Emerging Trends and Management for Sjögren Syndrome-Related Dry Eye Corneal Alterations
Abstract
:1. Introduction
2. Sjögren’s Syndrome Clinical Manifestations and Ocular Involvement
3. Glandular Ocular Involvement: Dry Eye Disease (DED) or Keratoconjunctivitis Sicca (KCS)
3.1. Histology, Biochemistry, Anatomy, and Physiology
3.1.1. Salivary and Lachrymal Glands
3.1.2. Tear Film
3.1.3. Meibomian Glands
3.1.4. Conjunctiva
3.1.5. Cornea
3.2. Clinical Characteristics
- -
- -
- More complex eye dryness (aqueous deficient + evaporative): Although SS-related DED is classified as a form of ADDE [23], an associated damage of conjunctival goblet cells and Meibomian glands may be present in 45–60% of cases [12,29]. SS-related DED should thus be regarded as a more complex and severe dry eye involving all tear film components, where the aqueous-deficient DED, considered the hallmark of the disease, is complicated by mucous and lipid layers instability and tears evaporation [39];
- -
- More severe ocular surface inflammation and damage: as compared with other DED forms, the SS-related DED is characterized by significantly higher levels of inflammatory biomarkers on the ocular surface [27] and by more severe ocular surface inflammation and damage and visual difficulties [6,7,12,17,38];
- -
- Neurotrophic component and typical discordance between clinical signs and symptoms: Patients with pSS are frequently affected by functional corneal nerves abnormalities. SS-related DED seems to be associated with less severe symptoms of ocular discomfort than non-SS DED forms or similar symptoms in the presence of significantly worse signs in SS patients [38], probably because of the higher degree of corneal nerves dysfunction found in SS patients, especially in more severe and long-lasting cases [40]. Conversely, other SS patients, especially women, may show corneal hypersensitivity, with neuropathic pain and light sensitivity, which may be caused by neuroinflammation [40]. The structural [32,34] and functional [40] corneal nerves abnormalities found in SS patients suggest that the SS-related dry eye disease may be regarded as a form of neurotrophic keratopathy [40];
- -
- -
- Higher infective risk: pSS patients show an increased risk of bacterial and fungal infections of the ocular surface [41]. The incidence of the infectious keratitis in the USA general population ranges between 0.0025% to 0.28%, whereas it is approximately 2% in SS patients, with a recurrence in 17% of cases [41];
- -
3.3. Pathogenesis
- -
- -
- Autoimmune attack against the ocular surface epithelium, conjunctival goblet cells, and Meibomian glands, with reduction in all tear film components, as well as epithelial cells autoantigens expression and proinflammatory molecules release [50];
- -
- -
- -
- Decreased levels of growth factors on the ocular surface, including epidermal growth factor secreted by the lachrymal gland, nerve growth factors (NGFs) released by the corneal nerves, and transforming growth factor produced by the conjunctival goblet cells [27], with impairment of the epithelial cell proliferation, differentiation, and migration;
- -
- Dysbiosis of the ocular surface, which may trigger an autoimmune attack via autoantigens-mimicry mechanisms and may increase infection susceptibility [28];
- -
- -
3.4. Treatment Strategies
- -
- -
- Multidisciplinary care team: The involvement of other specialists, such as odontostomatologists, rheumatologists, and immunologists, is of fundamental importance in order to establish glandular and extraglandular systemic involvement and disease activity. The presence of severe vision-threatening ocular complications or systemic manifestation requires a prompt multidisciplinary approach and systemic immunosuppressive therapy prescribed by a rheumatologist/immunologist [42,43,44,55,56];
- -
- Stepwise approach: the treatment should start with the patient’s education about useful lifestyle modifications (Table 2) and with the prescription of artificial tear substitutes (eyedrops, gels, or ointments), which remain the first-line therapy in patients with DED of any cause [59] (Table 3). Other topical (Table 3), mechanical (Table 3), systemic, or surgical therapies should be recommended as second-line approaches depending on the ocular and systemic associated complications [42,43,44,55,56]. The treatment of the SS-related KCS is generally limited to topical or mechanical approaches (Table 3). In a subset of SS patients, DED is unresponsive to common treatment, resulting in severe clinical manifestations, including recurrent corneal infections and persistent corneal abrasion or scarring, which may require more complex treatments.
4. Extraglandular Corneal Complications
4.1. Clinical Manifestations
4.2. Pathophysiological Mechanisms
- -
- High levels of proinflammatory cytokines, reduced levels of lacrimal gland-derived factors such as the epidermal growth factor, and neurodegeneration with decreased NGFs levels may impair the integrity, proliferation, differentiation, and migration of the ocular surface epithelium, leading to punctata keratoconjunctivitis, corneal erosion, and ulceration [45,46,47];
- -
- Neuroinflammation, neurodegeneration, and consequently decreased levels of nerve growth factors (NGFs) may lead to a neurotrophic keratopathy-like syndrome characterized by corneal hypo-aesthesia and reduced lachrymal reflex with decreased aqueous tear production, blinking impairment, and reduced trophism, apoptosis, and impaired regeneration of epithelial cells and stromal keratocytes, which may lead to corneal erosion/ulcer [45,46,47];
- -
- autoimmune attack against connective tissue proteins: As part of the AI connective tissue diseases (CTDs), SS represents a multisystem AI disorder affecting the connective tissue proteins [5]. Both connective and sclera-corneal tissues contain collagen and proteoglycans that may become the target of a cell-mediated and antibody-mediated autoimmune attack inducing the destruction of the peripheral corneal stroma via matrix metalloproteinases activation [88].
4.3. Therapeutic Interventions
- -
- Human amniotic membrane (AM) transplantation: AM implantation has shown several therapeutic properties, including analgesic, re-epithelizing, anti-fibrotic, antimicrobial, anti-inflammatory, anti-angiogenic, and anti-adhesive effects, and it has been successfully used as temporary biologic bandage in a wide range of the ocular surface disorders [105], such as severe SS-related KCS refractory to standard therapies or to promote healing of corneal ulceration/melting/perforations and PUKs [106,107];
- -
- Cyanoacrylate tissue adhesive (CTA) application represents a routine approach to corneal melts and small uncomplicated perforations (<2–3 mm in diameter) of different causes and also in pSS and sSS patients [108]. Although CTA application is generally used as a temporizing measure to delay penetrating keratoplasty, in 20–45% of cases, it provides satisfactory corneal healing, avoiding more invasive surgical interventions [108];
- -
- Conjunctival flap graft may be used to restore ocular surface integrity in different diseases [109];
- -
- -
- Full-thickness or lamellar corneal/scleral patch grafting is indicated in cases of corneal descemetocele, melting, perforation that cannot be closed with tissue adhesives or AM transplantation, or in eyes with acute inflammation, in which a traditional PK may be at high risk of rejection, aiming to postpone a larger refractive PK until systemic therapy with immunosuppressive and anti-collagenolytic treatments is effective [111]. Several case reports of tectonic corneal/scleral patch grafting performed in SS patients with descemetocele, melting, PUK, or perforation have been published [111,112];
- -
- -
- Keratoprosthesis implantation involves the implant of a synthetic structure that replaces the central portion of a diseased cornea, which is indicated in cases with end-stage corneal blindness and where a routine keratoplasty ± limbal stem cells transplantation and systemic immunosuppressant therapy may have a high risk of failure [116]. A total of 18 eyes affected by corneal melting/perforation in SS patients have been described in the literature [116].
5. Conclusions and Future Directions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Negrini, S.; Emmi, G.; Greco, M.; Borro, M.; Sardanelli, F.; Murdaca, G.; Indiveri, F.; Puppo, F. Sjögren’s syndrome: A systemic autoimmune disease. Clin. Exp. Med. 2022, 22, 9–25. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Akpek, E.K.; Bunya, V.Y.; Saldanha, I.J. Sjögren’s Syndrome: More Than Just Dry Eye. Cornea 2019, 38, 658–661. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Maleki-Fischbach, M.; Kastsianok, L.; Koslow, M.; Chan, E.D. Manifestations and management of Sjögren’s disease. Arthritis Res. Ther. 2024, 26, 43. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Kollert, F.; Fisher, B.A. Equal rights in autoimmunity: Is Sjögren’s syndrome ever ‘secondary’? Rheumatology 2020, 59, 1218–1225. [Google Scholar] [CrossRef] [PubMed]
- Asif, M.I.; Kalra, N.; Sharma, N.; Jain, N.; Sharma, M.; Sinha, R. Connective tissue disorders and eye: A review and recent updates. Indian. J. Ophthalmol. 2023, 71, 2385–2398. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Bjordal, O.; Norheim, K.B.; Rødahl, E.; Jonsson, R.; Omdal, R. Primary Sjögren’s syndrome and the eye. Surv. Ophthalmol. 2020, 65, 119–132. [Google Scholar] [CrossRef] [PubMed]
- Roszkowska, A.M.; Oliverio, G.W.; Aragona, E.; Inferrera, L.; Severo, A.A.; Alessandrello, F.; Spinella, R.; Postorino, E.I.; Aragona, P. Ophthalmologic Manifestations of Primary Sjögren’s Syndrome. Genes 2021, 12, 365. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Mathews, P.M.; Robinson, S.A.; Gire, A.; Baer, A.N.; Akpek, E.K. Extraglandular ocular involvement and morbidity and mortality in primary Sjögren’s Syndrome. PLoS ONE 2020, 15, e0239769. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Singh, S.; Das, A.V.; Basu, S. Ocular Involvement in Sjögren Syndrome: Risk Factors for Severe Visual Impairment and Vision-Threatening Corneal Complications. Am. J. Ophthalmol. 2021, 225, 11–17. [Google Scholar] [CrossRef] [PubMed]
- Iyer, G.; Srinivasan, B. Sjogren’s disease and the cornea: Bridging disciplines for enhanced ocular health. Kerala J. Ophthalmol. 2024, 36, 220–223. [Google Scholar] [CrossRef]
- Shah, R.; Amador, C.; Tormanen, K.; Ghiam, S.; Saghizadeh, M.; Arumugaswami, V.; Kumar, A.; Kramerov, A.A.; Ljubimov, A.V. Systemic diseases and the cornea. Exp. Eye Res. 2021, 204, 108455. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Villarreal-Gonzalez, A.J.; Jocelyn Rivera-Alvarado, I.; Rodriguez-Gutierrez, L.A.; Rodriguez-Garcia, A. Analysis of ocular surface damage and visual impact in patients with primary and secondary Sjögren syndrome. Rheumatol. Int. 2020, 40, 1249–1257. [Google Scholar] [CrossRef] [PubMed]
- Singh, S.; Chaudhary, S.; Das, A.V.; Basu, S. Presentation, aetiology and outcomes of corneal ulceration in Sjogren’s Syndrome. Eye 2023, 37, 3217–3220. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Beydon, M.; McCoy, S.; Nguyen, Y.; Sumida, T.; Mariette, X.; Seror, R. Epidemiology of Sjögren syndrome. Nat. Rev. Rheumatol. 2024, 20, 158–169. [Google Scholar] [CrossRef] [PubMed]
- Xuan, Y.; Zhang, X.; Wu, H. Impact of sex differences on the clinical presentation, pathogenesis, treatment and prognosis of Sjögren’s syndrome. Immunology 2024, 171, 513–524. [Google Scholar] [CrossRef] [PubMed]
- Baer, A.N.; Walitt, B. Update on Sjögren Syndrome and Other Causes of Sicca in Older Adults. Rheum. Dis. Clin. N. Am. 2018, 44, 419–436. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Wang, H.; Wang, P.B.; Chen, T.; Zou, J.; Li, Y.J.; Ran, X.F.; Li, Q.X. Analysis of Clinical Characteristics of Immune-Related Dry Eye. J. Ophthalmol. 2017, 2017, 8532397. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Shiboski, C.H.; Shiboski, S.C.; Seror, R.; Criswell, L.A.; Labetoulle, M.; Lietman, T.M.; Rasmussen, A.; Scofield, H.; Vitali, C.; Bowman, S.J.; 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, 35–45. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Bautista-Vargas, M.; Vivas, A.J.; Tobón, G.J. Minor salivary gland biopsy: Its role in the classification and prognosis of Sjögren’s syndrome. Autoimmun. Rev. 2020, 19, 102690. [Google Scholar] [CrossRef] [PubMed]
- Vílchez-Oya, F.; Balastegui Martin, H.; García-Martínez, E.; Corominas, H. Not all autoantibodies are clinically relevant. Classic and novel autoantibodies in Sjögren’s syndrome: A critical review. Front. Immunol. 2022, 13, 1003054. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Brott, N.R.; Zeppieri, M.; Ronquillo, Y. Schirmer Test. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2025. [Google Scholar] [PubMed]
- Srinivas, S.P.; Rao, S.K. Ocular surface staining: Current concepts and techniques. Indian. J. Ophthalmol. 2023, 71, 1080–1089. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Craig, J.P.; Nichols, K.K.; Akpek, E.K.; Caffery, B.; Dua, H.S.; Joo, C.K.; Liu, Z.; Nelson, J.D.; Nichols, J.J.; Tsubota, K.; et al. TFOS DEWS II Definition and Classification Report. Ocul. Surf. 2017, 15, 276–283. [Google Scholar] [CrossRef] [PubMed]
- Xu, K.P.; Katagiri, S.; Takeuchi, T.; Tsubota, K. Biopsy of labial salivary glands and lacrimal glands in the diagnosis of Sjögren’s syndrome. J. Rheumatol. 1996, 23, 76–82. [Google Scholar] [PubMed]
- Kaneko, N.; Chen, H.; Perugino, C.A.; Maehara, T.; Munemura, R.; Yokomizo, S.; Sameshima, J.; Diefenbach, T.J.; Premo, K.R.; Chinju, A.; et al. Cytotoxic CD8+ T cells may be drivers of tissue destruction in Sjögren’s syndrome. Sci. Rep. 2022, 12, 15427. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Pringle, S.; Verstappen, G.M.; van Ginkel, M.S.; Nakshbandi, U.; Girigoria, Z.; Bootsma, H.; van der Vegt, B.; Kroese, F.G.M. Lymphoepithelial lesions in the salivary glands of primary Sjögren’s syndrome patients: The perfect storm? Clin. Exp. Rheumatol. 2022, 40, 2434–2442. [Google Scholar] [CrossRef] [PubMed]
- Peng, J.; Feinstein, D.; DeSimone, S.; Gentile, P. A Review of the Tear Film Biomarkers Used to Diagnose Sjogren’s Syndrome. Int. J. Mol. Sci. 2024, 25, 10380. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Pal, S.; Vani, G.; Donthineni, P.R.; Basu, S.; Arunasri, K. Tear film microbiome in Sjogren’s and non-Sjogren’s aqueous deficiency dry eye. Indian. J. Ophthalmol. 2023, 71, 1566–1573. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Sullivan, D.A.; Dana, R.; Sullivan, R.M.; Krenzer, K.L.; Sahin, A.; Arica, B.; Liu, Y.; Kam, W.R.; Papas, A.S.; Cermak, J.M. Meibomian Gland Dysfunction in Primary and Secondary Sjögren Syndrome. Ophthalmic Res. 2018, 59, 193–205. [Google Scholar] [CrossRef] [PubMed]
- Wakamatsu, T.H.; Sato, E.A.; Matsumoto, Y.; Ibrahim, O.M.; Dogru, M.; Kaido, M.; Ishida, R.; Tsubota, K. Conjunctival in vivo confocal scanning laser microscopy in patients with Sjögren syndrome. Investig. Ophthalmol. Vis. Sci. 2010, 51, 144–150. [Google Scholar] [CrossRef] [PubMed]
- de Paiva, C.S.; Trujillo-Vargas, C.M.; Schaefer, L.; Yu, Z.; Britton, R.A.; Pflugfelder, S.C. Differentially Expressed Gene Pathways in the Conjunctiva of Sjögren Syndrome Keratoconjunctivitis Sicca. Front. Immunol. 2021, 12, 702755. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Matsumoto, Y.; Ibrahim, O.M.A.; Kojima, T.; Dogru, M.; Shimazaki, J.; Tsubota, K. Corneal In Vivo Laser-Scanning Confocal Microscopy Findings in Dry Eye Patients with Sjögren’s Syndrome. Diagnostics 2020, 10, 497. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Hao, R.; Ding, Y.; Li, X. Alterations in corneal epithelial dendritic cell in Sjogren’s syndrome dry eye and clinical correlations. Sci. Rep. 2022, 12, 11167. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Hao, R.; Chou, Y.; Ding, Y.; Liu, Z.; Wang, Y.; Ren, X.; Li, X. Corneal subbasal nerve analysis in patients with primary Sjogren’s syndrome: A novel objective grading method and clinical correlations. Int. Ophthalmol. 2023, 43, 779–793. [Google Scholar] [CrossRef] [PubMed]
- Borrego-Sanz, L.; Sáenz-Francés San Baldomero, F.; Díaz Valle, D.; Santos Bueso, E.; Sánchez Jean, R.; Martínez de la Casa, J.M.; Benítez Del Castillo, J.M.; García Feijóo, J.; Rodríguez Rodríguez, L. Comparison of corneal biomechanical properties of patients with dry eye secondary to Sjögren’s syndrome and healthy subjects. J. Fr. Ophtalmol. 2018, 41, 802–808. [Google Scholar] [CrossRef] [PubMed]
- Chung, J.K.; Kim, M.K.; Wee, W.R. Prognostic factors for the clinical severity of keratoconjunctivitis sicca in patients with Sjogren’s syndrome. Br. J. Ophthalmol. 2012, 96, 240–245. [Google Scholar] [CrossRef] [PubMed]
- Miyamoto, S.T.; Valim, V.; Fisher, B.A. Health-related quality of life and costs in Sjögren’s syndrome. Rheumatology 2021, 60, 2588–2601. [Google Scholar] [CrossRef] [PubMed]
- Xiong, F.; Pula, D.; Akpek, E.K.; Bunya, V.Y.; Shiboski, C.H.; Lietman, T.L.; Gonzales, J.A. Sjögren’s Versus Non-Sjögren’s Ocular Features: Similar Symptoms, But Significantly Worse Signs. Invest. Ophthalmol. Vis. Sci. 2024, 65, 23. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Wang, Y.; Qin, Q.; Liu, B.; Fu, Y.; Lin, L.; Huang, X.; Jin, X. Clinical Analysis: Aqueous-Deficient and Meibomian Gland Dysfunction in Patients With Primary Sjogren’s Syndrome. Front. Med. 2019, 6, 291. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Sanchez, V.; Dobzinski, N.; Fox, R.; Galor, A. Rethinking Sjögren Beyond Inflammation: Considering the Role of Nerves in Driving Disease Manifestations. Eye Contact Lens 2024, 50, 200–207. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Kim, S.Y.; Kim, S.H.; Yoon, C.H.; Kim, M.K.; Oh, J.Y. Infectious Keratitis in Patients with Ocular Sjögren’s Syndrome. Korean J. Ophthalmol. 2022, 36, 407–412. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Ramos-Casals, M.; Brito-Zerón, P.; Bombardieri, S.; Bootsma, H.; De Vita, S.; Dörner, T.; Fisher, B.A.; Gottenberg, J.E.; Hernandez-Molina, G.; Kocher, A.; et al. EULAR recommendations for the management of Sjögren’s syndrome with topical and systemic therapies. Ann. Rheum. Dis. 2020, 79, 3–18. [Google Scholar] [CrossRef] [PubMed]
- Price, E.J.; Baer, A.N. How to treat Sjögren’s syndrome. Rheumatology 2021, 60, 2574–2587. [Google Scholar] [CrossRef] [PubMed]
- Price, E.J.; Benjamin, S.; Bombardieri, M.; Bowman, S.; Carty, S.; Ciurtin, C.; Crampton, B.; Dawson, A.; Fisher, B.A.; Giles, I.; et al. British Society for Rheumatology guideline on management of adult and juvenile onset Sjögren disease. Rheumatology 2025, 64, 409–439. [Google Scholar] [CrossRef] [PubMed]
- Zhan, Q.; Zhang, J.; Lin, Y.; Chen, W.; Fan, X.; Zhang, D. Pathogenesis and treatment of Sjogren’s syndrome: Review and update. Front. Immunol. 2023, 14, 1127417. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Baldini, C.; Fulvio, G.; La Rocca, G.; Ferro, F. Update on the pathophysiology and treatment of primary Sjögren syndrome. Nat. Rev. Rheumatol. 2024, 20, 473–491. [Google Scholar] [CrossRef] [PubMed]
- Bron, A.J.; de Paiva, C.S.; Chauhan, S.K.; Bonini, S.; Gabison, E.E.; Jain, S.; Knop, E.; Markoulli, M.; Ogawa, Y.; Perez, V.; et al. TFOS DEWS II pathophysiology report. Ocul. Surf. 2017, 15, 438–510, Erratum in Ocul. Surf. 2019, 17, 842. https://doi.org/10.1016/j.jtos.2019.08.007. [Google Scholar] [CrossRef] [PubMed]
- Chen, Y.; Wang, S.; Alemi, H.; Dohlman, T.; Dana, R. Immune regulation of the ocular surface. Exp. Eye Res. 2022, 218, 109007. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Utomo, S.W.; Putri, J.F. Infections as Risk Factor of Sjögren’s Syndrome. Open Access Rheumatol. 2020, 12, 257–266. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Ogawa, Y.; Takeuchi, T.; Tsubota, K. Autoimmune Epithelitis and Chronic Inflammation in Sjögren’s Syndrome-Related Dry Eye Disease. Int. J. Mol. Sci. 2021, 22, 11820. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Chivasso, C.; Sarrand, J.; Perret, J.; Delporte, C.; Soyfoo, M.S. The Involvement of Innate and Adaptive Immunity in the Initiation and Perpetuation of Sjögren’s Syndrome. Int. J. Mol. Sci. 2021, 22, 658. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Khatri, B.; Tessneer, K.L.; Rasmussen, A.; Aghakhanian, F.; Reksten, T.R.; Adler, A.; Alevizos, I.; Anaya, J.-M.; Aqrawi, L.A.; Baecklund, E.; et al. Genome-wide association study identifies Sjögren’s risk loci with functional implications in immune and glandular cells. Nat. Commun. 2022, 13, 4287, Erratum in Nat. Commun. 2022, 13, 6519. https://doi.org/10.1038/s41467-022-34311-8; Erratum in Nat. Commun. 2023, 14, 598. https://doi.org/10.1038/s41467-023-36369-4. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Thorlacius, G.E.; Björk, A.; Wahren-Herlenius, M. Genetics and epigenetics of primary Sjögren syndrome: Implications for future therapies. Nat. Rev. Rheumatol. 2023, 19, 288–306. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Alam, J.; de Paiva, C.S.; Pflugfelder, S.C. Desiccation Induced Conjunctival Monocyte Recruitment and Activation—Implications for Keratoconjunctivitis. Front. Immunol. 2021, 12, 701415. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Jones, L.; Downie, L.E.; Korb, D.; Benitez-Del-Castillo, J.M.; Dana, R.; Deng, S.X.; Dong, P.N.; Geerling, G.; Hida, R.Y.; Liu, Y.; et al. TFOS DEWS II Management and Therapy Report. Ocul. Surf. 2017, 15, 575–628. [Google Scholar] [CrossRef] [PubMed]
- Amescua, G.; Ahmad, S.; Cheung, A.Y.; Choi, D.S.; Jhanji, V.; Lin, A.; Mian, S.I.; Rhee, M.K.; Viriya, E.T.; Mah, F.S.; et al. Dry Eye Syndrome Preferred Practice Pattern®. Ophthalmology 2024, 131, P1–P49. [Google Scholar] [CrossRef] [PubMed]
- Shih, K.C.; Lun, C.N.; Jhanji, V.; Thong, B.Y.; Tong, L. Systematic review of randomized controlled trials in the treatment of dry eye disease in Sjogren syndrome. J. Inflamm. 2017, 14, 26. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Brito-Zerón, P.; Retamozo, S.; Kostov, B.; Baldini, C.; Bootsma, H.; De Vita, S.; Dörner, T.; Gottenberg, J.E.; Kruize, A.A.; Mandl, T.; et al. Efficacy and safety of topical and systemic medications: A systematic literature review informing the EULAR recommendations for the management of Sjögren’s syndrome. RMD Open 2019, 5, e001064. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Barabino, S.; Benitez-Del-Castillo, J.M.; Fuchsluger, T.; Labetoulle, M.; Malachkova, N.; Meloni, M.; Utheim, T.P.; Rolando, M. Dry eye disease treatment: The role of tear substitutes, their future, and an updated classification. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 8642–8652. [Google Scholar] [CrossRef] [PubMed]
- Goldstein, M.H.; Silva, F.Q.; Blender, N.; Tran, T.; Vantipalli, S. Ocular benzalkonium chloride exposure: Problems and solutions. Eye 2022, 36, 361–368. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Pucker, A.D.; Ng, S.M.; Nichols, J.J. Over the counter (OTC) artificial tear drops for dry eye syndrome. Cochrane Database Syst. Rev. 2016, 2, CD009729. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Qiu, W.; Liu, Z.; Ao, M.; Li, X.; Wang, W. Punctal plugs versus artificial tears for treating primary Sjögren’s syndrome with keratoconjunctivitis SICCA: A comparative observation of their effects on visual function. Rheumatol. Int. 2013, 33, 2543–2548. [Google Scholar] [CrossRef] [PubMed]
- Vazirani, J.; Sridhar, U.; Gokhale, N.; Doddigarla, V.R.; Sharma, S.; Basu, S. Autologous serum eye drops in dry eye disease: Preferred practice pattern guidelines. Indian J. Ophthalmol. 2023, 71, 1357–1363. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Pan, Q.; Angelina, A.; Marrone, M.; Stark, W.J.; Akpek, E.K. Autologous serum eye drops for dry eye. Cochrane Database Syst. Rev. 2017, 2, CD009327. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Rodríguez Calvo-de-Mora, M.; Domínguez-Ruiz, C.; Barrero-Sojo, F.; Rodríguez-Moreno, G.; Antúnez Rodríguez, C.; Ponce Verdugo, L.; Hernández Lamas, M.D.C.; Hernández-Guijarro, L.; Villalvilla Castillo, J.; Fernández-Baca Casares, I.; et al. Autologous versus allogeneic versus umbilical cord sera for the treatment of severe dry eye disease: A double-blind randomized clinical trial. Acta Ophthalmol. 2022, 100, e396–e408. [Google Scholar] [CrossRef] [PubMed]
- Wróbel-Dudzińska, D.; Przekora, A.; Kazimierczak, P.; Ćwiklińska-Haszcz, A.; Kosior-Jarecka, E.; Żarnowski, T. The Comparison between the Composition of 100% Autologous Serum and 100% Platelet-Rich Plasma Eye Drops and Their Impact on the Treatment Effectiveness of Dry Eye Disease in Primary Sjogren Syndrome. J. Clin. Med. 2023, 12, 3126. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Aragona, P.; Stilo, A.; Ferreri, F.; Mobrici, M. Effects of the topical treatment with NSAIDs on corneal sensitivity and ocular surface of Sjögren’s syndrome patients. Eye 2005, 19, 535–539. [Google Scholar] [CrossRef] [PubMed]
- Rigas, B.; Huang, W.; Honkanen, R. NSAID-induced corneal melt: Clinical importance, pathogenesis, and risk mitigation. Surv. Ophthalmol. 2020, 65, 1–11. [Google Scholar] [CrossRef] [PubMed]
- Murtagh, P.; Comer, R.; Fahy, G. Corneal perforation in undiagnosed Sjögren’s syndrome following topical NSAID and steroid drops post routine cataract extraction. BMJ Case Rep. 2018, 2018, bcr2018225428. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Hammad, M.; Ahmed, L.; Seifelnasr, M. NSAID-induced corneal melt as the first presentation of Sjögren’s syndrome. BMJ Case Rep. 2024, 17, e258829. [Google Scholar] [CrossRef] [PubMed]
- Liu, S.H.; Saldanha, I.J.; Abraham, A.G.; Rittiphairoj, T.; Hauswirth, S.; Gregory, D.; Ifantides, C.; Li, T. Topical corticosteroids for dry eye. Cochrane Database Syst. Rev. 2022, 10, CD015070. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Aragona, P.; Spinella, R.; Rania, L.; Postorino, E.; Sommario, M.S.; Roszkowska, A.M.; Puzzolo, D. Safety and efficacy of 0.1% clobetasone butyrate eyedrops in the treatment of dry eye in Sjögren syndrome. Eur. J. Ophthalmol. 2013, 23, 368–376. [Google Scholar] [CrossRef] [PubMed]
- Lin, T.; Gong, L. Topical fluorometholone treatment for ocular dryness in patients with Sjögren syndrome: A randomized clinical trial in China. Medicine 2015, 94, e551. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Kapugi, M.; Cunningham, K. Corticosteroids. Orthop. Nurs. 2019, 38, 336–339. [Google Scholar] [CrossRef] [PubMed]
- Flts, A.; Medina, R.; Akpek, E.K. The evolution of cyclosporine treatments for treatment of ocular surface diseases. Curr. Opin. Allergy Clin. Immunol. 2024, 24, 360–367. [Google Scholar] [CrossRef] [PubMed]
- Sall, K.; Stevenson, O.D.; Mundorf, T.K.; Reis, B.L. Two multicenter, randomized studies of the efficacy and safety of cyclosporine ophthalmic emulsion in moderate to severe dry eye disease. CsA Phase 3 Study Group. Ophthalmology 2000, 107, 631–639, Erratum in Ophthalmology 2000, 107, 1220. [Google Scholar] [CrossRef] [PubMed]
- de Paiva, C.S.; Pflugfelder, S.C.; Ng, S.M.; Akpek, E.K. Topical cyclosporine A therapy for dry eye syndrome. Cochrane Database Syst. Rev. 2019, 9, CD010051. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Levy, O.; Labbé, A.; Borderie, V.; Hamiche, T.; Dupas, B.; Laroche, L.; Baudouin, C.; Bouheraoua, N. Increased corneal sub-basal nerve density in patients with Sjögren syndrome treated with topical cyclosporine A. Clin. Exp. Ophthalmol. 2017, 45, 455–463. [Google Scholar] [CrossRef] [PubMed]
- Haber, S.L.; Benson, V.; Buckway, C.J.; Gonzales, J.M.; Romanet, D.; Scholes, B. Lifitegrast: A novel drug for patients with dry eye disease. Ther. Adv. Ophthalmol. 2019, 11, 2515841419870366. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Guimaraes de Souza, R.; Yu, Z.; Stern, M.E.; Pflugfelder, S.C.; de Paiva, C.S. Suppression of Th1-Mediated Keratoconjunctivitis Sicca by Lifitegrast. J. Ocul. Pharmacol. Ther. 2018, 34, 543–549. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Ervin, A.M.; Law, A.; Pucker, A.D. Punctal occlusion for dry eye syndrome. Cochrane Database Syst. Rev. 2017, 6, CD006775. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Ahn, H.; Ji, Y.W.; Jun, I.; Kim, T.I.; Lee, H.K.; Seo, K.Y. Comparison of Treatment Modalities for Dry Eye in Primary Sjögren’s Syndrome. J. Clin. Med. 2022, 11, 463. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Lim, L.; Lim, E.W.L. Therapeutic Contact Lenses in the Treatment of Corneal and Ocular Surface Diseases-A Review. Asia Pac. J. Ophthalmol. 2020, 9, 524–532. [Google Scholar] [CrossRef] [PubMed]
- Romano, V.; Romano, D.; Semeraro, P.; Forbice, E.; Iaria, A.; Pizzolante, T.; Frassi, M.; Franceschini, F.; Semeraro, F. Therapeutic Hyper-CL soft contact lens in Sjögren’s syndrome. Am. J. Ophthalmol. Case Rep. 2022, 28, 101685. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Acharya, M.; Dave, A. Commentary: Corneal involvement in rheumatoid arthritis. Indian J. Ophthalmol. 2021, 69, 73–74. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Tzamalis, A.; Matsou, A.; Anastasopoulos, E.; Ziakas, N. Treatment of spontaneous corneal perforation secondary to undiagnosed Sjögren’s syndrome using regenerating agent and autologous serum eye drops. Eur. J. Ophthalmol. 2021, 31, NP17–NP21. [Google Scholar] [CrossRef] [PubMed]
- Hassanpour, K.; HElSheikh, R.; Arabi, A.; RFrank, C.; MElhusseiny, A.; KEleiwa, T.; Arami, S.; RDjalilian, A.; Kheirkhah, A. Peripheral Ulcerative Keratitis: A Review. J. Ophthalmic Vis. Res. 2022, 17, 252–275. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Cao, Y.; Zhang, W.; Wu, J.; Zhang, H.; Zhou, H. Peripheral Ulcerative Keratitis Associated with Autoimmune Disease: Pathogenesis and Treatment. J. Ophthalmol. 2017, 2017, 7298026. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Dutta Majumder, P.; Agarwal, S.; Shah, M.; Srinivasan, B.; K, P.; Iyer, G.; Sharma, N.; Biswas, J.; McCluskey, P. Necrotizing Scleritis: A Review. Ocul. Immunol. Inflamm. 2024, 32, 1405–1419. [Google Scholar] [CrossRef] [PubMed]
- Bamrolia, N.R.; Arora, R.; Yadava, U. Unusual presentation of a case of Sjogren’s syndrome with neurological and ocular manifestation. Contact Lens Anterior Eye 2012, 35, 85–88. [Google Scholar] [CrossRef] [PubMed]
- Narain, S.; Pyare, R.; Rath, P.D. A Rare Presentation: Bilateral Necrotizing Scleritis in Primary Sjögren’s Syndrome. Ocul. Immunol. Inflamm. 2025, 33, 146–148. [Google Scholar] [CrossRef] [PubMed]
- Brejchova, K.; Liskova, P.; Hrdlickova, E.; Filipec, M.; Jirsova, K. Matrix metalloproteinases in recurrent corneal melting associated with primary Sjörgen’s syndrome. Mol. Vis. 2009, 15, 2364–2372. [Google Scholar] [PubMed] [PubMed Central]
- Papas, A.S.; Sherrer, Y.S.; Charney, M.; Golden, H.E.; Medsger, T.A., Jr.; Walsh, B.T.; Trivedi, M.; Goldlust, B.; Gallagher, S.C. Successful Treatment of Dry Mouth and Dry Eye Symptoms in Sjögren’s Syndrome Patients With Oral Pilocarpine: A Randomized, Placebo-Controlled, Dose-Adjustment Study. J. Clin. Rheumatol. 2004, 10, 169–177. [Google Scholar] [CrossRef] [PubMed]
- Felberg, S.; Dantas, P.E.C.; Sato, E.H. Oral pilocarpine for the treatment of dry eye in patients with Sjögren’s syndrome. Arq. Bras. Oftalmol. 2022, 85, 269–276. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Yoon, C.H.; Lee, H.J.; Lee, E.Y.; Lee, E.B.; Lee, W.W.; Kim, M.K.; Wee, W.R. Effect of Hydroxychloroquine Treatment on Dry Eyes in Subjects with Primary Sjögren’s Syndrome: A Double-Blind Randomized Control Study. J. Korean Med. Sci. 2016, 31, 1127–1135. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Wang, S.Q.; Zhang, L.W.; Wei, P.; Hua, H. Is hydroxychloroquine effective in treating primary Sjogren’s syndrome: A systematic review and meta-analysis. BMC Musculoskelet. Disord. 2017, 18, 186. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Betül Türkoğlu, E.; Tuna, S.; Alan, S.; İhsan Arman, M.; Tuna, Y.; Ünal, M. Effect of Systemic Infliximab Therapy in Patients with Sjögren’s Syndrome. Turk. J. Ophthalmol. 2015, 45, 138–141. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Souza, F.B.; Porfírio, G.J.; Andriolo, B.N.; Albuquerque, J.V.; Trevisani, V.F. Rituximab Effectiveness and Safety for Treating Primary Sjögren’s Syndrome (pSS): Systematic Review and Meta-Analysis. PLoS ONE 2016, 11, e0150749. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Chu, L.L.; Cui, K.; Pope, J.E. Meta-Analysis of Treatment for Primary Sjögren’s Syndrome. Arthritis Care Res. 2020, 72, 1011–1021. [Google Scholar] [CrossRef] [PubMed]
- Álvarez-Rivas, N.; Sang-Park, H.; Díaz Del Campo, P.; Fernández-Castro, M.; Corominas, H.; Andreu, J.L.; Navarro-Compán, V. Efficacy of belimumab in Primary Sjögren’s syndrome: A systematic review. Reumatol. Clin. 2021, 17, 170–174. [Google Scholar] [CrossRef] [PubMed]
- Baer, A.N.; Gottenberg, J.E.; St Clair, E.W.; Sumida, T.; Takeuchi, T.; Seror, R.; Foulks, G.; Nys, M.; Mukherjee, S.; Wong, R.; 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, 80, 339–348. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Utine, C.A.; Birlik, M.; Özizmirliler, D.; Karakaş, A.; Akbulut, B.; Durak, I. TNF-α Inhibitors for the Management of Intractable Corneal Melt: Report of Three Cases and Review of the Literature. Eye Contact Lens 2021, 47, 372–377. [Google Scholar] [CrossRef] [PubMed]
- Kate, A.; Basu, S. Systemic Immunosuppression in Cornea and Ocular Surface Disorders: A Ready Reckoner for Ophthalmologists. Semin. Ophthalmol. 2022, 37, 330–344. [Google Scholar] [CrossRef] [PubMed]
- Srivastava, A.; Makarenkova, H.P. Innate Immunity and Biological Therapies for the Treatment of Sjögren’s Syndrome. Int. J. Mol. Sci. 2020, 21, 9172. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Sanders, F.W.B.; Huang, J.; Alió Del Barrio, J.L.; Hamada, S.; McAlinden, C. Amniotic membrane transplantation: Structural and biological properties, tissue preparation, application and clinical indications. Eye 2024, 38, 668–679. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Shafer, B.; Fuerst, N.M.; Massaro-Giordano, M.; Palladino, V.; Givnish, T.; Macchi, I.; Sulewski, M.E.; Orlin, S.E.; Bunya, V.Y. The use of self-retained, cryopreserved amniotic membrane for the treatment of Sjögren syndrome: A case series. Digit. J. Ophthalmol. 2019, 25, 21–25. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Chen, Z.; Lao, H.Y.; Liang, L. Update on the application of amniotic membrane in immune-related ocular surface diseases. Taiwan J. Ophthalmol. 2021, 11, 132–140. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Anchouche, S.; Harissi-Dagher, M.; Segal, L.; Racine, L.; Darvish-Zargar, M.; Robert, M.C. Cyanoacrylate Tissue Adhesive for the Treatment of Corneal Thinning and Perforations: A Multicenter Study. Cornea 2020, 39, 1371–1376. [Google Scholar] [CrossRef] [PubMed]
- Zemba, M.; Stamate, A.C.; Tataru, C.P.; Branisteanu, D.C.; Balta, F. Conjunctival flap surgery in the management of ocular surface disease (Review). Exp. Ther. Med. 2020, 20, 3412–3416. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Jan, R.L.; Ho, C.H.; Wang, J.J.; Tseng, S.H.; Chang, Y.S. Associations between Sjögren Syndrome, Sociodemographic Factors, Comorbid Conditions, and Scleritis in a Taiwanese Population-Based Study. J. Pers. Med. 2022, 12, 105. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Asfuroğlu, Y.; Karaca, E.E.; Orman, C.; Evren Kemer, Ö. Clinical and surgical outcomes of tectonic corneal patch grafts: Insights from a tertiary referral hospital. BMC Ophthalmol. 2024, 24, 516. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Choi, W.; Lee, S.S.; Park, Y.G.; Yoon, K.C. A case of necrotizing keratoscleritis in primary Sjogren’s syndrome. Korean J. Ophthalmol. 2011, 25, 275–277. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Gurnani, B.; Kaur, K. Therapeutic Keratoplasty. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2025. [Google Scholar] [PubMed]
- Artaechevarria Artieda, J.; Estébanez-Corrales, N.; Sánchez-Pernaute, O.; Alejandre-Alba, N. Peripheral Ulcerative Keratitis in a Patient with Bilateral Scleritis: Medical and Surgical Management. Case Rep. Ophthalmol. 2020, 11, 500–506. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Elhaddad, O.; Achiron, A.; Yahalomi, T.; Avadhanam, V.; Tole, D.; Darcy, K. Modified “Top-up sandwich” technique for the management of corneal perforations: A case report and a literature review. Am. J. Ophthalmol. Case Rep. 2022, 29, 101765. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Sharma, S.; Donthineni, P.R.; Iyer, G.; Chodosh, J.; de la Paz, M.F.; Maskati, Q.; Srinivasan, B.; Agarwal, S.; Basu, S.; Shanbhag, S.S. Keratoprosthesis in dry eye disease. Indian J. Ophthalmol. 2023, 71, 1154–1166. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Wang, B.; Chen, S.; Zheng, Q.; Li, Y.; Zhang, X.; Xuan, J.; Liu, Y.; Shi, G. Early diagnosis and treatment for Sjögren’s syndrome: Current challenges, redefined disease stages and future prospects. J. Autoimmun. 2021, 117, 102590. [Google Scholar] [CrossRef] [PubMed]
- Zeng, W.; Zhou, X.; Yu, S.; Liu, R.; Quek, C.W.N.; Yu, H.; Tay, R.Y.K.; Lin, X.; Feng, Y. The Future of Targeted Treatment of Primary Sjögren’s Syndrome: A Focus on Extra-Glandular Pathology. Int. J. Mol. Sci. 2022, 23, 14135. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Fox, R.I.; Fox, C.M.; McCoy, S.S. Emerging treatment for Sjögren’s disease: A review of recent phase II and III trials. Expert. Opin. Emerg. Drugs 2023, 28, 107–120. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Almulhim, A. Therapeutic Targets in the Management of Dry Eye Disease Associated with Sjögren’s Syndrome: An Updated Review of Current Insights and Future Perspectives. J. Clin. Med. 2024, 13, 1777. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Nepp, J.; Knoetzl, W.; Prinz, A.; Hoeller, S.; Prinz, M. Management of moderate-to-severe dry eye disease using chitosan-N-acetylcysteine (Lacrimera®) eye drops: A retrospective case series. Int. Ophthalmol. 2020, 40, 1547–1552. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Moawad, P.; Shamma, R.; Hassanein, D.; Ragab, G.; El Zawahry, O. Evaluation of the effect of topical tacrolimus 0.03% versus cyclosporine 0.05% in the treatment of dry eye secondary to Sjogren syndrome. Eur. J. Ophthalmol. 2022, 32, 673–679. [Google Scholar] [CrossRef] [PubMed]
- Moscovici, B.K.; Holzchuh, R.; Sakassegawa-Naves, F.E.; Hoshino-Ruiz, D.R.; Albers, M.B.; Santo, R.M.; Hida, R.Y. Treatment of Sjögren’s syndrome dry eye using 0.03% tacrolimus eye drop: Prospective double-blind randomized study. Contact Lens Anterior Eye 2015, 38, 373–378. [Google Scholar] [CrossRef] [PubMed]
- Tauber, J.; Davitt, W.F.; Bokosky, J.E.; Nichols, K.K.; Yerxa, B.R.; Schaberg, A.E.; LaVange, L.M.; Mills-Wilson, M.C.; Kellerman, D.J. Double-masked, placebo-controlled safety and efficacy trial of diquafosol tetrasodium (INS365) ophthalmic solution for the treatment of dry eye. Cornea 2004, 23, 784–792, Erratum in Cornea 2007, 26, 514. [Google Scholar] [CrossRef] [PubMed]
- Sun, X.; Liu, L.; Liu, C. Topical diquafosol versus hyaluronic acid for the treatment of dry eye disease: A meta-analysis of randomized controlled trials. Graefes Arch. Clin. Exp. Ophthalmol. 2023, 261, 3355–3367. [Google Scholar] [CrossRef] [PubMed]
- Yokoi, N.; Kato, H.; Kinoshita, S. The increase of aqueous tear volume by diquafosol sodium in dry-eye patients with Sjögren’s syndrome: A pilot study. Eye 2016, 30, 857–864. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Tauber, J.; Laurie, G.W.; Parsons, E.C.; Odrich, M.G.; Lacripep Study Group; Lacripep Study Group. Lacripep for the Treatment of Primary Sjögren-Associated Ocular Surface Disease: Results of the First-In-Human Study. Cornea 2023, 42, 847–857. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Wang, S.; Wang, M.; Liu, Y.; Hu, D.; Gu, L.; Fei, X.; Zhang, J. Effect of Rapamycin Microspheres in Sjögren Syndrome Dry Eye: Preparation and Outcomes. Ocul. Immunol. Inflamm. 2019, 27, 1357–1364. [Google Scholar] [CrossRef] [PubMed]
- Castrejón-Morales, C.Y.; Granados-Portillo, O.; Cruz-Bautista, I.; Ruiz-Quintero, N.; Manjarrez, I.; Lima, G.; Hernández-Ramírez, D.F.; Astudillo-Angel, M.; Llorente, L.; Hernández-Molina, G. Omega-3 and omega-6 fatty acids in primary Sjögren’s syndrome: Clinical meaning and association with inflammation. Clin. Exp. Rheumatol. 2020, 38 (Suppl. 126), 34–39. [Google Scholar] [PubMed]
- Kawahara, A. Treatment of Dry Eye Disease (DED) in Asia: Strategies for Short Tear Film Breakup Time-Type DED. Pharmaceutics 2023, 15, 2591. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Schaefer, L.; Trujillo-Vargas, C.M.; Midani, F.S.; Pflugfelder, S.C.; Britton, R.A.; de Paiva, C.S. Gut Microbiota From Sjögren syndrome Patients Causes Decreased T Regulatory Cells in the Lymphoid Organs and Desiccation-Induced Corneal Barrier Disruption in Mice. Front. Med. 2022, 9, 852918. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Abughanam, G.; Elkashty, O.A.; Liu, Y.; Bakkar, M.O.; Tran, S.D. Mesenchymal Stem Cells Extract (MSCsE)-Based Therapy Alleviates Xerostomia and Keratoconjunctivitis Sicca in Sjogren’s Syndrome-Like Disease. Int. J. Mol. Sci. 2019, 20, 4750, Erratum in Int. J. Mol. Sci. 2021, 22, 894. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Shin, S.; Yoon, S.G.; Kim, M.; Cheon, E.J.; Jeon, Y.; Lee, H.J.; Chung, S.H. The Effect of Mesenchymal Stem Cells on Dry Eye in Sjogren Syndrome Mouse Model. Int. J. Mol. Sci. 2023, 24, 1039. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Teos, L.Y.; Zheng, C.Y.; Liu, X.; Swaim, W.D.; Goldsmith, C.M.; Cotrim, A.P.; Baum, B.J.; Ambudkar, I.S. Adenovirus-mediated hAQP1 expression in irradiated mouse salivary glands causes recovery of saliva secretion by enhancing acinar cell volume decrease. Gene Ther. 2016, 23, 572–579. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Alevizos, I.; Zheng, C.; Cotrim, A.P.; Liu, S.; McCullagh, L.; Billings, M.E.; Goldsmith, C.M.; Tandon, M.; Helmerhorst, E.J.; Catalán, M.A.; et al. Late responses to adenoviral-mediated transfer of the aquaporin-1 gene for radiation-induced salivary hypofunction. Gene Ther. 2017, 24, 176–186. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Baeza, D.C.; Sanchez, V.; Goldhardt, R.; McCoy, S.S.; Tang, F.; Baer, A.; Fox, R.; Galor, A. The eye as a window to oral and pain symptoms in Sjögren’s disease. Br. J. Ophthalmol. 2025, bjo-2024-326748. [Google Scholar] [CrossRef] [PubMed]
Tissue/Organ | Diagnostic Method | Findings |
---|---|---|
Salivary and lacrimal glands | Histology [24,25,26] | Infiltration of lymphocytes CD4+ T helper cells (Th1 and Th17), CD8+ T cytotoxic cells, B lymphocytes, plasma cells, macrophages, fibroblasts, and dendritic and mast cells; reduced levels of T regulatory cells |
Destruction of the gland tubuloacinar architecture and atrophy in late stages | ||
High levels of pro-inflammatory cytokines, especially interferon (IFN)-γ and interleukin (IL)-17 | ||
Presence of autoantibodies, such as anti-SSA/Ro and anti-SSB/La | ||
Presence of Epstein–Barr virus antigens | ||
Ectopic lymphoid germinal center-like structures containing autoreactive B lymphocytes | ||
Tear film | Chemical and biochemical analyses [27] | Tear film hyperosmolarity |
Enhanced levels of proinflammatory cytokines (e.g., IL-1α, IL-1β, IL-6, IL-8, IL-10, IL-17, IL-33, TNF-α, and IFN-γ), matrix metalloproteinase (MMP-9), and defensins | ||
Over-expression of HLA-DR antigens | ||
Decreased levels of lysozyme, lactoferrin, and IgA | ||
Presence of autoantibodies, including anti-Ro/SSA, anti-La/SSB, ANA, and rheumatoid factor | ||
Decreased levels of epidermal growth factor secreted by the lacrimal gland | ||
Decreased levels of nerve growth factor secreted by the ocular surface nerves | ||
Decreased levels of a mucin and transforming growth factor (TGF)-β produced by the conjunctival goblet cells | ||
Upregulation of several proteins related to the oxidative stress, such as the APEX1 and S100 | ||
Microbiological analyses [28] | Tear film microbiome alterations, with predominant pro-inflammatory bacteria | |
Meibomian glands and eyelids | Slit lamp, keratography [29] | Anterior blepharitis |
Meibomian gland dysfunction, with reduced number of Meibomian glands, increased number of gland orifices occlusion, and metaplasia | ||
Decreased thickness and stability of the tear film lipid layer and increased tears evaporability | ||
Conjunctiva | In vivo confocal scanning laser microscopy, impression cytology [30,31] | Conjunctival epithelial cells decreased density and morphological alterations, with microcystic degeneration |
Significant reduction in goblet cell density and decreased tear film mucous layer (xerophthalmia); | ||
Infiltration of lymphocytes CD4+ T helper cells (Th1 and Th17), CD8+ T cytotoxic cells, B lymphocytes, plasma cells, macrophages, fibroblasts, and dendritic and mast cells; reduced levels of T regulatory cells | ||
Presence of autoantibodies, including anti-Ro/SSA, anti-La/SSB, ANA, and rheumatoid factor | ||
Increased levels of proinflammatory cytokine (e.g., IL-1, IL-6, and IFN-γ) | ||
Over-expression of human leukocytes antigen (HLA)-DR and HLA-DQ and CD40 antigens on both lymphocytes and conjunctival epithelial cells | ||
Over-expression of antigen intercellular adhesion molecule (ICAM)-1, which may induce apoptosis of the epithelial cells | ||
Conjunctival epithelial squamous metaplasia or keratinization (rare) | ||
Cicatrizing conjunctivitis (rare) | ||
Cornea | In vivo confocal scanning laser microscopy [32,33,34] | Morphological alteration of all epithelial layers |
Decreased cell density of superficial epithelium and outer and inner layers of the wing cells | ||
Reduced or increased basal epithelial cell density | ||
Squamous metaplasia or keratinization of the superficial epithelium (rare) | ||
Decreased or increased stromal keratocyte density | ||
Increased stromal dendritic cell density and activation | ||
Increased Bowman’s and stromal collagen degradation and stromal thinning | ||
Sub-basal nerve plexus morphological abnormalities (increased nerve reflectivity, width, and tortuosity) | ||
Sub-basal nerve plexus reduced density and length | ||
Ocular Response Analyzer [35] | Reduced corneal stiffness |
Maintaining good ocular hygiene |
Wearing protective glasses |
Avoiding smoking |
Avoiding dry or windy environments |
Limiting activities associated with reduced blink rate, such as reading or using computer |
Limiting medications that may decrease tears production, including anti-cholinergic drugs (antidepressant, anxiolytics, and anti-psychotics), muscarinic antagonists (e.g., tamsulosin and ipratropium), anti-histamines, opiates, anti-hypertensives (e.g., beta-blockers and ACE inhibitors), proton pump inhibitors (e.g., omeprazole), etc. |
Therapeutic Option | Active Components | Therapeutic Effects | Drawbacks and Side Effects | Efficacy in DED Patients (Level of Evidence) | Efficacy in SS Patients (Level of Evidence) | Guidelines Recommendation in SS-Related DED [42,43,44,55,56] |
---|---|---|---|---|---|---|
Artificial tear eye drops, lubricating gels, and ointments [59] | Hyaluronic acid, hydroxypropyl methylcellulose, carboxy methylcellulose, polyvinyl alcohol, carbopol, polyvinylpyrrolidone, polyethylene glycol, dextran, and polyacrylic acid liquid polyols | Reduction in friction between ocular surface and lids and increase in ocular surface regularity | Lack of tear components essential for ocular surface homeostasis (e.g., growth factors, vitamins, and immunoglobulins) and preservative-related toxicity [60] | Moderate to high [59,61] | Low [62] | First-line therapy in preservative-free formulations |
Autologous or homologous serum prepared as eyedrops [63] | Blood serum containing anti-infective agents (lysozyme, lactoferrin, and IgA), anti-inflammatory molecules (cytokines, interleukin receptor antagonists, and matrix metalloproteinase inhibitors), epitheliotrophic substances (fibronectin, vitamin A, epidermal growth factor, and transforming growth factor-β) [63]. | Ocular surface hydration and nutrition, stimulation of epithelial cells and nerves regeneration, and anti-inflammatory and anti-infective actions [63] | Risk of infections, input of proinflammatory cytokines in cases of severe SS systemic disease (prefer allogenic serum), and high preparation costs [63] | Low [64] | Low to moderate [65,66] | Suggested in cases of severe DED |
Non-steroidal anti-inflammatory drugs (NSAIDs) eyedrops [67] | Diclofenac, bromfenac, flurbiprofen, indomethacin, ketorolac, and nepafenac | Anti-inflammatory and analgesic effects | Corneal sensitivity decrease, epithelium healing delay [68], and association with corneal melting/perforations in general population [68] and SS patients [69,70] | Low to moderate in reducing ocular discomfort and some dry eye signs [67] | Low to moderate in reducing ocular discomfort and some dry eye signs [67] | Better to avoid |
Corticosteroids eyedrops [71] | Dexamethasone, methylprednisolone, fluorometholone, and hydrocortisone | Anti-inflammatory effects | Glaucoma, cataract, herpes simplex virus reactivation, epithelial healing delay, stromal collagen production inhibition, and increased risk of corneal/scleral perforation [71] | Low to moderate in improving DED symptoms; low in improving DED signs [71] | High in reducing ocular surface inflammation and improving DED symptoms; moderate in improving DED signs [72,73,74] | Suggested for short-term or pulse therapy (2–4 weeks) in patients who do not respond to other therapies |
Immunosuppressant agents eyedrops | Cyclosporine A [75] | Downregulation of proinflammatory cytokines, T-cells recruitment, and apoptosis | Burning sensation after instillation (60% of cases) | Controversial: high [76] or poor [77]; moderate in treatment of MGD [75] | Controversial evidence: high [76] or moderate [78] | Suggested in cases of moderate-to-severe DED and MGD |
Biologic agent eyedrops | Lifitegrast [79,80] | Integrin antagonist inhibiting antigen-presenting cells, CD4+ T cells activation and migration, and intracellular adhesion molecule-1 (ICAM-1) | High in improving signs and symptoms in non-SS DED patients [79] and in improving clinical DED signs in pSS animal models [80] | None | Approved by the FDA to reduce ocular surface inflammation in DED patients [79]. | |
Temporary or permanent punctal plugs [81] | Tear deflux reduction, tear meniscus increase | Increased risk of infections and accumulation of pro-inflammatory cytokines on the ocular surface | Controversial: high [62] or poor [81] | High [62] or moderate [82] | Suggested in cases of moderate-to-severe DED | |
Therapeutic contact lenses [83] | Ocular surface protection from eyelids and environment, increased permanence of artificial tears or drugs on the ocular surface, desiccation reduction, corneal healing promotion, and discomfort relieve | Mechanical damage of the surface, epithelial swelling, hypoxic, and infective and inflammatory complications | Moderate to high [83] | Limited [84] | Suggested in cases of moderate-to-severe DED or in presence of eyelids abnormalities |
Drug Class (Administration Route) | Active Component | Therapeutic Effects | Drawbacks and Side Effects | Efficacy in SS Patients (Level of Evidence) | Guidelines Recommendations [42,43,44,55,56] |
---|---|---|---|---|---|
Systemic corticosteroids (oral/intravenous) [74] | Oral prednisolone; intravenous methylprednisolone | Anti-inflammatory and immunosuppressant effects | Hypertension, hyperglycemia, weight gain, lipid profile alteration, osteoporosis and Cushingoid features, and increased risk of infections | Poor [57,58] | First-line systemic approach in cases of pSS patients with severe ocular sight-threatening conditions, such as corneal melting, PUK, necrotizing scleritis, or vasculitis |
Parasympathomimetic agonists (oral) | Pilocarpine (Cevimeline, not commercially available in Europe) | Parasympathomimetic muscarinic receptor agonist stimulating exocrine gland secretion | Nausea, excessive sweating, flushing, increased salivation, and urinary frequency | High improvement of mouth dryness; low to moderate in DED [93,94] | FDA-approved for managing moderate to severe; not recommended for dry eye |
Disease-modifying anti-rheumatic drugs (DMARDs)(oral) | Hydroxychloroquine | T-cell inhibition | Nausea, diarrhea, rash, hair changes, muscle weakness, anemia, and maculopathy | None in cases of non-SS DED [95] and SS-related DED [96] | First choice to treat fatigue and joint pain in SS patients |
Systemic immunosuppressive agents: Antimetabolites [103] | Methotrexate | Nucleic acid synthesis/cell proliferation inhibition | Gastrointestinal intolerance, elevated liver enzymes, stomatitis, dry eye, etc. | Some evidence in preventing ocular complications and treating PUK in RA patients | First choice to treat joint disease in SS patients; second choice (after systemic CSs) in sterile corneal ulcer/melt and PUK |
Azathioprine | Nucleic acid synthesis/cell proliferation inhibition | Gastrointestinal intolerance, bone marrow suppression, hepatotoxicity, etc. | None | Second choice (after systemic CSs) in PUK treatment | |
Mycophenolic acid | Nucleic acid synthesis/cell proliferation inhibition | Gastrointestinal intolerance and hepatotoxicity | Some evidence in reducing symptoms (not signs) of DED in pSS patients | None | |
Systemic immunosuppressive agents: T-cell inhibitors (oral) [103] | Cyclosporine A | T-cell inhibition | High blood pressure, inflamed gums, tremors, nausea, diarrhea, headache, etc. | None/very poor [57,58] | None/possible use in refractory SS-related DED |
Systemic immunosuppressive agents: Biologic agents (intravenous) [103,104] | Belimumab | Monoclonal antibody against the B-cell activating factor (BAFF) | Infusion reactions, increased risk of infections, etc. | Moderate in cases of systemic extraglandular manifestation [100] | High systemic disease activity, parotid enlargement, lymphadenopathies, and articular manifestation |
Infliximab | Tumor necrosis factor (TNF)-α inhibitor | Infusion reactions, increased risk of infections, etc. | None in SS-related DED [97]; moderate as adjuvant therapy in SS-related sterile corneal melting [102] and in PUK, scleritis, and vasculitis in RA patients [103] | Possible role in extraglandular ocular manifestation in SS and RA patients | |
Adalimumab | TNF-α inhibitor | Infusion reactions, increased risk of infections, etc. | Moderate in PUK, scleritis, and vasculitis in RA patients [103] | Possible role in case of extraglandular ocular manifestation in sSS patients | |
Abatacept | Selective inhibitor of T cells | Infusion reactions, increased risk of infections, etc. | None in pSS patients [101]; moderate in glandular and extraglandular ocular manifestations in sSS-RA patients [103]. | Possible role in case of DED and extraglandular ocular manifestation in sSS patients | |
Rituximab | Monoclonal antibody directed against the protein CD20 expressed on B cells | Infusion reactions, increased risk of infections, etc. | Poor to moderate in pSS-related DED [98]; none in pSS systemic disease activity and extraglandular systemic manifestation [99]; moderate for PUK and scleritis in RA patients [103] | Possible role in pSS-related DED and PUK and scleritis in RA patients | |
Tocilizumab | Interleukin(IL)-6 inhibitor | Infusion reactions, increased risk of infections, etc. | Promising results in PUK and necrotizing scleritis in systemic lupus erythematosus (SLE) and RA patients [103] | Possible role in PUK and scleritis in SLE and RA patients | |
Anakinra, tocilizumab, efalizumab, etanercept, and baminercept | Anti-IL-1 (anakinra), anti-IL6 (tocilizumab), anti-CD11 (efalizumab), anti-TNF-α (etanercept), and anti-lymphotoxin B receptor (baminercept) | Infusion reactions, increased risk of infections, etc. | None [99] | None | |
Antibiotics (oral) | Doxycycline | Antibiotic effect and MMPs inhibition with anti-collagenolytic effect | Headache, vomiting, diarrhea, etc. | None in pSS patients [57]; high to moderate in MGD [57] | Anterior blepharitis or MGD |
Human amniotic membrane (AM) transplantation | AM is a semi-transparent structure representing the deepest layer of the fetal membranes, and it has shown several therapeutic properties, including analgesic, re-epithelizing, anti-fibrotic, anti-microbial, anti-inflammatory, anti-angiogenic, and anti-adhesive effects. AM transplantation has been successfully used as temporary biologic bandage in a wide range of the ocular surface disorders, including chemical burns, persistent epithelial defects, ocular pemphigoid, bullous keratopathy, corneal ulcer, melting, impeding or full-thickness perforation, etc. [105]. The lack of immunogenicity avoids the need for immunosuppressive therapy. AM can be transplanted with or without sutures, using cyanoacrylate or fibrin glue and a therapeutic contact lens [105]. The benefits of its transplantation are temporary, with a median transplant duration of 3–4 weeks, and most patients complain of foreign body sensation and blurred vision during the AM implant [105]. Single or multilayer AM transplantation has shown to be beneficial in SS patients with severe KCS refractory to standard therapies and to promote healing of corneal ulceration/melting/perforations and peripheral ulcerative keratitis (PUK) [106,107]. |
Cyanoacrylate tissue adhesive (CTA) application | This represents a routinely approach to severe corneal/scleral thinning, melts, and impending or small uncomplicated corneal/scleral perforation (<2–3 mm in diameter) of different causes, such as infectious keratitis, trauma, neurotrophic or exposure keratopathy, and autoimmune diseases, including SS patients [108]. CTA application aims to re-establish the globe integrity (90% of success rate), to inhibit inflammatory cells blocking keratolysis worsening, and to contain infection due to its additional bacteriostatic effects [108]. Although generally used as a temporizing measure to delay the PK, CTA application can provide satisfactory corneal healing avoiding more invasive surgical interventions in 20–45% of cases [108]. |
Conjunctival flap graft | This may be used to restore ocular surface integrity in different chronic diseases, including severe DED and neurotrophic or neuroparalytic or bullous keratopathy [109]. |
Tarsorrhaphy | This involves the temporary or permanent joining of part or all of the upper and lower eyelids in order to partially or completely close the eye. It is indicated in cases of severe ocular surface damage in order to retain tears for a longer time and to reduce surface exposure [110]. Some cases of tarsorrhaphy have been described in SS patients [110]. |
Full-thickness or lamellar corneal/scleral patch grafting | This involves the placement of a donor corneal/scleral patch to reinforce weakened areas, with the aim to restore globe integrity (tectonic use) [111]. It is indicated in cases of corneal thinning, descemetocele, melting, and perforation that cannot be closed with tissue adhesives or AM transplantation or in eyes with acute inflammation, in which a traditional penetrating keratoplasty may be at high risk of rejection, aiming to postpone a larger refractive PK until systemic immunosuppressive therapy is effective [111]. Advantages over penetrating keratoplasty include lower risk of rejection, addition of tissue to the cornea/sclera that reduce the risk of recurrent perforation, and removal of necrotic epithelium and stroma that are sources of pro-inflammatory molecules that maintain the keratolysis/sclerolysis [111]. The most common indications for the corneal/scleral patch grafting are PUK, infectious keratitis, trauma, and limbal stem cell deficiency, with a reported rate of success, defined as globe integrity achievement, of 60–90% [111]. Several case reports of tectonic corneal/scleral patch grafting performed in SS patients with descemetocele, melting, PUK, or perforation have been published [111,112]. |
Tectonic lamellar or penetrating keratoplasty (PK) | This is indicated in cases of large central corneal perforations (>3 mm in diameter) of different causes (tectonic keratoplasty). Large-diameter grafts (9–9.5 mm) may be indicated to remove inflamed tissue but have a high risk of rejection due to the adjacent limbal vasculature [113]. Small-diameter tectonic grafts, ranging from 3 to 5.5 mm in diameter, are associate with poor visual outcome [113]. Keratoplasty requires a therapy with CSs, cyclosporine, and other systemic immunosuppressives [113]. Tectonic lamellar or penetrating keratoplasty have been described in SS patients [5,114,115]. |
Keratoprosthesis implantation | This involves the implant of a synthetic structure that replaces the central portion of a diseased cornea. It is indicated in cases with end-stage corneal blindness and where a routine keratoplasty ± limbal stem cells transplantation and systemic immunosuppressant therapy may have a high risk of failure [116]. Typical indications are severe dry eye with coexisting chronic cicatrizing conjunctivitis, adnexal disorders, and limbal stem cell deficiency such as chemical and thermal ocular burns, Stevens–Johnson syndrome, ocular mucous membrane pemphigoid, and SS [116]. Keratoprosthesis has the advantages of a relatively rapid visual recovery without requiring systemic immunosuppression [116]. A total of 18 eyes affected by corneal melting/perforation in SS patients have been described in the literature [116] |
Therapeutic Agent/Approach | Therapeutic Effects | Scientific Evidences |
---|---|---|
Umbilical cord serum eyedrops | Serum components intake | Promising results in reducing sign and symptoms of SS-related DED [65] |
Platelet-rich plasma eyedrops | Plasma components intake | Superior to autologous serum in improving dry eye symptoms in pSS patients [66] |
Acetylcysteine 5–10% eyedrops | Mucolytic effect and collagenase inhibition | Possible use in cases of keratitis filamentosa and corneal melt [121] |
Tacrolimus 0.1% eyedrops | T-cell immunosuppression | Effective in improving signs and symptoms of DED in SS patients [123], with efficacy comparable to topical cyclosporine A [122]; burning after instillation (80% of cases) [123] |
Diquafosol (3% eyedrops) | Antagonist of the P2Y2 purinergic receptor increasing tear and mucin production | Controversial results: no differences against placebo in a large RCT of DED patients including a small percentage of SS patients [124]; more effective than artificial tears in DED patients in a recent meta-analysis [125]; effective and safe to treat SS-related DED in small cohort [126] |
Regenerating agents (RGTA) eye drops: Poly-carboxymethyl glucose sulphate eyedrops | Drugs bioengineered to replace the heparin sulphate molecules of the corneal/scleral stroma, with reparative and regenerative effects | Effective as adjuvant therapy of corneal melting in pSS patients [86]. |
Lacripep eyedrops | Amino acid fragment of lacritin, a regulator of the ocular surface homeostasis | Encouraging results in improving signs and symptoms of DED in pSS [127] |
Rapamycin eyedrops (pSS animal models) | Immunosuppression | Improves tear secretion and reduces ocular surface damage in pSS animal model [128] |
Remibrutinib | Selective Bruton’s tyrosine kinase and B-cell inhibition | Effective in reducing systemic disease activity in SS patients (ocular manifestations not evaluated) [45,118,119,120] |
Dazodalibep | CD40 ligand antagonist suppressing stimulatory signals amongst T and B cells and antigen-presenting cells | Effective in reducing systemic disease activity in SS patients [45,118,119,120] |
Epratuzumab | Humanizedanti-CD22 monoclonal antibody | Effective in reducing systemic disease activity in pSS patients [45,118,119,120] |
Iguratimod | Anti-rheumatic agent | Effective in reducing systemic disease activity in SS patients [45,118,119,120] |
Ianalumab | Human monoclonal antibody able to deplete B cells | Effective in reducing systemic disease activity in pSS patients [45,118,119,120] |
Leflunomide combined with hydroxychloroquine | Type I IFN-associated proteins downregulation | Effective in reducing systemic disease activity in pSS patients [45,118,119,120] |
Iscalimab (CFZ533) | Anti-CD40 monoclonal antibody | Effective in reducing systemic disease activity in pSS patients [45,118,119,120] |
Sequential administration of Belimumab and Rituximab | Monoclonal antibody against B-cell activating factor (Belimumab); monoclonal antibody against CD20 protein of the B cells (Rituximab) | Effective in reducing systemic disease activity in pSS patients [45,118,119,120] |
Baricitinib | Janus kinase (JAK) inhibitor | Promising results in improving signs and symptoms of DED in pSS; effective in reducing systemic disease activity in pSS patients [45,118,119,120] |
Interleukin 2 | T-cell suppression when administered in low doses | Effective in reducing systemic diseases activity in SS patients [45,118,119,120] |
Interferon α-2b | Immunomodulation | Effective in improving signs and symptoms of xerostomia in SS patients at low dose; at high doses, used to treat hepatitis C or multiple myeloma, it has been associated with the development of dry eye and SS [45,118,119,120] |
RO5459072 | Cysteine protease cathepsin/T-cell inhibitor | Promising results in reducing systemic diseases activity in pSS patients [45,118,119,120] |
Diet supplements (omega-3 and omega-6 fatty acids, oral hyaluronic acid, astaxanthin, maquiberry extract, 1and bilberry extract) | Anti-oxidant, anti-inflammatory, anti-apoptotic, and immunomodulatory effects | Poor-to-moderate efficacy in reducing DED signs and/or symptoms [57,58,129,130]; no data on SS patients are available so far |
Microbiota transfer (pSS animal models) | Microbiota dysbiosis treatment | Germ-free mice colonized with human intestinal microbiota derived from pSS patients showed reduced CD4+T-regulatory cells and greater tendency of corneal barrier disruption when compared with germ-free mice colonized with intestinal microbiota of healthy subjects [131] |
Stem cells transplantation (pSS animal models) | Trophic, regenerative, and immunomodulatory effects (possible graft rejection) | Mesenchymal stem cells or allogenic multipotent hematopoietic stem cells transplantation on the ocular surface has shown promising results in increasing tear secretion, tear film stability, and epithelial and goblet cells regeneration and in reducing ocular surface flogosis [132]. The administration of mesenchymal stem cells by intra-lacrimal gland and subconjunctival injections has proven to inhibit epithelial cells autophagy, to reduce lymphocytic infiltration, and to decrease the objective signs of dry eye [133]. |
Gene transfer (animal models and human clinical trials) | Genetic integrity restitution | Adenoviral-mediated gene transfer or ultrasound assisted non-viral gene transfer of the AQP1 gene into damaged salivary glands are under investigation in several animal models [134] and in few clinical trials [135] and have shown promising preliminary results in increasing the parotid salivary flow. |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Salvetat, M.L.; Pellegrini, F.; D’Esposito, F.; Musa, M.; Tognetto, D.; Giglio, R.; Foti, R.; Gagliano, C.; Zeppieri, M. Emerging Trends and Management for Sjögren Syndrome-Related Dry Eye Corneal Alterations. Appl. Sci. 2025, 15, 5076. https://doi.org/10.3390/app15095076
Salvetat ML, Pellegrini F, D’Esposito F, Musa M, Tognetto D, Giglio R, Foti R, Gagliano C, Zeppieri M. Emerging Trends and Management for Sjögren Syndrome-Related Dry Eye Corneal Alterations. Applied Sciences. 2025; 15(9):5076. https://doi.org/10.3390/app15095076
Chicago/Turabian StyleSalvetat, Maria Letizia, Francesco Pellegrini, Fabiana D’Esposito, Mutali Musa, Daniele Tognetto, Rosa Giglio, Roberta Foti, Caterina Gagliano, and Marco Zeppieri. 2025. "Emerging Trends and Management for Sjögren Syndrome-Related Dry Eye Corneal Alterations" Applied Sciences 15, no. 9: 5076. https://doi.org/10.3390/app15095076
APA StyleSalvetat, M. L., Pellegrini, F., D’Esposito, F., Musa, M., Tognetto, D., Giglio, R., Foti, R., Gagliano, C., & Zeppieri, M. (2025). Emerging Trends and Management for Sjögren Syndrome-Related Dry Eye Corneal Alterations. Applied Sciences, 15(9), 5076. https://doi.org/10.3390/app15095076