486
Views
0
CrossRef citations to date
0
Altmetric
Review

Emerging treatment for Sjögren’s disease: a review of recent phase II and III trials

, & ORCID Icon
Pages 107-120 | Received 20 Jan 2023, Accepted 28 Apr 2023, Published online: 08 May 2023

References

  • Fox RI. Sjögren’s syndrome. Lancet. 2005;366(9482):321–331.
  • Shiboski CH, Shiboski SC, Seror R, et al. 2016 American College of Rheumatology/European League Against Rheumatism classification criteria for primary sjögren’s syndrome: a consensus and data‐driven methodology involving three international patient cohorts. Arthritis Rheumatol. 2017;69(1):35–45. DOI:10.1002/art.39859
  • Jousse‐joulin S, Gatineau F, Baldini C, et al. Weight of salivary gland ultrasonography compared to other items of the 2016 ACR/EULAR classification criteria for primary Sjögren’s syndrome. J Intern Med. 2020;287(2):180–188. DOI:10.1111/joim.12992
  • Rasmussen A, Radfar L, Lewis D, et al. Previous diagnosis of Sjögren’s syndrome as rheumatoid arthritis or systemic lupus erythematosus. Rheumatology. 2016;55(7):1195–1201. DOI:10.1093/rheumatology/kew023
  • Fox RI, Fox CM. Sjögren syndrome: why do clinical trials fail? Rheum Dis Clin. 2016;42(3):519–530.
  • Löfgren CD, Isberg P-E, Christersson C. Screening for oral dryness in relation to salivary flow rate addresses the need for functional tests of saliva. Oral Health Prev Dent. 2010;8:243–252.
  • Nichols K K, Nichols J J, MPH M, et al. The Lack of Association Between Signs and Symptoms in Patients With Dry Eye Disease. Cornea. 2004;23(8);762–770. DOI:10.1097/01.ico.0000133997.07144.9e
  • Ryu O, Atkinson J, Hoehn G, et al. Identification of parotid salivary biomarkers in Sjögren’s syndrome by surface-enhanced laser desorption/ionization time-of-flight mass spectrometry and two-dimensional difference gel electrophoresis. Rheumatology. 2006;45(9):1077–1086. DOI:10.1093/rheumatology/kei212
  • Kalk W, Vissink A, Spijkervet F, et al. Sialometry and sialochemistry: diagnostic tools for Sjögren’s syndrome. Ann Rheumatic Dis. 2001;60(12):1110–1116. DOI:10.1136/ard.60.12.1110
  • Akpek EK, Wu HY, Karakus S, et al. Differential diagnosis of Sjögren versus non-Sjögren dry eye through tear film biomarkers. Cornea. 2020;39(8):991–997. DOI:10.1097/ICO.0000000000002299
  • McCoy SS, Woodham M, Bartels CM, et al. Symptom-based cluster analysis categorizes sjögren’s disease subtypes: an international cohort study highlighting disease severity and treatment discordance. Arthritis Rheumatol. 2022;74(9):1569–1579. DOI:10.1002/art.42238
  • Tarn JR, Howard-Tripp N, Lendrem DW, et al. Symptom-based stratification of patients with primary sjögren’s syndrome: multi-dimensional characterisation of international observational cohorts and reanalyses of randomised clinical trials. Lancet Rheumatol. 2019;1(2):e85–94. DOI:10.1016/S2665-9913(19)30042-6
  • Verstappen GM, Pringle S, Bootsma H, et al. Epithelial–immune cell interplay in primary Sjögren syndrome salivary gland pathogenesis. Nat Rev Rheumatol. 2021;17(6):333–348. DOI:10.1038/s41584-021-00605-2
  • Ridgewell D, Thalayasingam N, Ng W-F. Sjögren’s syndrome: shedding light on emerging and key drug targets. Expert Opin Ther Targets. 2022;26(10):1–14.
  • Verstappen GM, Gao L, Pringle S, et al. The transcriptome of paired major and minor salivary gland tissue in patients with primary Sjögren’s syndrome. Front Immunol. 2021;12:681941.
  • Mingueneau M, Boudaoud S, Haskett S, et al. Cytometry by time-of-flight immunophenotyping identifies a blood Sjögren’s signature correlating with disease activity and glandular inflammation. J Allergy Clin Immunol. 2016 Jun;137(6):1809–1821.e12.
  • James JA, Guthridge JM, Chen H, et al. Unique Sjögren’s syndrome patient subsets defined by molecular features. Rheumatology (Oxford). 2020 Apr 1;59(4):860–868. DOI:10.1093/rheumatology/kez335
  • Bodewes IL, Al-Ali S, van Helden-Meeuwsen CG, et al. Systemic interferon type I and type II signatures in primary Sjögren’s syndrome reveal differences in biological disease activity. Rheumatology. 2018;57(5):921–930. DOI:10.1093/rheumatology/kex490
  • Seror R, Bootsma H, Saraux A, et al. Defining disease activity states and clinically meaningful improvement in primary Sjögren’s syndrome with EULAR primary Sjögren’s syndrome disease activity (ESSDAI) and patient-reported indexes (ESSPRI). Ann Rheumatic Dis. 2016;75(2):382–389. DOI:10.1136/annrheumdis-2014-206008
  • Mak A, Kow NY. The pathology of T cells in systemic lupus erythematosus. J Ofimmunol Res. 2014;2014:1–8.
  • Lee JJ, Park YJ, Park M, et al. Longitudinal analysis of symptom-based clustering in patients with primary Sjogren’s syndrome: a prospective cohort study with a 5-year follow-up period. J Transl Med. 2021;19(1):1–8. DOI:10.1186/s12967-021-03051-6
  • Dev NWS, Seror R, Baron G, et al. Development and preliminary validation of the Sjogren’s tool for assessing response (STAR): a consensual composite score for assessing treatment effect in primary Sjogren’s syndrome. Ann Rheumatic Dis. 2022;81:189–190.
  • Bootsma H, Arends S, de Wolff L, et al. POS0193 EVALUATION of CRESS in the PHASE 2 RANDOMISED PLACEBO-CONTROLLED STUDY of SEQUENTIAL BELIMUMAB/RITUXIMAB ADMINISTRATION in PATIENTS with PRIMARY SJÖGREN’S SYNDROME. Annals of Rheumatic Disease. 2022.
  • Segal B, Bowman SJ, Fox PC, et al. Primary Sjögren’s syndrome: health experiences and predictors of health quality among patients in the United States. Health Qual Life Outcomes. 2009;7(1):1–9. DOI:10.1186/1477-7525-7-46
  • Miyamoto ST, Valim V, Fisher BA. Health-related quality of life and costs in Sjögren’s syndrome. Rheumatology. 2021;60(6):2588–2601.
  • Lee T-Y, Slejko JF, Davies-Teye BB, et al. Economic burden of interstitial lung disease in a commercially insured population with Sjögren syndrome in the United States. J Manag Care Spec Pharm. 2022;28(7):786–794. DOI:10.18553/jmcp.2022.28.7.786
  • Callaghan R, Prabu A, Allan R, et al. Direct healthcare costs and predictors of costs in patients with primary Sjögren’s syndrome. Rheumatology. 2007;46(1):105–111. DOI:10.1093/rheumatology/kel155
  • Bowman SJ, Pierre YS, Sutcliffe N, et al. Estimating indirect costs in primary Sjögren’s syndrome. J Rheumatol. 2010;37(5):1010–1015. DOI:10.3899/jrheum.090734
  • Sivakumar GK, Patel J, Malvankar-Mehta MS, et al. Work productivity among Sjögren’s syndrome and non-Sjögren’s dry eye patients: a systematic review and meta-analysis. Eye. 2021;35(12):3243–3257. DOI:10.1038/s41433-020-01282-3
  • Koh S, Maeda N, Hori Y, et al. Effects of Suppression of Blinking on Quality of Vision in Borderline Cases of Evaporative Dry Eye. Cornea. 2008;27(3):275–278. DOI:10.1097/ICO.0b013e31815be9c8
  • Bejarano MV, Romanini F, Pellet AC, et al. Work productivity and activity impairment in patients with primary Sjögren’s syndrome. Clin Exp Rheumatol. 2021;39(6):S93–99. DOI:10.55563/clinexprheumatol/6rd9mr
  • W-F N, Bowman SJ, Griffiths B. United kingdom primary sjögren’s syndrome registry— a united effort to tackle an orphan rheumatic disease. Rheumatology. 2011;50(1):32–39.
  • Symmons D, Ng WF, Bowman SJ, et al. United kingdom primary sjögren’s syndrome registry-a united effort to tackle an orphan rheumatic disease. Rheumatology. 2011;50(1):32–39. DOI:10.1093/rheumatology/keq240
  • Gao M, Zhao L, Liang R, et al. Evaluation of the efficacy and safety of topical 0.05% cyclosporine eye drops (II) in the treatment of dry eye associated with primary Sjögren’s syndrome. Ocul Immunol Inflamm. 2022;1–7.
  • Doctor MB, Basu S. Lacrimal gland insufficiency in aqueous deficiency dry eye disease: recent advances in pathogenesis, diagnosis, and treatment. Semin Ophthalmol. 2022;37(7–8):801–812. Taylor & Francis. DOI:10.1080/08820538.2022.2075706
  • Eom Y, Song JS, Kim HM. Effectiveness of topical cyclosporin a 0.1%, diquafosol tetrasodium 3%, and their combination, in dry eye disease. J Ocul Pharmacol Ther. 2022;38(10):682–694.
  • Michelson PE, Fox RI. Overview of management of dry eye associated with Sjögren’s syndrome. Sjögren’s Syndrome: Springer; 2011. p. 179–201.
  • Chrcanovic B, Kisch J, Wennerberg A, et al. Dental implants in patients with Sjögren’s syndrome: a case series and a systematic review. International Journal of Oral and Maxillofacial Surgery. 2019;48(9):1250–1259. DOI:10.1016/j.ijom.2019.02.005
  • Dawson L, Caulfield V, Stanbury J, et al. Hydroxychloroquine therapy in patients with primary Sjögren’s syndrome may improve salivary gland hypofunction by inhibition of glandular cholinesterase. Rheumatology. 2005;44(4):449–455. DOI:10.1093/rheumatology/keh506
  • Hashimoto N, Nakazawa T, Iwasaki T, et al. POS0706 LONG-TERM HYDROXYCHLOROQUINE TREATMENT IMPROVES ESSPRI and ESSDAI in PATIENTS with PRIMARY SJOGREN’S SYNDROME. Annals of Rheumatic disease. 2022.
  • Wang S-Q, Zhang L-W, Wei P, et al. Is hydroxychloroquine effective in treating primary Sjogren’s syndrome: a systematic review and meta-analysis. BMC Musculoskelet Disord. 2017;18(1):1–13. DOI:10.1186/s12891-017-1543-z
  • Gottenberg J-E, Ravaud P, Puéchal X, et al. Effects of hydroxychloroquine on symptomatic improvement in primary Sjögren syndrome: the JOQUER randomized clinical trial. JAMA. 2014;312(3):249–258. DOI:10.1001/jama.2014.7682
  • Rynes RI. Hydroxychloroquine treatment of rheumatoid arthritis. Am j med. 1988;85(4):18–22.
  • Manoussakis M, Moutsopoulos H. Antimalarials in Sjogren’s syndrome-the greek experience. Lupus. 1996;5(1_suppl):28–30.
  • Crofford LJ, Mease PJ, Simpson SL, et al. Fibromyalgia relapse evaluation and efficacy for durability of meaningful relief (FREEDOM): a 6-month, double-blind, placebo-controlled trial with pregabalin. Pain. 2008;136(3):419–431. DOI:10.1016/j.pain.2008.02.027
  • Van Woerkom J, Kruize A, Geenen R, et al. Safety and efficacy of leflunomide in primary Sjögren’s syndrome: a phase II pilot study. Ann Rheumatic Dis. 2007;66(8):1026–1032. DOI:10.1136/ard.2006.060905
  • Pepple S, Arnold J, Vital EM, et al. P245 identifying predictors of short-term response to rituximab in extra-glandular primary Sjogren’s syndrome. Rheumatology. 2022;61(Supplement_1):keac133. 244. DOI:10.1093/rheumatology/keac133.244
  • Dass S, Bowman SJ, Vital EM, et al. Reduction of fatigue in sjögren syndrome with rituximab: results of a randomised, double-blind, placebo-controlled pilot study. Ann Rheumatic Dis. 2008;67(11):1541–1544. DOI:10.1136/ard.2007.083865
  • Devauchelle-Pensec V, Mariette X, Jousse-Joulin S, et al. Tolerance and Efficacy of Rituximab in Primary Sjogren Syndrome (Tears): results of a randomized controlled trial [conference paper]. Ann Rheum Dis. 2012 Jun;71:75.
  • Fisher BA, Everett CC, Rout J, et al. Effect of rituximab on a salivary gland ultrasound score in primary Sjögren’s syndrome: results of the TRACTISS randomised double-blind multicentre substudy. Ann Rheum Dis. 2018 Mar;77(3):412–416.
  • Mariette X, Barone F, Baldini C, et al. A randomized, phase II study of sequential belimumab and rituximab in primary Sjögren’s syndrome. JCI Insight. 2022;7(23).
  • de Wolff L, van Nimwegen JF, Mossel E, et al. editors. Long-term abatacept treatment for 48 weeks in patients with primary Sjögren’s syndrome: the open-label extension phase of the ASAP-III trial. Semin Arthritis Rheum. 2022;53:151955. Elsevier. DOI:10.1016/j.semarthrit.2022.151955
  • Endocrinopathy TSsS RI. Abatacept ameliorates both glandular and extraglandular involvements in patients with Sjögren’s syndrome associated with rheumatoid arthritis: findings from an open-label. 2022.
  • Chowdhury F. The longitudinal effects of rituximab in the Trial for Anti-B Cell Therapy in Primary Sjögren’s Syndrome (TRACTISS). 2022.
  • Sankar V, Brennan MT, Kok MR, et al. Etanercept in Sjögren’s syndrome: a twelve‐week randomized, double‐blind, placebo‐controlled pilot clinical trial. Arthritis Rheumatol. 2004;50(7):2240–2245. DOI:10.1002/art.20299
  • Felten R, Devauchelle-Pensec V, Seror R, et al. Interleukin 6 receptor inhibition in primary Sjögren syndrome: a multicentre double-blind randomised placebo-controlled trial. Ann Rheumatic Dis. 2021;80(3):329–338. DOI:10.1136/annrheumdis-2020-218467
  • Verstappen GM, van Nimwegen JF, Vissink A, et al. The value of rituximab treatment in primary Sjögren’s syndrome. Clin Immunol. 2017;182:62–71.
  • Bowman SJ, Everett CC, O’Dwyer JL, et al. Randomized controlled trial of rituximab and cost-effectiveness analysis in treating fatigue and oral dryness in primary Sjögren’s syndrome. Arthritis Rheumatol. 2017 Jul;69(7):1440–1450.
  • Quartuccio L, Fabris M, Salvin S, et al. Controversies on rituximab therapy in sjögren syndrome-associated lymphoproliferation. Int J Rheumatol. 2009;2009. DOI:10.1155/2009/424935
  • Chen YH, Wang XY, Jin X, et al. Rituximab therapy for primary Sjögren’s syndrome. Front Pharmacol. 2021;12:731122.
  • Merino‐vico A, Frazzei G, van Hamburg JP, et al. Targeting B cells and plasma cells in autoimmune diseases: from established treatments to novel therapeutic approaches. Eur J Immunol. 2023 Jan;53(1):e2149675.
  • Mariette X, Seror R, Quartuccio L, et al. Efficacy and safety of belimumab in primary Sjögren’s syndrome: results of the BELISS open-label phase II study. Ann Rheumatic Dis. 2015;74(3):526–531. DOI:10.1136/annrheumdis-2013-203991
  • De Vita S, Quartuccio L, Seror R, et al. Efficacy and safety of belimumab given for 12 months in primary Sjögren’s syndrome: the BELISS open-label phase II study. Rheumatology. 2015;54(12):2249–2256. DOI:10.1093/rheumatology/kev257
  • Bowman SJ, Fox R, Dörner T, et al. Safety and efficacy of subcutaneous ianalumab (VAY736) in patients with primary Sjögren’s syndrome: a randomised, double-blind, placebo-controlled, phase 2b dose-finding trial. Lancet. 2022;399(10320):161–171. DOI:10.1016/S0140-6736(21)02251-0
  • Baer AN, Gottenberg JE, St Clair EW, et al. Efficacy and safety of abatacept in active primary Sjögren’s syndrome: results of a phase III, randomised, placebo-controlled trial. Ann Rheum Dis. 2021 Mar;80(3):339–348.
  • Fisher BA, Szanto A, W-F N, et al. Assessment of the anti-CD40 antibody iscalimab in patients with primary Sjögren’s syndrome: a multicentre, randomised, double-blind, placebo-controlled, proof-of-concept study. Lancet Rheumatol. 2020;2(3):e142–152. DOI:10.1016/S2665-9913(19)30135-3
  • Karnell JL, Rieder SA, Ettinger R, et al. Targeting the CD40-CD40L pathway in autoimmune diseases: humoral immunity and beyond. Adv Drug Delivery Rev. 2019;141:92–103.
  • van der Heijden E HM, Hartgring S AY, Kruize A A, et al. Additive immunosuppressive effect of leflunomide and hydroxychloroquine supports rationale for combination therapy for Sjögren’s syndrome. Expert Review of Clinical Immunology. 2019;15(7):801–808. DOI:10.1080/1744666X.2019.1624527
  • Norheim KB, Harboe E, Gøransson LG, et al. Interleukin-1 inhibition and fatigue in primary Sjögren’s syndrome–a double blind, randomised clinical trial. PLoS ONE. 2012;7(1):e30123. DOI:10.1371/journal.pone.0030123
  • Chiche L, Cornec D. Mysterious uncoupled clinical symptoms and interferon signature in Sjögren’s syndrome: limitations of current approaches for unravelling complexity? Rheumatology. 2020;59(1):5–6.
  • Fisher B, Barone F, Jobling K, et al. OP0202 effect of RSLV-132 on fatigue in patients with primary Sjögren’s syndrome–results of aphase II randomised, double-blind, placebo-controlled, proof of concept study. Annals of Rheumatic disease. 2019;78:177.
  • Bai W, Yang F, Xu H, et al. A multi-center, prospective, open-label, randomized study to explore efficacy and safety of baricitinib in active primary sjogren’s syndrome patients. Trials. . 2023 Feb 15;24(1):112.
  • Price E, Bombardieri M, Kivitz A, et al. Safety and efficacy of filgotinib, lanraplenib and tirabrutinib in Sjögren’s syndrome: a randomized, phase 2, double-blind, placebo-controlled study. Rheumatology. 2022;61(12):4797–4808. DOI:10.1093/rheumatology/keac167
  • Ringheim GE, Wampole M, Oberoi K. Bruton’s tyrosine kinase (BTK) inhibitors and autoimmune diseases: making sense of BTK inhibitor specificity profiles and recent clinical trial successes and failures. Front Immunol. 2021;12:662223.
  • Watterson SH, Liu Q, Beaudoin Bertrand M, et al. Discovery of branebrutinib (BMS-986195): astrategy for identifying ahighly potent and selective covalent inhibitor providing rapid invivo inactivation of bruton’s tyrosine kinase (BTK). J Med Chem. 2019 Apr 11;62(7):3228–3250. DOI:10.1021/acs.jmedchem.9b00167. Epub 2019 Mar 29.
  • Nair JJ, Singh TP. Sjogren’s syndrome: review of the aetiology, pathophysiology & potential therapeutic interventions. J Clin Exp Dent. 2017;9(4):e584.
  • Zhang H, Gao D, Ji L, et al.Ab0030 the biological function and mechanism exploration of taci in sjogren’s syndrome. Annals of the Rheumatic Diseases. 2022;81:1149–1150.
  • Vugmeyster Y, Seshasayee D, Chang W, et al. A soluble BAFF antagonist, BR3-fc, decreases peripheral blood B cells and lymphoid tissue marginal zone and follicular B cells in cynomolgus monkeys. Am J Pathol. 2006;168(2):476–489. DOI:10.2353/ajpath.2006.050600
  • Clair EW S, Baer AN, Wei C, et al. Clinical efficacy and safety of baminercept, a lymphotoxin β receptor fusion protein, in primary sjögren’s syndrome: results from a phase II randomized, double‐Blind, placebo‐Controlled trial. Arthritis Rheumatol. 2018;70(9):1470–1480. DOI:10.1002/art.40513
  • Seror R, Nocturne G, Mariette X. Current and future therapies for primary sjögrensyndrome. Nat Rev Rheumatol. 2021;17(8):475–486.
  • Mariette X, Bombardieri M, Alevizos I, et al., editors. A phase 2a study of MEDI5872 (AMG557), a fully human anti-ICOS ligand monoclonal antibody in patients with primary sjogren’s syndrome. Arthritis Rheumatol. 2019;71(suppl 10). Accessed May 3, 2023. https://acrabstracts.org/abstract/a-phase-2a-study-of-medi5872-amg557-a-fully-human-anti-icos-ligand-monoclonal-antibody-in-patients-with-primary-sjogrens-syndrome
  • Chatzis L, Vlachoyiannopoulos PG, Tzioufas AG, et al. New frontiers in precision medicine for sjogren’s syndrome. Expert Rev Clin Immunol. 2021;17(2):127–141. DOI:10.1080/1744666X.2021.1879641
  • Ritter J, Chen Y, Stefanski L, et al. Current and future treatment in primary sjögren’s syndrome – a still challenging development. Joint Bone Spine. 2022;89(6):105406. DOI:10.1016/j.jbspin.2022.105406
  • Chen H, Qi X, Li Y, et al. Iguratimod treatment reduces disease activity in early primary sjögren’s syndrome: an open-label pilot study. Mod Rheumatol. 2021;31(2):394–398. DOI:10.1080/14397595.2020.1789335
  • Smyth P, Sasiwachirangkul J, Williams R, et al. Cathepsin S (CTSS) activity in health and disease-A treasure trove of untapped clinical potential. Mol Aspect Med. 2022;88:101106.
  • Dörner T, Zeher M, Laessing U, et al. OP0250 A randomised, double-blind study to assess the safety, tolerability and preliminary efficacy of leniolisib (CDZ173) in patients with primary sjÖgren’s syndrome. Annals of the Rheumatic Diseases. 2018;77:174.
  • Juarez M, Diaz N, Johnston GI, et al. A phase 2 randomized, double-blind, placebo-controlled, proof-of-concept study of oral seletalisib in primary sjögren’s syndrome. Rheumatology. 2021;60(3):1364–1375. DOI:10.1093/rheumatology/keaa410
  • Mavragani CP, Moutsopoulos HM. Sjögren’s syndrome: old and new therapeutic targets. J Autoimmun. 2020;110:102364.
  • Pinto ACPN, Piva SR, da Silva Vieira AG, et al. Transcranial direct current stimulation for fatigue in patients with sjogren’s syndrome: a randomized, double-blind pilot study. Brain Stimulation. 2021;14(1):141–151. DOI:10.1016/j.brs.2020.12.004
  • Kedor C, Zernicke J, Hagemann A, et al. A phase II investigator-initiated pilot study with low-dose cyclosporine a for the treatment of articular involvement in primary sjögren’s syndrome. Clin Rheumatol. 2016;35(9):2203–2210. DOI:10.1007/s10067-016-3360-4
  • Luo J, Ming B, Zhang C, et al. IL-2 inhibition of th17 generation rather than induction of treg cells is impaired in primary sjögren’s syndrome patients. Front Immunol. 2018;9:1755.
  • He J, Chen J, Miao M, et al. Efficacy and safety of low-dose interleukin 2 for primary sjögren syndrome: a randomized clinical trial. JAMA Netw Open. 2022;5(11):e2241451. DOI:10.1001/jamanetworkopen.2022.41451
  • Pérez-Jeldres T, Alvarez-Lobos M, Rivera-Nieves J. Targeting sphingosine-1-phosphate signaling in immune-mediated diseases: beyond multiple sclerosis. Drugs. 2021;81(9):985–1002.
  • Zaheer M, Wang C, Bian F, et al. Protective role of commensal bacteria in sjögren syndrome. J Autoimmun. 2018;93:45–56.
  • Seror R, Gottenberg JE, Devauchelle-Pensec V, et al. European league against rheumatism sjögren’s syndrome disease activity index and european league against rheumatism sjögren’s syndrome patient-reported index: a complete picture of primary sjögren’s syndrome patients. Arthritis Care Res (Hoboken). 2013 Aug;65(8):1358–1364.
  • James K, Al-Ali S, Tarn J, et al. A transcriptional signature of fatigue derived from patients with primary sjögren’s syndrome. PLoS ONE. 2015;10(12):e0143970. DOI:10.1371/journal.pone.0143970
  • Mariette X, Criswell LA. Primary sjögren’s syndrome. N Engl J Med. 2018;378(10):931–939.
  • Brito-Zeron P, Baldini C, Bootsma H, et al. Sjögren syndrome. Nat Rev Dis Primers. 2016;2(1):1–20. DOI:10.1038/nrdp.2016.47
  • Beuerman RW, Mircheff A, Pflugfelder SC, et al. The lacrimal functional unit. dry eye and ocular surface disorders. (NY): Marcel Dekker; 2004. p. 11–39.
  • Fox RI, Stern M. Sjögren’s syndrome: mechanisms of pathogenesis involve interaction of immune and neurosecretory systems. Scand J Rheumatol. 2002;31(2):3–13.
  • Friedman DI. New frontiers in the treatment and prevention of migraine: a closer look at the role of emerging GCRP targeted therapies. J Man Care Med. 2019;22(2):13–19.
  • Dolgin E. FDA go-ahead for myasthenia gravis agent. Biotechnol. 2022 Jan;40(1):4. DOI:10.1038/s41587-021-01198-7
  • Steinfeld S editor. Effects of TNF blockade with infliximab in patients with primary sjogren’s syndrome: an open pilot study. European League Against Rheumatism (EULAR)/European Congress of Rheumatology: Prague Czech Republic. Jun 13-16. 2001.
  • Steinfeld SD, Demols P, Salmon I, et al. Notice of retraction of two articles (“infliximab in patients with primary sjögren’s syndrome: apilot study” and “infliximab in patients with primary sjögren’s syndrome: one‐year followup”). Arthritis Rheum. 2013 Mar;65(3):814.
  • Price E. Biologics in sjogren’s syndrome. In: Jain, N., Duggal, L, editors. Handbook of biologics for rheumatological disorders. Springer; 2022. p. 75–84. https://doi.org/10.1007/978-981-16-7200-2_7
  • Mariette X, Ravaud P, Steinfeld S, et al. editors. Infliximab in primary sjogren’s syndrome: open extension 2 of the TRIPSS study in 64 patients. In Arthritis and rheumatism. 2004. WILEY. WILEYLISS DIV JOHN WILEY & SONS INC, 111 RIVER ST, HOBOKEN, NJ 07030 USA
  • Moutsopoulos NM, Katsifis GE, Angelov N, et al. Lack of efficacy of etanercept in sjogren syndrome correlates with failed suppression of tumour necrosis factor and systemic immune activation. Ann Rheumatic Dis. 2008;67(10):1437–1443. DOI:10.1136/ard.2007.077891
  • van der Heijden EHM, Blokland SLM, Hillen MR, et al. Leflunomide–hydroxychloroquine combination therapy in patients with primary sjögren’s syndrome (repurpSS-I): a placebo-controlled, double-blinded, randomised clinical trial. Lancet Rheumatol. 2020;2(5):e260–269. DOI:10.1016/S2665-9913(20)30057-6
  • Devauchelle-Pensec V, Mariette X, Jousse-Joulin S, et al. Treatment of primary sjögren syndrome with rituximab a randomized trial [article]. Ann Intern Med. 2014;160(4):233–242. DOI:10.7326/M13-1085
  • Chu LL, Cui K, Pope JE. Meta‐analysis of treatment for primary sjögren’s syndrome. Arthritis Care Res. 2020;72(7):1011–1021.
  • Silverman GJ, Boyle DL. Understanding the mechanistic basis in rheumatoid arthritis for clinical response to anti‐CD20 therapy: the b‐cell roadblock hypothesis. Immunol Rev. 2008;223(1):175–185.
  • Chen M-H, Chen C-K, Chou H-P, et al. Rituximab therapy in primary sjögren’s syndrome with interstitial lung disease: a retrospective cohort study. Clin Exp Rheumatol. 2016;34(6):1077–1084.
  • Smolen J, Nash P, Tahir H, et al. Ianalumab (VAY736), a Dual Mode of Action Biologic Combining BAFF Receptor Inhibition with B Cell Depletion, for Treatment of Primary Sjögren’s Syndrome: Results of an International Randomized, Placebo Controlled Dose Range Finding Study in 190 Patients [abstract]. Arthritis rheumatol. 2019;71:(suppl 10). https://acrabstracts.org/abstract/ianalumab-vay736-a-dual-mode-of-action-biologic-combining-baff-receptor-inhibition-with-b-cell-depletion-for-treatment-of-primary-sjogrens-syndrome-results-of-an-international-randomized/. Accessed May 3.
  • Dörner T, Bowman S, Fox R, et al. POS0692 IANALUMAB (VAY736) SAFETY and EFFICACY in PATIENTS with SJOGREN’S SYNDROME: 52 WEEK RESULTS from a RANDOMISED, PLACEBO-CONTROLLED, PHASE 2B DOSE-RANGING TRIAL. BMJ Publishing Group Ltd; 2021.
  • Arbitman L, Furie R, Vashistha H. B cell-targeted therapies in systemic lupuserythematosus. J Autoimmun. 2022;102873. DOI:10.1016/j.jaut.2022.102873
  • Xu D, Zhang S, Huang C, et al. Efficacy and safety of telitacicept in primary sjögren’s syndrome: a randomized, double-blind, placebo-controlled, phase 2 trial. ACR AnnualConference; Philadelphia PA, 2022
  • Dhillon S. Telitacicept: first approval. Drugs. 2021;81(14):1671–1675.
  • van Nimwegen JF, Mossel E, van Zuiden GS, et al. Abatacept treatment for patients with early active primary sjögren’s syndrome: a single-centre, randomised, double-blind, placebo-controlled, phase 3 trial (ASAP-III study). Lancet Rheumatol. 2020;2(3):e153–163. DOI:10.1016/S2665-9913(19)30160-2
  • Xu H, Li X, Liu D, et al. Follicular T-helper cell recruitment governed by bystander B cells and ICOS-driven motility. Nature. 2013 Apr 25;496(7446):523–527. DOI:10.1038/nature12058
  • Posada J, Valadkhan S, Burge D, et al. Improvement of severe fatigue following nuclease therapy in patients with primary sjögren’s syndrome: a randomized clinical trial. Arthritis Rheumatol. 2020 Aug 15;73(1):143–150. DOI:10.1002/art.41489
  • Driedonks TA, Nolte-‘t Hoen EN. Circulating Y-RNAs in extracellular vesicles and ribonucleoprotein complexes; implications for the immune system. Front Immunol. 2019;9:3164.
  • Katze MG, Fornek JL, Palermo RE, et al. Innate immune modulation by RNA viruses: emerging insights from functional genomics. Nat Rev Immunol. 2008;8(8):644–654. DOI:10.1038/nri2377
  • Nocturne G, Mariette X. Interferon signature in systemic autoimmune diseases: what does it mean? BMJ Specialist J. 2022;8(2):e002687.
  • Zhang X, Miao M, Zhang R, et al. Efficacy and safety of low-dose interleukin-2 in combination with methotrexate in patients with active rheumatoid arthritis: a randomized, double-blind, placebo-controlled phase 2 trial. Signal Transduct Target Ther. 2022;7(1):1–8. DOI:10.1038/s41392-022-00887-2
  • Saadoun D, Rosenzwajg M, Joly F, et al. Regulatory T-cell responses to low-dose interleukin-2 in HCV-induced vasculitis. N Engl J Med. 2011 Dec 1;365(22):2067–2077. DOI:10.1056/NEJMoa1105143
  • He J, Zhang X, Wei Y, et al. Low-dose interleukin-2 treatment selectively modulates CD4(+) T cell subsets in patients with systemic lupus erythematosus. Nat Med. 2016 Sep;22(9):991–993.
  • Miao M, Hao Z, Guo Y, et al. Short-term and low-dose IL-2 therapy restores the th17/treg balance in the peripheral blood of patients with primary sjögren’s syndrome. Ann Rheumatic Dis. 2018;77(12):1838–1840. DOI:10.1136/annrheumdis-2018-213036
  • Graßhoff H, Comdühr S, Monne LR, et al. Low-dose IL-2 therapy in autoimmune and rheumatic diseases. Front Immunol. 2021;12:648408.
  • Lorenzo-Vizcaya A, Fasano S, Isenberg DA. Bruton’s tyrosine kinase inhibitors: a new therapeutic target for the treatment of SLE? Immunotargets Ther. 2020;9:105.
  • Bruton OC. Agammaglobulinemia. Pediatrics. 1952 Jun;9(6):722–728.
  • Corneth OB, de Bruijn MJ, Rip J, et al. Enhanced expression of bruton’s tyrosine kinase in B cells drives systemic autoimmunity by disrupting T cell homeostasis. J Immunol. 2016 Jul 1;197(1):58–67. DOI:10.4049/jimmunol.1600208
  • Nicolson PLR, Hughes CE, Watson S, et al. Inhibition of btk by btk-specific concentrations of ibrutinib and acalabrutinib delays but does not block platelet aggregation mediated by glycoprotein VI. Haematologica. 2018 Dec;103(12):2097–2108.
  • Kaul M, End P, Cabanski M, et al. Remibrutinib (LOU064): a selective potent oral BTK inhibitor with promising clinical safety and pharmacodynamics in a randomized phase I trial. Clin Transl Sci. 2021;14(5):1756–1768. DOI:10.1111/cts.13005
  • Strand V, O’Quinn S, Furie RA, et al. Clinical meaningfulness of a british isles lupus assessment group-based composite lupus assessment response in terms of patient-reported outcomes in moderate to severe systemic lupus erythematosus: a post-hoc analysis of the phase 3 TULIP-1 and TULIP-2 trials of anifrolumab. Lancet Rheumatol. 2022;4(3):e198–207.
  • Chaussabel D, Quinn C, Shen J, et al. A modular analysis framework for blood genomics studies: application to systemic lupus erythematosus. Immunity. 2008 Jul 18;29(1):150–164. DOI:10.1016/j.immuni.2008.05.012
  • Bodewes IL, Björk A, Versnel MA, et al. Innate immunity and interferons in the pathogenesis of sjögren’s syndrome. Rheumatology. 2021;60(6):2561–2573. DOI:10.1093/rheumatology/key360
  • Nezos A, Gravani F, Tassidou A, et al. Type I and II interferon signatures in sjogren’s syndrome pathogenesis: contributions in distinct clinical phenotypes and sjogren’s related lymphomagenesis. J Autoimmun. 2015;63:47–58.
  • Seguier J, Jouve E, Bobot M, et al. Paradoxical association between blood modular interferon signatures and quality of life in patients with systemic lupus erythematosus, Rheumatology, Volume 59, Issue 8, August 2020. https://doi.org/10.1093/rheumatology/kez541
  • Chiche L, Seguier J, Gentile S, et al., editors. Unexpected association between health-related quality of life and the blood interferon modular transcriptional signatures in patients with systemic lupus erythematosus. 2018 ACR/ARHP Annual Meeting; 2018.
  • Bentley D, Fisher BA, Barone F, et al. A randomized, double-blind, placebo-controlled, parallel group study on the effects of a cathepsin S inhibitor in primary sjögren’s syndrome. Rheumatology (Oxford). 2023 Mar 2. DOI:10.1093/rheumatology/kead092
  • Levavi H, Lancman G, Gabrilove J. Impact of rituximab on COVID-19 outcomes. Ann Hematol. 2021;100(11):2805–2812.
  • Bacman S, Berra A, Sterin-Borda L, et al. Muscarinic acetylcholine receptor antibodies as a new marker of dry eye sjogren syndrome. Invest Ophthalmol Visual Sci. 2001;42(2):321–327.
  • Goebeler M, Bata‐Csörgő Z, De Simone C, et al. Treatment of pemphigus vulgaris and foliaceus with efgartigimod, a neonatal fc receptor inhibitor: a phase II multicentre, open‐label feasibility trial. Br J Dermatol. 2022;186(3):429–439. DOI:10.1111/bjd.20782
  • Lünemann JD. Getting specific: targeting fc receptors in myasthenia gravis. Nat Rev Neurol. 2021;17(10):597–598.
  • Nelke C, Spatola M, Schroeter CB, et al. Neonatal fc receptor–Targeted therapies in neurology. Neurotherapeutics. 2022;19(3):1–12. DOI:10.1007/s13311-021-01175-7

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.