scispace - formally typeset
Search or ask a question
Journal ArticleDOI

A phase 2 randomized, double-blind, placebo-controlled, proof-of-concept study of oral seletalisib in primary Sjögren’s syndrome

TL;DR: Despite enrolment challenges, seletalisib demonstrated a trend towards clinical improvement in patients with PSS and demonstrated efficacy in reducing size and organisation of salivary gland inflammatory foci and in target engagement, thus reducing PI3K-mTOR signalling compared with placebo.
Abstract: Objectives This phase 2 proof-of-concept study (NCT02610543) assessed efficacy, safety and effects on salivary gland inflammation of seletalisib, a potent and selective PI3Kδ inhibitor, in patients with moderate-to-severe primary Sjogren's syndrome (PSS). Methods Adults with PSS were randomized 1:1 to seletalisib 45 mg/day or placebo, in addition to current PSS therapy. Primary end points were safety and tolerability and change from baseline in EULAR Sjogren's Syndrome Disease Activity Index (ESSDAI) score at week 12. Secondary end points included change from baseline at week 12 in EULAR Sjogren's Syndrome Patient Reported Index (ESSPRI) score and histological features in salivary gland biopsies. Results Twenty-seven patients were randomized (seletalisib n = 13, placebo n = 14); 20 completed the study. Enrolment challenges led to early study termination with loss of statistical power (36% vs 80% planned). Nonetheless, a trend for improvement in ESSDAI and ESSPRI [difference vs placebo: -2.59 (95% CI: -7.30, 2.11; P=0.266) and -1.55 (95% CI: -3.39, 0.28), respectively] was observed at week 12. No significant changes were seen in saliva and tear flow. Serious adverse events (AEs) were reported in 3/13 of patients receiving seletalisib vs 1/14 for placebo and 5/13 vs 1/14 discontinued due to AEs, respectively. Serum IgM and IgG concentrations decreased in the seletalisib group vs placebo. Seletalisib demonstrated efficacy in reducing size and organisation of salivary gland inflammatory foci and in target engagement, thus reducing PI3K-mTOR signalling compared with placebo. Conclusion Despite enrolment challenges, seletalisib demonstrated a trend towards clinical improvement in patients with PSS. Histological analyses demonstrated encouraging effects of seletalisib on salivary gland inflammation and organisation. Trial registration https://clinicaltrials.gov, NCT02610543.

Summary (3 min read)

Introduction

  • Primary Sjögren’s syndrome (PSS) is a chronic, inflammatory, autoimmune disease characterised by focal lymphocytic infiltration and progressive functional impairment of the exocrine glands, often associated with systemic symptoms, B-cell hyperactivation, autoantibody formation, and increased risk of lymphoma development (1-3).
  • Two controlled trials of rituximab (5, 6) and two studies of abatacept (7, 8) did not meet their primary endpoints.
  • Conversely, a small study with a CD40-targeted antibody, CFZ533, reported significant amelioration of European League Against Rheumatism Sjögren’s Syndrome Disease Activity Index , and a trend towards improvement in Sjögren’s Syndrome Patient Reported Index (9).
  • In a mouse model of PSS, treatment with seletalisib blocked PI3Kδ activity, demonstrated by significant reduction of pS6 phosphorylation and decreased accumulation of B and T lymphocytes and plasma cells.
  • Treatment with seletalisib also reduced autoantibody titres and improved saliva production (3), providing a strong rationale for the evaluation of seletalisib in patients with PSS.

Study design

  • 1 to oral seletalisib 45 mg once daily or matching placebo, in addition to current PSS therapy, according to a predetermined randomisation schedule (supplementary materials, section Methods), also known as Patients were randomised 1.
  • The study comprised a 12-week treatment period and a 4-week safety follow-up.

Patients

  • Eligible patients were aged 18–75 years with moderate-to-severe PSS meeting the American-European Consensus Group criteria (20), total ESSDAI score ≥5, and positive for anti-Sjögren's-syndrome-related antigen A (SSA; Ro) and/or antiSjögren's-syndrome-related antigen B (SSB; La) autoantibodies.
  • Eligibility required an unstimulated salivary flow rate of >0 mL/15 min and a salivary gland biopsy in the prior 12 months, or during screening.
  • Additional inclusion and exclusion criteria are provided in Supplementary Table S1.
  • The study was conducted in accordance with the Declaration of Helsinki and the protocol approved by national or regional independent ethics committees.
  • Written informed consent was obtained from all patients before enrolment.

Endpoints

  • The primary objectives were safety and tolerability and systemic disease activity.
  • Exploratory efficacy endpoints included changes in salivary gland biopsies at Week 12 and those listed in the supplementary materials, section Methods.
  • Safety and tolerability assessments included incidence of adverse events, changes in clinical laboratory measurements, vital signs, and 12-lead electrocardiograms (ECGs).
  • Staining was detected by immunohistochemistry using the Leica Bond RX automated staining system (Leica Biosystems, Milton Keynes, UK) (21).
  • Manual immunofluorescence staining for CD20, CD138, CD3 and pS6 was performed as previously described (3) on sequential sections.

Statistical analysis

  • Safety analyses were based on the safety set, which comprised all randomised patients who received at least one dose of study drug.
  • Efficacy analyses were based on the full analysis set, which comprised all patients in the safety set who had at least one post-baseline efficacy assessment.
  • The primary efficacy endpoint, change from baseline in ESSDAI at Week 12, was analysed by a mixed model for repeated measures (MMRM) to account for missing data, with covariates of treatment, visit, baseline ESSDAI score, and treatment by visit interaction.
  • The minimal clinically important improvement in ESSDAI score was defined as a decrease of three points.
  • A post-hoc Bayesian analysis was conducted to interpret the results based on the limited observed data (supplementary materials, section Methods).

Patient disposition and demographics

  • Fifty-one patients were screened, 27 patients randomised (seletalisib n=13, placebo n=14), and 20 completed the study (Fig. 1).
  • Due to enrolment challenges, the study was terminated early; nearly 2 years was needed to recruit the 51 patients.
  • Moreover, due to safety concerns related to potential interaction, the number of concomitant medications allowed during the study were limited, further restricting the inclusion criteria.
  • Baseline characteristics were generally well balanced between the two groups.

Histology

  • Salivary gland biopsies were obtained from consenting patients at baseline (seletalisib n=13, placebo n=13), and Week 12 (seletalisib n=7, placebo n=11).
  • Lymphocytic foci, B and T cell segregation, and germinal centres were identified with H&E, CD3/CD20 and CD21 staining, respectively (Fig. 3A, Supplementary Fig. S4).
  • At baseline, minor salivary gland biopsies showed broadly similar features between groups although percentage infiltration and focus score were slightly higher in the placebo group.
  • Of seletalisibtreated patients with histological data at Week 12, 6/7 were ESSDAI responders (greater than 3-point reduction in ESSDAI score), and also had decreases in infiltration, foci with follicular dendritic cells, segregation, and germinal centres.
  • PS6 staining decreased in the seletalisib group versus placebo in B cells, but not in T cells or plasma cells (Supplementary Fig. S6).

Discussion

  • PSS still represents an unmet clinical need.
  • This reduction may be related to missing data, where two patients receiving placebo trending toward worsening and one patient receiving seletalisib trending toward improvement had missing Week 12 ESSDAI scores.
  • The authors also observed significant changes in salivary gland pathology: salivary gland infiltrates from seletalisib-treated patients showed decreases in both the organization and extent of lymphocytic infiltration.
  • This suggests that PI3Kδ inhibition affects cell aggregation in the salivary glands, compromising the process of T/B cell activation, as reflected in the observed decrease in immunoglobulins.
  • In line with the results in this study, leniolisib, another PI3Kδ inhibitor, also showed a modest improvement in biological endpoints but did not achieve significant clinical results in patients with PSS (23).

Figure legends

  • (A) Change from baseline in ESSDAI score; (B) Percentage of patients with a ≥3 point reduction from baseline in ESSDAI score; (C) Change from baseline in ESSPRI score (full analysis set).
  • Fig. 3. Histological results from paired minor salivary gland biopsies.
  • (A) Representative sections from placebo-treated and seletalisib-treated patients at screening/baseline and Week 12.
  • In H&E stained sections, lymphocytic foci are indicated with arrows; margins of the foci are marked with green lines.
  • Individual patient data from paired biopsies (at baseline and Week 12) are shown for each group.

Disclosure statement

  • MJ, ND, and PW are employees and shareholders of UCB Pharma.
  • AP was an employee of UCB Pharma at the time this study was conducted, is currently employed by Exscientia Ltd, and holds share options for UCB Pharma.
  • BAF reports personal consultancy fees from BMS, Novartis, and Roche. MK and JSB report no competing interests.
  • SJB reports grants from Roche and UCB Pharma and personal consultancy fees from AstraZeneca, BMS, Celgene, GFK, MedImmune, MT Pharma, Novartis, ONO Pharmaceuticals, UCB Pharma, and Xtlbio.

Data sharing statement

  • Due to the small sample size in this trial, Individual Patient Data cannot be adequately anonymised and there is a reasonable likelihood that individual participants could be re-identified.
  • For this reason, data from this trial cannot be shared.

Did you find this useful? Give us your feedback

Content maybe subject to copyright    Report

University of Birmingham
A phase 2 randomised, double-blind, placebo-
controlled, proof-of-concept study of oral seletalisib
in primary Sjögren’s syndrome
Juarez, Maria; Diaz, Nieves; Johnston, Geoffrey; Nayar, Saba; Payne, Andrew; Helmer, Eric;
Cain, Dionne; Williams, Paulette; Devauchelle-Pensec, Valerie; Fisher, Benjamin; Giacomelli,
Roberto; Gottenberg, Jacques-Eric; Guggino, Giuliana; Kvarnstrom, Marika; Mariette, Xavier;
Ng, Wan-Fai; Rosas, Jose; Burson, Juan; Triolo, Giovanni; Barone, Francesca
DOI:
10.1093/rheumatology/keaa410
License:
None: All rights reserved
Document Version
Peer reviewed version
Citation for published version (Harvard):
Juarez, M, Diaz, N, Johnston, G, Nayar, S, Payne, A, Helmer, E, Cain, D, Williams, P, Devauchelle-Pensec, V,
Fisher, B, Giacomelli, R, Gottenberg, J-E, Guggino, G, Kvarnstrom, M, Mariette, X, Ng, W-F, Rosas, J, Burson,
J, Triolo, G, Barone, F & Bowman, S 2020, 'A phase 2 randomised, double-blind, placebo-controlled, proof-of-
concept study of oral seletalisib in primary Sjögren’s syndrome', Rheumatology.
https://doi.org/10.1093/rheumatology/keaa410
Link to publication on Research at Birmingham portal
Publisher Rights Statement:
This is a pre-copyedited, author-produced version of an article accepted for publication in Rheumatology following peer review. The version
of record, Maria Juarez, Nieves Diaz, Geoffrey I Johnston, Saba Nayar, Andrew Payne, Eric Helmer, Dionne Cain, Paulette Williams, Valerie
Devauchelle-Pensec, Benjamin A Fisher, Roberto Giacomelli, Jacques-Eric Gottenberg, Giuliana Guggino, Marika Kvarnström, Xavier
Mariette, Wan Fai Ng, José Rosas, Juan Sánchez Bursón, Giovanni Triolo, Francesca Barone, Simon J Bowman, A phase 2 randomized,
double-blind, placebo-controlled, proof-of-concept study of oral seletalisib in primary Sjögren’s syndrome, Rheumatology, ,keaa410 is
available online at: https://doi.org/10.1093/rheumatology/keaa410
General rights
Unless a licence is specified above, all rights (including copyright and moral rights) in this document are retained by the authors and/or the
copyright holders. The express permission of the copyright holder must be obtained for any use of this material other than for purposes
permitted by law.
•Users may freely distribute the URL that is used to identify this publication.
•Users may download and/or print one copy of the publication from the University of Birmingham research portal for the purpose of private
study or non-commercial research.
•User may use extracts from the document in line with the concept of ‘fair dealing’ under the Copyright, Designs and Patents Act 1988 (?)
•Users may not further distribute the material nor use it for the purposes of commercial gain.
Where a licence is displayed above, please note the terms and conditions of the licence govern your use of this document.
When citing, please reference the published version.
Take down policy
While the University of Birmingham exercises care and attention in making items available there are rare occasions when an item has been
uploaded in error or has been deemed to be commercially or otherwise sensitive.
If you believe that this is the case for this document, please contact UBIRA@lists.bham.ac.uk providing details and we will remove access to
the work immediately and investigate.
Download date: 10. Aug. 2022

Seletalisib SS0004 manuscript_Rheumatology
Page 1
A phase 2 randomised, double-blind, placebo-controlled,
proof-of-concept study of oral seletalisib in primary Sjögren’s
syndrome
Maria Juarez,
1
* Nieves Diaz,
1
* Geoffrey I Johnston,
1
Saba Nayar,
2
Andrew Payne,
3†
Eric Helmer,
4†
Dionne Cain,
5
Paulette Williams,
6
Valerie Devauchelle-Pensec,
7
Benjamin A Fisher,
8,9
Roberto Giacomelli,
10
Jacques-Eric Gottenberg,
11
Giuliana
Guggino,
12
Marika Kvarnström,
13
Xavier Mariette,
14
Wan Fai Ng,
15
Jose Rosas,
16
Juan Sanchez Burson,
17
Giovanni Triolo,
12
Francesca Barone,
2
* Simon J Bowman
8
*
*Dr Maria Juarez, Dr Nieves Diaz, Dr Francesca Barone and Professor Simon J
Bowman contributed equally to this paper
Employees of UCB Pharma at the time the study was conducted
1
Translational Medicine, UCB Pharma, Slough, UK
2
Institute of Inflammation and Ageing,
University of Birmingham, Birmingham, UK
3
Discovery Biology, UCB Pharma, Slough, UK
4
Quantitative Clinical Pharmacology, UCB Pharma, Slough, UK
5
Global Clinical Sciences and Operations, UCB Pharma, Slough, UK
6
Statistical Science and Innovation, UCB Pharma, Raleigh, NC, USA
7
Department of Rheumatology, Brest University, Cavale Blanche Hospital, Brest,
France
8
National Institute for Health Research (NIHR) Birmingham Biomedical Research
Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
9
Rheumatology Research Group, Institute of Inflammation and Ageing, University of
Birmingham, Birmingham, UK
10
Rheumatology Unit, Department of Biotechnological and Applied Clinical Sciences,
University of L'Aquila, L’Aquila, Italy
11
National Reference Centre For Rare Systemic Auto-Immune Diseases, Department
of Rheumatology, Strasbourg University Hospital, University of Strasbourg, IBMC,
CNRS UPR 3572, Strasbourg, France
12
Department of Health Promotion, Mother and Child Care, Internal Medicine and
Medical Specialties, Rheumatology Section, University of Palermo, Palermo, Italy
13
Department of Medicine, Rheumatology Unit, Karolinska Institutet, Stockholm,
Sweden

Seletalisib SS0004 manuscript_Rheumatology
Page 2
14
Department of Rheumatology,
Université Paris-Sud, Assistance Publique –
Hôpitaux de Paris, Hôpitaux Universitaires Paris-Sud, INSERM UMR1184, Le
Kremlin Bicêtre, France
15
Translational and Clinical Research Institute, Newcastle University & NIHR
Newcastle Biomedical Research Centre, Newcastle upon Tyne, UK
16
Department of Rheumatology, Hospital Marina Baixa, Villajoyosa, Spain
17
Department of Rheumatology, Infanta Luisa Hospital, Sevilla, Spain
Corresponding author:
Francesca Barone
Rheumatology Research Group, Institute of Inflammation and Ageing
Queen Elizabeth Hospital
Birmingham B15 2WB, UK
Email: f.barone@bham.ac.uk
ORCiD iD: 0000-0002-5287-9614
Running header: Phase 2 seletalisib study in primary Sjögren’s syndrome
Previous presentations in part: The results have been previously published as
an abstract for EULAR 2019 (Juarez M et al. Ann Rheum Dis. 2019;78[Suppl
2]:1692–1693).
Word count: 3,121

Seletalisib SS0004 manuscript_Rheumatology
Page 3
Abstract
Objectives
This phase 2 proof-of-concept study (NCT02610543) assessed efficacy, safety and
effects on salivary gland inflammation of seletalisib, a potent and selective PI3Kδ
inhibitor, in patients with moderate-to-severe primary Sjögren’s syndrome (PSS)
.
Methods
Adults with PSS were randomised 1:1 to seletalisib 45 mg/day or placebo, in addition
to current PSS therapy. Primary endpoints were safety and tolerability and change
from baseline in ESSDAI score at Week 12. Secondary endpoints included change
from baseline at Week 12 in ESSPRI score and histological features in salivary gland
biopsies.
Results
Twenty-seven patients were randomised (seletalisib n=13, placebo n=14); 20
completed the study. Enrolment challenges led to early study termination with loss of
statistical power (36% vs 80% planned). Nonetheless, a trend for improvement in
ESSDAI and ESSPRI (difference vs placebo: –2.59 [95% CI –7.30, 2.11; P=0.266]
and –1.55 [95% CI –3.39, 0.28], respectively) was observed at Week 12. No
significant changes were seen in saliva and tear flow. Serious adverse events (AEs)
were reported in 3/13 of patients receiving seletalisib versus 1/14 for placebo and
5/13 versus 1/14 discontinued due to AEs, respectively. Serum IgM and IgG
concentrations decreased in the seletalisib group versus placebo. Seletalisib
demonstrated efficacy in reducing size and organisation of salivary gland
inflammatory foci and in target engagement, thus reducing PI3K-mTOR signalling
compared with placebo.
Conclusion
Despite enrolment challenges, seletalisib demonstrated a trend towards clinical
improvement in patients with PSS. Histological analyses demonstrated encouraging
effects of seletalisib on salivary gland inflammation and organization.
Trial registration number:
NCT02610543

Seletalisib SS0004 manuscript_Rheumatology
Page 4
Keywords
Phosphatidylinositol 3-kinase delta (PI3Kδ); primary Sjögren’s syndrome; seletalisib;
proof-of-concept; histology
Key messages
Seletalisib demonstrated a trend towards clinical improvement in patients with
PSS despite enrolment-related underpowering
Histological analyses demonstrated encouraging effects of seletalisib on
salivary gland inflammation and organization
Seletalisib or other PI3Kδ inhibitors could be new effective drugs in PSS
requiring future development

Citations
More filters
Journal ArticleDOI
TL;DR: In this paper, Vanhaesebroeck et al. review efforts to understand and therapeutically exploit the biology of PI3Kα and PI3kδ pathway inhibitors, highlighting lessons learned and future opportunities.
Abstract: Overactive phosphoinositide 3-kinase (PI3K) in cancer and immune dysregulation has spurred extensive efforts to develop therapeutic PI3K inhibitors. Although progress has been hampered by issues such as poor drug tolerance and drug resistance, several PI3K inhibitors have now received regulatory approval — the PI3Kα isoform-selective inhibitor alpelisib for the treatment of breast cancer and inhibitors mainly aimed at the leukocyte-enriched PI3Kδ in B cell malignancies. In addition to targeting cancer cell-intrinsic PI3K activity, emerging evidence highlights the potential of PI3K inhibitors in cancer immunotherapy. This Review summarizes key discoveries that aid the clinical translation of PI3Kα and PI3Kδ inhibitors, highlighting lessons learnt and future opportunities PI3K signalling is one of the most frequently aberrantly activated pathways in cancer. However, the development of therapeutic PI3K pathway inhibitors has faced challenges including poor drug tolerance and drug resistance. Here, Vanhaesebroeck et al. review efforts to understand and therapeutically exploit the biology of PI3Kα and PI3Kδ — the key targets of currently approved PI3K inhibitors, highlighting lessons learned and future opportunities.

149 citations

Journal ArticleDOI
TL;DR: A review of the available evidence for systemic treatments for primary sjogren syndrome (pSS) can be found in this article, where the authors discuss advances in outcome assessment, the current evidence for DMARD use and an overview of promising future therapies.
Abstract: Primary Sjogren syndrome (pSS) is a systemic autoimmune disease that is characterized by a triad of symptoms that affect all patients (dryness, pain and fatigue). In addition, systemic involvement can affect between one-third and one-half of patients. The management of patients with pSS has been negatively affected by a lack of effective treatments; however, knowledge of the epidemiology of pSS has increased, and advances in developing classification criteria, systemic disease activity scoring and patient-reported outcomes have been made during the past decade. Progress has also been made in understanding the mechanisms that underlie the pathogenesis of pSS, which has enabled a more targeted therapeutic approach to be taken. At present, therapeutic decisions rely on the evaluation of symptoms and systemic manifestations and are mostly formed on the basis of experience rather than evidence, and on similarities with other autoimmune diseases, although the 2019 management recommendations from EULAR are now being used to inform clinical management of pSS. This Review summarizes the available evidence for systemic treatments for pSS and includes discussions of advances in outcome assessment, the current evidence for DMARD use and an overview of promising future therapeutics. Advances in understanding the pathogenic mechanisms of primary Sjogren syndrome (pSS) and the development of new outcome measures are aiding drug development for this disease. This Review describes current treatments and highlights promising candidates for future therapies for pSS.

42 citations

Journal ArticleDOI
TL;DR: A composite responder index that includes all main disease features in a single tool and is designed for use as a primary endpoint in pSS RCTs is designed.
Abstract: Objective To develop a composite responder index in primary Sjögren’s syndrome (pSS): the Sjögren’s Tool for Assessing Response (STAR). Methods To develop STAR, the NECESSITY (New clinical endpoints in primary Sjögren’s syndrome: an interventional trial based on stratifying patients) consortium used data-driven methods based on nine randomised controlled trials (RCTs) and consensus techniques involving 78 experts and 20 patients. Based on reanalysis of rituximab trials and the literature, the Delphi panel identified a core set of domains with their respective outcome measures. STAR options combining these domains were proposed to the panel for selection and improvement. For each STAR option, sensitivity to change was estimated by the C-index in nine RCTs. Delphi rounds were run for selecting STAR. For the options remaining before the final vote, a meta-analysis of the RCTs was performed. Results The Delphi panel identified five core domains (systemic activity, patient symptoms, lachrymal gland function, salivary gland function and biological parameters), and 227 STAR options combining these domains were selected to be tested for sensitivity to change. After two Delphi rounds, a meta-analysis of the 20 remaining options was performed. The candidate STAR was then selected by a final vote based on metrological properties and clinical relevance. Conclusion The candidate STAR is a composite responder index that includes all main disease features in a single tool and is designed for use as a primary endpoint in pSS RCTs. The rigorous and consensual development process ensures its face and content validity. The candidate STAR showed good sensitivity to change and will be prospectively validated by the NECESSITY consortium in a dedicated RCT.

16 citations

Journal ArticleDOI
TL;DR: Wang et al. as discussed by the authors discuss the disappointing results of recent trials of primary Sjogren syndrome (pSS) and explain recent negative results of tocilizumab and abatacept, apart from the inefficacy of these biologicals in pSS.
Abstract: We read with interest the correspondence related to our article, ‘Interleukin 6 receptor inhibition in primary Sjogren syndrome: a multicentre double-blind randomised placebo-controlled trial’, by Wang et al 1 who discuss the disappointing results of recent trials of primary Sjogren syndrome (pSS) To explain recent negative results of tocilizumab and abatacept, apart from the inefficacy of these biologicals in pSS, Wang et al discuss the contribution of the design of the study First, they discuss the choice of a primary endpoint based on the EULAR Sjogren’s Syndrome Disease Activity Index (ESSDAI) As mentioned by the authors, this score captures only the systemic complications of the disease, observed in 30%–50% …

9 citations

Journal ArticleDOI
TL;DR: The progress in discovering novel treatments for primary sjogren syndrome (pSS) seem to be focused on searching new biological therapies as highly-selective drugs that can be effective without the adverse effects related to the wide, nonspecific immunosuppression induced by the drugs currently used.
Abstract: Introduction: Primary Sjogren syndrome (pSS) is a systemic autoimmune disease that may affect 3 in 1,000 people within the general population. The therapeutic scenario is complex, and no therapy has proved to be able to modify the natural course of the disease, nor to prevent the most severe systemic complications.Areas covered: Recently, the EULAR 2020 Recommendations for pSS have underlined the low level of evidence supporting efficacious therapeutic approaches, lacking a definition of specific treatment targets and being far from the 'disease modification' concept that is frequently used in other diseases. Herein, the authors review the status of current targeted therapies and provide the reader with their expert opinion.Expert opinion: The progress in discovering novel treatments for pSS seem to be focused on searching new biological therapies as highly-selective drugs that can be effective without the adverse effects related to the wide, nonspecific immunosuppression induced by the drugs currently used. Most likely, the more disruptive therapeutic approach in pSS that could be seen in a few years is the use of combination strategies targeting different etiopathogenic pathways.

7 citations

References
More filters
Journal ArticleDOI
TL;DR: These classification criteria for Sjögren's syndrome have been re-examined by consensus group members, who have introduced some modifications, more clearly defined the rules for classifying patients with primary or secondary SS, and provided more precise exclusion criteria.
Abstract: Classification criteria for Sjogren's syndrome (SS) were developed and validated between 1989 and 1996 by the European Study Group on Classification Criteria for SS, and broadly accepted. These have been re-examined by consensus group members, who have introduced some modifications, more clearly defined the rules for classifying patients with primary or secondary SS, and provided more precise exclusion criteria.

4,914 citations

Journal ArticleDOI
TL;DR: Different biochemical and structural features of p110δ suggest divergent functional/regulatory capacities for this PI3K, which is exclusively found in leukocytes.
Abstract: Phosphoinositide 3-kinases (PI3Ks) are a family of lipid kinases that have been implicated in signal transduction through tyrosine kinase- and heterotrimeric G-protein-linked receptors. We report herein the cloning and characterization of p110delta, a novel class I PI3K. Like p110alpha and p110beta, other class I PI3Ks, p110delta displays a broad phosphoinositide lipid substrate specificity and interacts with SH2/SH3 domain-containing p85 adaptor proteins and with GTP-bound Ras. In contrast to the widely distributed p110alpha and beta, p110delta is exclusively found in leukocytes. In these cells, p110alpha and delta both associate with the p85alpha and beta adaptor subunits and are similarly recruited to activated signaling complexes after treatment with the cytokines interleukin 3 and 4 and stem cell factor. Thus, these class I PI3Ks appear not to be distinguishable at the level of p85 adaptor selection or recruitment to activated receptor complexes. However, distinct biochemical and structural features of p110delta suggest divergent functional/regulatory capacities for this PI3K. Unlike p110alpha, p110delta does not phosphorylate p85 but instead harbors an intrinsic autophosphorylation capacity. In addition, the p110delta catalytic domain contains unique potential protein-protein interaction modules such as a Pro-rich region and a basic-region leucine-zipper (bZIP)-like domain. Possible selective functions of p110delta in white blood cells are discussed.

410 citations

Journal ArticleDOI
TL;DR: The understanding of the PI3Ks in immunity is guided by fundamental discoveries made in simpler model organisms as well as by appreciating new adaptations of this signaling module in mammals in general and in immune cells in particular.
Abstract: Phosphoinositide 3-kinases (PI3Ks) control many important aspects of immune cell development, differentiation, and function. Mammals have eight PI3K catalytic subunits that are divided into three classes based on similarities in structure and function. Specific roles for the class I PI3Ks have been broadly investigated and are relatively well understood, as is the function of their corresponding phosphatases. More recently, specific roles for the class II and class III PI3Ks have emerged. Through vertebrate evolution and in parallel with the evolution of adaptive immunity, there has been a dramatic increase not only in the genes for PI3K subunits but also in genes for phosphatases that act on 3-phosphoinositides and in 3-phosphoinositide-binding proteins. Our understanding of the PI3Ks in immunity is guided by fundamental discoveries made in simpler model organisms as well as by appreciating new adaptations of this signaling module in mammals in general and in immune cells in particular.

355 citations

Journal ArticleDOI
TL;DR: This multicenter, double-blind, placebo-controlled, randomized trial found that rituximab alleviated some symptoms at week 6 but did not alleviate symptoms or improve global activity score at month 6 in adults with recent-onset or systemic pSS.
Abstract: BACKGROUND: Primary Sjogren syndrome (pSS) is an autoimmune disorder characterized by ocular and oral dryness or systemic manifestations. OBJECTIVE: To evaluate efficacy and harms of rituximab in adults with recent-onset or systemic pSS. DESIGN: Randomized, placebo-controlled, parallel-group trial conducted between March 2008 and January 2011. Study personnel (except pharmacists), investigators, and patients were blinded to treatment group. (ClinicalTrials.gov: NCT00740948). SETTING: 14 university hospitals in France. PATIENTS: 120 patients with scores of 50 mm or greater on at least 2 of 4 visual analogue scales (VASs) (global disease, pain, fatigue, and dryness) and recent-onset (< 10 years) biologically active or systemic pSS. INTERVENTION: Randomization (1:1 ratio) to rituximab (1 g at weeks 0 and 2) or placebo. MEASUREMENTS: Primary end point was improvement of at least 30 mm in 2 of 4 VASs by week 24. RESULTS: No significant difference between groups in the primary end point was found (difference, 1.0% [95% CI, -16.7% to 18.7%]). The proportion of patients with at least 30-mm decreases in at least two of the four VAS scores was higher in the rituximab group at week 6 (22.4% vs. 9.1%; P = 0.036). An improvement of at least 30 mm in VAS fatigue score was more common with rituximab at weeks 6 (P < 0.001) and 16 (P = 0.012), and improvement in fatigue from baseline to week 24 was greater with rituximab. Adverse events were similar between groups except for a higher rate of infusion reactions with rituximab. LIMITATION: Low disease activity at baseline and a primary outcome that may have been insensitive to detect clinically important changes. CONCLUSION: Rituximab did not alleviate symptoms or disease activity in patients with pSS at week 24, although it alleviated some symptoms at earlier time points.

305 citations

Journal ArticleDOI
TL;DR: The acquisition of lymphoid features byinflammatory foci in SS is critically associated with the enlargement of the inflammatory foci and with the expression of CXCL13 and CCL21 within the infiltrate, but is not associated with their expression by epithelial cells.
Abstract: Objective Ectopic lymphoneogenesis can occur in the salivary glands of Sjogren's syndrome (SS) patients and is associated with local antigen-driven B cell responses, autoantibody formation, and potential lymphomatous transformation. CXCL13 and CCL21 have been identified in salivary glands, but their role in ectopic lymphoneogenesis in SS remains unclear. This study aimed to evaluate the microanatomic association between CXCL13 and CCL21 expression and the acquisition of lymphoid features in periductal foci. Methods Salivary glands from 37 SS patients and 9 chronic sialadenitis patients were analyzed by immunohistochemistry for T cell/B cell segregation, CD21+ follicular dendritic cell networks, and peripheral lymph node addressin (PNAd)–positive high endothelial venules (HEVs) in relationship to the size of the aggregates and the expression of CXCL13 and CCL21 within infiltrating cells, epithelium, and endothelium. Results Grade 1 aggregates (10–50 lymphocytes) demonstrated predominance of nonorganized CD3+ cells, while grade 2 (>50 lymphocytes) and grade 3 (>50 with germinal centers) showed a progressive increase in CD20+ B cells and T cell/B cell segregation. This higher degree of lymphoid organization was significantly related to an increased expression of CXCL13 within infiltrating cells and PNAd+ HEV–associated CCL21-producing cells. Conversely, no association between lymphoid organization and lymphoid chemokine expression by epithelial cells was observed. Conclusion The acquisition of lymphoid features by inflammatory foci in SS is critically associated with the enlargement of the inflammatory foci and with the expression of CXCL13 and CCL21 within the infiltrate, but is not associated with their expression by epithelial cells. These data strongly support an active participation of CXCL13 and CCL21 in regulating the progressive organization and maintenance of periductal foci.

235 citations

Frequently Asked Questions (2)
Q1. What contributions have the authors mentioned in the paper "University of birmingham a phase 2 randomised, double-blind, placebo- controlled, proof-of-concept study of oral seletalisib in primary sjögren’s syndrome" ?

Citation for published version ( Harvard ): Juarez, M, Diaz, N, Johnston, G, Nayar, S, Payne, A, Helmer, E, Cain, D, Williams, P, Devauchelle-Pensec, V, Fisher, B, Giacomelli, R, Gottenberg, J-E, Guggino, G, Kvarnstrom, M, Mariette, X, Ng, W-F, Rosas, J, Burson, J, Triolo, G, Barone, F & Bowman, S 2020, ' A phase 2 randomised, double-blind, placebo-controlled, proof-ofconcept study of oral seletalisib in primary Sjögren ’ s syndrome ', Rheumatology. 

Larger studies with specific PI3Kδ inhibitors and potentially with patient stratification could be considered in the future.