treatment of severe dry
revised version of the European criteria proposed eye disease: a multicenter randomized trial. Eur J
by the American-European Consensus Group. Ophthalmol 2016; 26: 287–296.
Ann Rheum Dis 2002; 61: 554–558.
55. Vivino FB, et al. Pilocarpine tablets for the
45. Seror R, Ravaud P, Bowman S, et al. EULAR treatment of dry mouth and dry eye symptoms in
Sjögren’s Syndrome Disease Activity Index patients with Sjögren’s syndrome: a randomized,
(ESSDAI): development of a consensus systemic placebo-controlled, fixed-dose, multicenter trial.
disease activity index in primary Sjögren’s P92-01 Study Group. Arch Intern Med 1999; 159:
syndrome. Ann Rheum Dis 2010; 69: 1103–1109. 174–181.

52 journals.sagepub.com/home/tab
N Del Papa and C Vitali

56. Petrone D, Condemi JJ, Fife R, et al. A double- 68. Pijpe J, van Imhoff GW, Spijkervet FK, et al.
blind, randomized, placebo-controlled study Rituximab treatment in patients with primary
of cevimeline in Sjögren’s syndrome patients Sjögren’s syndrome: an open-label phase II study.
with xerostomia and keratoconjunctivitis sicca. Arthritis Rheum 2005; 52: 2740–2750.
Arthritis Rheum 2002; 46: 748–754.
69. Devauchelle-Pensec V, Pennec Y, Morvan J,
57. Foulks GN, Forstot SL, Donshik PC, et al. et al. Improvement of Sjögren’s syndrome after
Clinical guidelines for management of dry eye two infusions of rituximab (anti-CD20). Arthritis
associated with Sjögren’s disease. Ocul Surf 2015; Rheum 2007; 57: 310–317.
13: 118–132.
70. Meiners PM, Arends S, Brouwer E, et al.
58. Le Gall M, Cornec D, Pers JO, et al. A Responsiveness of disease activity indices ESSPRI
prospective evaluation of dental and periodontal and ESSDAI in patients with primary Sjögren’s
status in patients with suspected Sjögren’s syndrome treated with rituximab. Ann Rheum Dis
syndrome. Joint Bone Spine 2016; 83: 235–236. 2012; 71: 1297–1302.
59. van der Reijden WA, van der Kwaak H, 71. Carubbi F, Cipriani P, Marrelli A, et al. Efficacy
Vissink A, et al. Treatment of xerostomia with and safety of rituximab treatment in early primary
polymer-based saliva substitutes in patients with Sjögren’s syndrome: a prospective, multi-center,
Sjögren’s syndrome. Arthritis Rheum 1996; 39: follow-up study. Arthritis Res Ther 2013; 15:
57–63. R172.
60. Strietzel FP, Lafaurie GI, Mendoza GR, 72. Dass S, Bowman SJ, Vital EM, et al. Reduction
et al. Efficacy and safety of an intraoral of fatigue in Sjögren’s syndrome with rituximab:
electrostimulation device for xerostomia relief: results of a randomized double-blind, placebo-
a multicenter, randomized trial. Arthritis Rheum controlled pilot study. Ann Rheum Dis 2008; 67:
2011; 63: 180–190. 1541–1544.
61. Saraux A, Pers JO, Devauchelle-Pensec V, et al. 73. Meijer JM, Meiners PM, Vissink A, et al.
Treatment of primary Sjögren’s syndrome. Nat Effectiveness of rituximab treatment in primary
Rev Rheumatol 2016; 12: 456–471. Sjögren’s syndrome: a randomized, double-blind,
placebo-controlled trial. Arthritis Rheum 2010; 62:
62. Carsons SE, Vivino FB, Parke A, et al. Treatment
960–968.
guidelines for rheumatologic manifestations of
Sjögren’s: use of biologics, management of fatigue 74. Devauchelle-Pensec V, Mariette X, Jousse-Joulin
and inflammatory musculoskeletal pain. Arthritis S, et al. Treatment of primary Sjögren’s syndrome
Care Res 2017; 69: 517–527. with rituximab: a randomized trial. Ann Intern
Med 2014; 160: 233–242.
63. Gottenberg JE, Ravaud P, Puechal X, et al.
Effects of hydroxychloroquine on symptomatic 75. Bowman SJ, Everett CC, O’Dwyer JL, et al.
improvement in primary Sjögren syndrome: the Randomized controlled trial of rituximab and
JOQUER randomized clinical trial. JAMA 2014; cost-effectiveness analysis in treating fatigue and
312: 249–258. oral dryness in primary Sjögren’s syndrome.
Arthritis Rheumatol 2017; 69: 1440–1450.
64. Mavragani CP and Moutsopoulos HM.
Conventional therapy of Sjögren’s syndrome. Clin 76. St Clair EW, Levesque MC, Prak ET, et al.;
Rev Allergy Immunol 2007; 32: 284–291. Autoimmunity Centers of Excellence. Rituximab
therapy for primary Sjögren’s syndrome: an
65. Clark EA and Ledbetter JA. How does B cell
open-label clinical trial and mechanistic analysis.
depletion therapy work, and how can it be
Arthritis Rheum 2013; 65: 1097–1106.
improved? Ann Rheum Dis 2005; 64(Suppl. 4):
iv77–iv80. 77. Verstappen GM, van Nimwegen JF, Vissink A,
et al. The value of rituximab treatment in primary
66. Pijpe J, Meijer JM, Bootsma H, et al. Clinical and
Sjögren’s syndrome. Clin Immunol 2017; 182:
histologic evidence of salivary gland restoration
62–71.
supports the efficacy of rituximab treatment in
Sjögren’s syndrome. Arthritis Rheum 2009; 60: 78. Gottenberg JE, Cinquetti G, Larroche C, et al.
3251–3256. Efficacy of rituximab in systemic manifestations
of primary Sjögren’s syndrome: results in 78
67. Alunno A, Carubbi F, Bistoni O, et al. Interleukin
patients of the AutoImmune and Rituximab
(IL)-17-producing pathogenic T lymphocytes
registry. Ann Rheum Dis 2013; 72: 1026–1031.
co-express CD20 and are depleted by rituximab
in primary Sjögren’s syndrome: a pilot study. Clin 79. Goede V, Fischer K, Engelke A, et al.
Exp Immunol 2016; 184: 284–292. Obinutuzumab as frontline treatment of chronic

journals.sagepub.com/home/tab 53
Therapeutic Advances in Musculoskeletal Disease 10(2)

lymphocytic leukemia: updated results of the proof of concept ASAP study). Ann Rheum Dis
CLL11 study. Leukemia 2015; 29: 1602–1604. 2014; 73: 1393–1396.
80. Mariette X, Seror R, Quartuccio L, et al. Efficacy 88. Sankar V, Brennan MT, Kok MR, et al.
and safety of belimumab in primary Sjögren’s: Etanercept in Sjögren’s syndrome: a twelve-week
results of the BELISS open-label phase II study. randomized, double-blind, placebo controlled
Ann Rheum Dis 2015; 74: 526–531. pilot clinical trial. Arthritis Rheum 2004; 50:
2240–2245.
81. De Vita S, Quartuccio L, Seror R, et al. Efficacy
and safety of belimumab given for 12 months in 89. Mariette X, Ravaud P, Steinfeld S, et al.
primary Sjögren’s syndrome: the BELISS open- Inefficacy of infliximab in primary Sjögren’s
label phase II study. Rheumatology 2015; 54: syndrome: results of the randomized, controlled
2249–2256. Trial of Remicade in Primary Sjögren’s Syndrome
(TRIPSS). Arthritis Rheum 2004; 50: 1270–1276.
82. Quartuccio L, Mavragani CP and Nezos A.
Type I interferon predicts biological effect of 90. Norheim KB, Harboe E, Goransson LG, et al.
belimumab on rheumatoid factor positive B-cells Interleukin-1 inhibition and fatigue in primary
in Sjögren’s syndrome: results from the BELISS Sjögren’s syndrome: a double blind, randomised
trial. Ann Rheum Dis 2016; 75(Suppl. 2): 294. clinical trial. PLoS One 2012; 7: e30123.
83. Pontarini E, Fabris M, Quartuccio L, et al. 91. Norman P. Selective JAK inhibitors in
Treatment with belimumab restores B cell development for rheumatoid arthritis. Expert Opin
subsets and their expression of B cell activating Invest Drugs 2014; 23: 1067–1077.
factor receptor in patients with primary Sjögren’s
92. St. Clair EW, Baer AN, Noaiseh G, et al. The
syndrome. Rheumatology 2015; 54: 1429–1434.
clinical efficacy and safety of baminercept, a
84. De Vita S, Quartuccio L, Salvin S, et al. 2014. lymphotoxin-beta receptor fusion protein, in
Sequential therapy with belimumab followed by primary Sjögren’s syndrome: results from a
rituximab in Sjögren’s syndrome associated with randomized, double-blind, placebo-controlled
B-cell lymphoproliferation and overexpression of phase II trial. Arthritis Rheum 2015;
BAFF: evidence for long-term efficacy. Clin Exp 67(Suppl. 10): 3844 (abstract 3203).
Rheumatol 32: 490–494.
93. Tsuboi H, Hagiwara S, Asashima H, et al.
85. Steinfeld SD, Tant L, Burmester GR, et al. Comparison of performance of the 2016 ACR-
Epratuzumab (humanised anti-CD22 antibody) EULAR classification criteria for primary
in primary Sjögren’s syndrome: an open-label Sjögren’s syndrome with other sets of criteria
phase I/II study. Arthritis Res Ther 2006; 8: 1–11. in Japanese patients. Ann Rheum Dis 2017; 76:
1980–1985.
86. Adler S, Korner M, Forger F, et al. Evaluation
of histologic, serologic, and clinical changes 94. Vitali C and Del Papa N. Classification and
in response to abatacept treatment of primary diagnostic criteria in Sjögren’s syndrome: a long-
Sjögren’s syndrome: a pilot study. Arthritis Care standing and still open controversy. Ann Rheum
Res 2013; 65: 1862–1868. Dis 2017; 76: 1953–1954.
Visit SAGE journals online
journals.sagepub.com/ 87. Meiners PM, Vissink A, Kroese FGM, et al. 95. Campar A and Isenberg DA. Primary Sjögren’s
home/tab
Abatacept treatment reduces disease activity in syndrome activity and damage indices
SAGE journals early primary Sjögren’s syndrome (open-label comparison. Eur J Clin Invest 2010; 40: 636–644.

54 journals.sagepub.com/home/tab

You might also like