Your new experience awaits. Try the new design now and help us make it even better

MINI REVIEW article

Front. Ophthalmol., 19 November 2025

Sec. Inflammatory Eye Diseases

Volume 5 - 2025 | https://doi.org/10.3389/fopht.2025.1700163

This article is part of the Research TopicMolecular Mechanisms and Pathophysiology of Dry Eye Disease and Meibomian Gland DysfunctionView all 6 articles

Cyclosporine a in the treatment of dry eye disease: a narrative review

Xiaoyan Bian&#x;Xiaoyan Bian1†Jun Ma&#x;Jun Ma2†Yunxia Liu&#x;Yunxia Liu3†Yuelan FengYuelan Feng3Zhiqiang Liu*Zhiqiang Liu4*Bozhou Zhang*Bozhou Zhang1*Baoyu Huang*Baoyu Huang5*
  • 1Department of Corneal Diseases, Baotou Chaoju Eye Hospital, Baotou, China
  • 2Department of Ophthalmology, Benxi Central Hospital, Benxi, Liaoning, China
  • 3Department of Ophthalmology, The First Affiliated Hospital of Baotou Medical College of Inner Mongolia University of Science and Technology, Baotou, China
  • 4Department of Ophthalmology, Yantai Huaxia Kang’ai Ophthalmic Hospital, Yantai, China
  • 5Department of Ophthalmology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China

Dry eye disease (DED) is a common chronic ocular surface disorder that significantly impacts quality of life. Its pathogenesis involves disruption of immune regulatory mechanisms and ocular surface inflammation, which mutually reinforce each other in a vicious cycle. Conventional treatments, such as artificial tears and meibomian gland care, alleviate symptoms but often fail to control underlying inflammation. Anti-inflammatory therapy is therefore crucial. Traditional agents like corticosteroids provide rapid relief but carry risks with long-term use. Cyclosporine A, an immunosuppressant, offers unique advantages by inhibiting T-cell activation, reducing pro-inflammatory cytokines, enhancing tear secretion, and restoring the ocular surface. Clinical and experimental studies have consistently demonstrated its efficacy and safety in improving tear production, relieving symptoms, repairing ocular surface structures, and slowing disease progression. This review summarizes the mechanisms, recent clinical evidence, and future perspectives of topical cyclosporine A in DED treatment, providing a reference for rational clinical use and novel therapeutic development.

Introduction

Dry eye disease (DED) is a common multifactorial disorder of the ocular surface. The pathophysiological mechanisms of DED include tear film instability and shortened tear break-up time (TBUT), tear hyperosmolarity, ocular surface inflammation and damage, and neurosensory abnormalities (1). In 2007, the International Dry Eye Workshop (DEWS I) first defined DED and identified inflammation as one of its major pathogenic mechanisms (2, 3).

Any insult to ocular surface function, such as reduced tear secretion, increased tear evaporation, or environmental irritation, can lead to tear film instability and elevated tear osmolarity, which in turn activate inflammatory cells on the ocular surface and trigger inflammatory cascades (4, 5). Consequently, controlling ocular surface inflammation and restoring microenvironmental stability are considered key strategies for DED management.

Current first-line treatments, including artificial tear supplementation, meibomian gland massage, warm compresses, and lifestyle modifications, can relieve symptoms to some extent but fail to fundamentally suppress the inflammatory process. Traditional anti-inflammatory agents, such as corticosteroids and nonsteroidal anti-inflammatory drugs, can rapidly improve symptoms but are unsuitable for long-term use due to adverse effects including intraocular pressure elevation, cataract formation, and corneal epithelial toxicity (6).

In recent years, emerging anti-inflammatory therapies such as lifitegrast, tacrolimus, and biologics targeting interleukin pathways have broadened the therapeutic landscape. Placing Cyclosporine A (CsA) within this context allows a clearer understanding of its advantages, such as long-term tolerability and immune-modulating specificity, as well as its limitations in onset speed and patient adherence (7). The latest TFOS DEWS II report further emphasized the role of inflammation in DED pathogenesis and recommended topical CsA as a long-term therapeutic option (6). Therefore, a comprehensive review of the mechanisms of action and clinical evidence of CsA in DED may provide valuable guidance for optimizing treatment strategies and improving patient outcomes.

This review aimed to provide an updated and integrative overview of the pharmacological mechanisms, formulation development, and clinical efficacy of topical CsA in the treatment of DED. Unlike earlier reviews that mainly summarized early clinical experiences, this work emphasizes recent evidence, comparative perspectives with other anti-inflammatory agents, and future research directions.

Literature search strategy

A comprehensive literature search was conducted in PubMed, Embase, and Web of Science from January 2000 to August 2025 using the keywords “dry eye disease,” “cyclosporine A,” “ocular surface,” “formulation,” and “clinical trial.” Randomized controlled trials, cohort studies, and relevant experimental investigations were included. Reviews, case reports, and conference abstracts were excluded. Reference lists of retrieved articles were manually screened to identify additional studies.

Mechanisms of CsA in the treatment of DED

CsA, a well-characterized immunosuppressant, inhibits T-cell activation, reduces inflammatory mediator release, and improves lacrimal gland function, thereby effectively alleviating DED signs and symptoms with favorable long-term safety. In addition, CsA is a cyclic, non-ribosomal neutral peptide with a molecular weight of 1,202 g/mol, marked hydrophobicity (LogP = 1.4–3.0), and extremely low aqueous solubility (6.6×106 μg/ml, temperature-dependent), with only 0.008 mg/ml solubility in water at room temperature. These physicochemical properties limit its ocular delivery and bioavailability in topical formulations (Figure 1) (8).

Figure 1
Diagram illustrating the mechanism of action for topical CsA. CsA interacts with cyclophilin A and affects lacrimal gland epithelial cells and conjunctival goblet cells. It inhibits calcineurin, reducing IL-2 production, leading to decreased T-cell activation and cytokine release, ultimately enhancing tear film stability and goblet cell density while reducing ocular surface inflammation.

Figure 1. Mechanism of action of topical cyclosporine A (CsA) in dry eye disease. Topically applied CsA penetrates the ocular surface and binds to cyclophilin A within lacrimal gland epithelial cells and T lymphocytes. The CsA–cyclophilin A complex inhibits calcineurin activity, leading to decreased interleukin-2 (IL-2) transcription and suppression of T-cell activation and cytokine release. This cascade reduces ocular surface inflammation, stabilizes the tear film, and increases conjunctival goblet cell density, thereby improving the ocular surface microenvironment in dry eye disease.

At the cellular level, CsA penetrates the cytoplasm of T cells and binds to cyclophilin A, thereby inhibiting calcineurin phosphatase activity and blocking the transcription of IL-2 and other T cell activation-related genes (8). Additionally, CsA can bind to cyclophilin D to inhibit apoptosis or programmed cell death (8, 9), and suppress T cell–mediated inflammatory responses by blocking p38 and JNK signaling pathways. In the inflammatory milieu of DED, levels of inflammatory mediators such as IL-1, IL-2, IL-6, tumor necrosis factor-α (TNF-α), and matrix metalloproteinases (MMPs) are markedly elevated in tears and conjunctival epithelial cells. These mediators activate dendritic cells and recruit T lymphocytes, which further release cytokines, impair lacrimal functional unit function, damage corneal and conjunctival goblet cells, reduce mucin secretion, and amplify apoptosis and inflammation, thus perpetuating the vicious inflammatory cycle. CsA can effectively disrupt this cycle, reduce ocular surface T lymphocyte infiltration and inflammatory cytokine production, improve corneal and conjunctival epithelial apoptosis (10, 11), and significantly increase goblet cell density and function (12).

CsA also restores tear secretion suppressed by inflammation, a process that may not only depend on inflammation resolution but also involve stimulation of muscarinic and sensory nerves to release neurotransmitters (13, 14). In DED, MMP concentrations and activity in tears are elevated (15), leading to degradation of basement membrane components and tight junction proteins such as ZO-1 and occluding (16, 17), thereby weakening the corneal barrier. CsA treatment reduces MMP levels, helping repair and maintain ocular surface barrier integrity. In terms of mucin metabolism, CsA increases the proportion of secretory goblet cells and enhances intracellular mucin content, enabling faster mucin synthesis and accumulation compared to untreated conditions (18).

Furthermore, in vivo confocal microscopy studies have shown that CsA can reverse DED-induced activation of corneal Langerhans cells, loss of subbasal corneal nerve fiber density, and increased nerve tortuosity, while restoring corneal sensitivity (19, 20). Recovery of corneal nerve function can reduce the release of neuropeptides associated with neurogenic inflammation and even strengthen reflex arcs to stimulate blinking and tear secretion, thereby lowering tear osmolarity and creating favorable conditions for corneal nerve regeneration (20). Therefore, even patients with mild ocular surface inflammation may benefit from the multifaceted mechanisms of CsA.

Clinical formulations of CsA

CsA has demonstrated definite efficacy and low toxicity in the treatment of DED. However, its large molecular weight, hydrophobicity, and low permeability limit the optimization of drug concentration, stability, and administration routes. To improve water solubility and ocular permeability, the concept of “nanoemulsion” was proposed, which consists of oil, surfactant, and co-surfactant, and is more easily absorbed than traditional emulsions (Table 1).

Table 1
www.frontiersin.org

Table 1. Comparison of commercially available topical cyclosporine A formulations for dry eye disease.

In 2002, the U.S. FDA approved Restasis® (Allergan, Irvine, CA, USA), a 0.05% CsA anionic oil-in-water nanoemulsion for the treatment of inflammation-related tear secretion reduction, with a particle size of approximately 159.3 nm. Moreover, its oil-based high viscosity and poor stability often cause blurred vision, burning sensation, and conjunctival hyperemia, leading to poor tolerability and low bioavailability (21, 22). In China, Zirun® (Xingqi Pharmaceutical Co., Ltd., Shenyang, China) (0.05% CsA clear nano-microemulsion, particle size about 24.2 nm) can show efficacy within 3 months, with blurred vision being rarely reported as an adverse event (23).

Ikervis® (Santen Pharmaceutical Co., Ltd., Osaka, Japan), a 0.1% CsA cationic oil-in-water nanoemulsion, was approved by the European Union in 2015. By electrostatically interacting with the mucin layer, it prolongs retention time on the ocular surface, thereby improving bioavailability. Additionally, cationic emulsions themselves may promote tear secretion and modulate inflammatory cytokine expression (24, 25). In 2018, the FDA approved OTX-101 (Cequa™; Sun Pharmaceutical Industries Ltd., Mumbai, India), a 0.09% CsA nanomicellar aqueous solution, with a particle size of 10–80 nm. It can easily diffuse through scleral pores, with CsA concentrations in the conjunctiva and sclera being 3.60-fold and 3.44-fold higher than those with Restasis, respectively. It shows faster onset and higher stability, though ocular irritation and potential drug leakage remain possible adverse effects (26, 27).

CyclASol® (Novaliq GmbH, Heidelberg, Germany), a 0.1% CsA semifluorinated alkane anhydrous solution contains no preservatives or surfactants, has high solubilizing capacity for hydrophobic drugs and good biocompatibility, and can reduce blurred vision as well as the risk of iatrogenic DED, with good tolerability (2830). To overcome the rapid clearance of eye drops by tears, in situ gel systems can prolong drug release and improve bioavailability, though they have low oxygen permeability and may cause blurred vision (31).

CsA-loaded contact lenses (CL), including Brij-52 microemulsion, porous carrier, and amyloid nanofilm types, enhance bioavailability by 50%–82% compared with Restasis. They also prolong drug action and promote corneal healing without affecting optical performance (32, 33). Other novel formulations, including low-concentration CsA–trehalose nanoemulsion, lipid nanocapsules, cationic hyaluronic acid-coated formulations, and cyclodextrin-based oil-free CsA eye drops, have all demonstrated potential in improving bioavailability and tolerability (3436).

Clinical application of CsA

Currently, CsA alleviates inflammation and symptoms in moderate-to-severe DED and Sjögren’s syndrome (SS) (3742), enhancing goblet cell density, mucin secretion, and tear film stability with efficacy comparable to corticosteroids (13, 43).

Multiple clinical studies have confirmed that CsA eye drops effectively improve ocular surface disease index (OSDI), tear film BUT, Schirmer’s test results, corneal fluorescein staining, and ocular discomfort (3739). For example, the foundational 0.05% CsA emulsion (Restasis®) has been shown to improve Schirmer scores, BUT, and corneal staining, although it may require more than 6 months to demonstrate full efficacy. A multicenter study involving 240 Chinese patients showed that 0.05% CsA eye drops (II) (produced by Shenyang Xingqi Eye Medicine Co., Ltd., specification: 0.4ml: 0.2mg) administered twice daily for moderate-to-severe DED led to better total ocular scores than control after one week, with noticeable improvement within 1–2 months, and significantly superior outcomes compared with the control group after 3 months (23, 44). In a dose-ranging, randomized trial, the therapeutic effect of CsA generally emerges after several weeks of use, and study have shown that 0.05% and 0.1% concentrations achieve similar efficacy, with higher concentrations offering no obvious advantage (39).

In a retrospective analysis involving patients with meibomian gland dysfunction (MGD), CsA can reduce inflammation, improve eyelid margin scurf, telangiectasia, and meibum stasis, and enhance BUT and tear secretion (45). A Korean prospective, randomized, single-blinded, controlled clinical study demonstrated that 0.05% CsA nanoemulsion combined with warm compresses was more effective than sodium hyaluronate eye drops with warm compresses in treating MGD (46).

In cases of iatrogenic DED, such as post-LASIK and post-cataract surgery, CsA also shows good efficacy. One year after LASIK, CsA treatment restored Schirmer’s test and BUT to preoperative levels, improving DED indicators by approximately 30%, significantly better than the control group (47). In a prospective, open-label, randomized, controlled study among post-cataract surgery patients, three months of 0.05% CsA significantly improved Schirmer’s test results and BUT, and reduced dry eye symptom scores (48). A comparison between 3% diquafosol sodium and 0.05% CsA for post-cataract surgery DED found that the former improved tear secretion faster, while the latter was more effective in improving visual aberrations and required less frequent dosing (48).

In graft-versus-host disease (GVHD)-related DED, topical CsA improves dry eye symptoms, corneal sensitivity, tear evaporation rate, BUT, corneal staining, and conjunctival inflammatory cell infiltration (49). Preconditioning with topical CsA before hematopoietic stem cell transplantation (HSCT) can reduce postoperative dry eye severity, suggesting a preventive potential (50). An in vivo confocal microscopy study indicated that long-term use of preservative-containing medications in glaucoma patients often causes ocular surface damage; long-term CsA use can alleviate dry eye symptoms and prevent fibrosis (51).

Numerous randomized controlled trials worldwide have further confirmed the significant efficacy of CsA in moderate-to-severe DED. A multicenter study in China showed that combining 0.05% CsA with artificial tears for 8–12 weeks significantly improved OSDI score, corneal staining, tear secretion, and BUT, with an effective rate exceeding 70% (52). Phase III clinical trials abroad demonstrated that 0.1% CsA treatment for 6 months significantly improved ocular symptoms and reduced conjunctival HLA-DR expression (53). The latest approved 0.09% CsA nano-formulation has shown greater improvement in tear secretion and ocular surface symptoms in clinical studies (26). Clinical trials with different CsA concentrations indicate that 0.05% CsA is most effective in improving ocular symptoms, with higher concentrations showing no significant additional benefit (22).

Adverse effects of CsA

The incidence and severity of adverse effects from topical CsA are largely concentration dependent. Low-concentration formulations show minimal systemic absorption and an excellent safety profile. The most frequently reported reactions are mild-to-moderate ocular irritation symptoms, such as stinging, burning, blurred vision, and conjunctival hyperemia, with no serious local or systemic adverse events observed (5456). These irritation symptoms are often attributed not only to the active CsA molecule but also significantly to the formulation vehicle, such as the castor oil-based emulsion in traditional preparations, which can cause poor tolerability. In a clinical trial involving 1039 DED patients, CsA was well tolerated (57).

Reported adverse event rates vary across concentrations and formulations, with approximately 25.3% in the 0.05% group and 29% in the 0.1% group, predominantly involving mild-to-moderate eye pain or burning (58, 59). Differences in formulation may influence the incidence of adverse reactions.

Differences in formulation may influence the incidence of adverse reactions. Differences in vehicle composition may contribute to this variation. Transient stinging or burning upon instillation remains the main cause of treatment discontinuation in a minority of patients, highlighting the need for improved formulation tolerability. Nanoemulsion and cationic emulsion vehicles have been shown to lessen discomfort and enhance adherence compared with traditional oil-based preparations (6062). Long-term follow-up has not identified any CsA-related systemic toxicity or sight-threatening adverse events, supporting its suitability for chronic use (63). Furthermore, combining CsA with artificial tears or short-term corticosteroids may reduce early irritation and improve compliance (6466).

Regarding CL-based delivery systems, potential risks include bacterial resistance, ocular toxicity from hydrogel–drug interactions, and transient systemic exposure due to initial burst release (67). Emerging multifunctional biosensor–CL systems aim to reduce systemic toxicity, but remain in the research stage.

Prospective

Currently, due to CsA’s strong hydrophobicity, the bioavailability of topical formulations remains low, limiting its clinical performance. Premature discontinuation often leads to symptom relapse, underscoring the need to define standardized treatment duration and tapering strategies. In addition, combination therapy has gained attention as a means to enhance efficacy and tolerability. Short-term topical corticosteroids can accelerate symptom relief in the early phase of CsA therapy by suppressing acute inflammation, while subsequent maintenance with CsA supports long-term control (68). Artificial tears remain indispensable as a complementary therapy to improve lubrication and patient comfort, and their concurrent use does not interfere with CsA’s pharmacodynamics (69). Secretagogues such as diquafosol or rebamipide may also provide synergistic benefits by improving mucin and aqueous tear secretion. Future research should clarify the optimal timing, sequence, and duration of these combination strategies to maximize therapeutic benefit while minimizing ocular irritation (70). Future investigations should also emphasize head-to-head comparisons between CsA and other immunomodulators (lifitegrast, tacrolimus), as well as the long-term impact of CsA on neurosensory recovery and ocular microbiome balance.

Future DED treatment strategies should be more individualized, taking into account different etiologies, disease courses, and severities, and formulating multi-level, long-term treatment plans. Incorporating biomarkers such as tear cytokine levels, HLA-DR expression, and patient genotypes into therapeutic decision-making could enable precision medicine approaches for CsA therapy, ultimately improving both efficacy and adherence in chronic DED management.

Conclusion

In conclusion, with deeper understanding of DED pathophysiology and advances in delivery technologies, CsA’s application prospects in dry eye treatment are promising. Through continuous optimization of treatment regimens and innovative formulations, safer, more effective, and more personalized dry eye management can be achieved, ultimately improving patients` quality of life. Further comparative and mechanistic research is needed to determine CsA’s position among current and emerging therapies, guiding evidence-based, individualized treatment approaches.

Author contributions

XB: Funding acquisition, Writing – review & editing, Writing – original draft, Resources, Software, Formal Analysis, Visualization, Project administration, Supervision, Methodology, Validation, Conceptualization, Investigation, Data curation. JM: Writing – review & editing, Conceptualization, Writing – original draft. YL: Visualization, Project administration, Data curation, Methodology, Formal Analysis, Validation, Conceptualization, Supervision, Writing – original draft, Software, Resources, Writing – review & editing, Investigation, Funding acquisition. YF: Conceptualization, Data curation, Visualization, Writing – original draft. ZL: Supervision, Conceptualization, Project administration, Investigation, Validation, Writing – review & editing, Methodology, Data curation, Funding acquisition, Software, Writing – original draft, Formal Analysis, Resources, Visualization. BZ: Formal Analysis, Visualization, Data curation, Resources, Writing – review & editing, Project administration, Validation, Funding acquisition, Software, Methodology, Supervision, Conceptualization, Investigation, Writing – original draft. BH: Formal Analysis, Project administration, Data curation, Methodology, Visualization, Conceptualization, Validation, Funding acquisition, Software, Writing – original draft, Writing – review & editing, Supervision, Resources, Investigation.

Funding

The author(s) declare financial support was received for the research and/or publication of this article. This study was supported by the Baotou Municipal Health and Wellness Science and Technology Project (No. wsjkkj2022100). The funding body had no role in the design of the study, data collection, analysis, interpretation of data, or in writing the manuscript.

Acknowledgments

We would acknowledge the use of artificial intelligence tools that were used for language improvement.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Generative AI statement

The author(s) declare that no Generative AI was used in the creation of this manuscript.

Any alternative text (alt text) provided alongside figures in this article has been generated by Frontiers with the support of artificial intelligence and reasonable efforts have been made to ensure accuracy, including review by the authors wherever possible. If you identify any issues, please contact us.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Golden MI, Meyer JJ, Zeppieri M, and Patel BC. Dry eye syndrome. Treasure Island (FL: StatPearls (2025). ineligible companies. Disclosure: Jay Meyer declares no relevant financial relationships with ineligible companies. Disclosure: Marco Zeppieri declares no relevant financial relationships with ineligible companies. Disclosure: Bhupendra Patel declares no relevant financial relationships with ineligible companies.

Google Scholar

2. The definition and classification of dry eye disease: report of the Definition and Classification Subcommittee of the International Dry Eye WorkShop (2007). Ocul Surf. (2007) 5:75–92. doi: 10.1016/s1542-0124(12)70081-2

PubMed Abstract | Crossref Full Text | Google Scholar

3. Craig JP, Nichols KK, Akpek EK, Caffery B, Dua HS, Joo CK, et al. TFOS DEWS II definition and classification report. Ocul Surf. (2017) 15:276–83. doi: 10.1016/j.jtos.2017.05.008

PubMed Abstract | Crossref Full Text | Google Scholar

4. Cukurova F, Coco G, Lixi F, and Giannaccare G. Current and emerging pharmacological treatment options for patients with dry eye disease. Expert Rev Clin Pharmacol. (2025) 18:485–501. doi: 10.1080/17512433.2025.2543380

PubMed Abstract | Crossref Full Text | Google Scholar

5. Guo S, Kang J, Yan K, Li J, Wang R, Qin D, et al. Prostaglandin F2alpha exacerbated dry eye by promoting lacrimal gland fibrosis progression through the activation of the RhoA/ROCKs signaling pathway. Ocul Surf. (2025) 38:155–69. doi: 10.1016/j.jtos.2025.07.004

PubMed Abstract | Crossref Full Text | Google Scholar

6. Craig JP, Nelson JD, Azar DT, Belmonte C, Bron AJ, Chauhan SK, et al. TFOS DEWS II report executive summary. Ocul Surf. (2017) 15:802–12. doi: 10.1016/j.jtos.2017.08.003

PubMed Abstract | Crossref Full Text | Google Scholar

7. Charreau B, Coupel S, Boulday G, and Soulillou JP. Cyclosporine inhibits class II major histocompatibility antigen presentation by xenogeneic endothelial cells to human T lymphocytes by altering expression of the class II transcriptional activator gene. Transplantation. (2000) 70:354–61. doi: 10.1097/00007890-200007270-00021

PubMed Abstract | Crossref Full Text | Google Scholar

8. Shen Y, Yu Y, Chaurasiya B, Li X, Xu Y, Webster TJ, et al. Stability, safety, and transcorneal mechanistic studies of ophthalmic lyophilized cyclosporine-loaded polymeric micelles. Int J Nanomedi. (2018) 13:8281–96. doi: 10.2147/IJN.S173691

PubMed Abstract | Crossref Full Text | Google Scholar

9. Mandal A, Gote V, Pal D, Ogundele A, and Mitra AK. Ocular pharmacokinetics of a topical ophthalmic nanomicellar solution of cyclosporine (Cequa(R)) for dry eye disease. Pharm Res. (2019) 36:36. doi: 10.1007/s11095-018-2556-5

PubMed Abstract | Crossref Full Text | Google Scholar

10. Jin R, Li Y, Li L, Kim J, Yoon HJ, and Yoon KC. Comparative analysis of 0.1% cyclosporin A cationic emulsion and 0.05% cyclosporin A emulsion in murine dry eye cases with different severities. Exp Ther Med. (2021) 22:1363. doi: 10.3892/etm.2021.10797

PubMed Abstract | Crossref Full Text | Google Scholar

11. Strong B, Farley W, Stern ME, and Pflugfelder SC. Topical cyclosporine inhibits conjunctival epithelial apoptosis in experimental murine keratoconjunctivitis sicca. Cornea. (2005) 24:80–5. doi: 10.1097/01.ico.0000133994.22392.47

PubMed Abstract | Crossref Full Text | Google Scholar

12. Moon I, Kang HG, Yeo A, Noh H, Kim HC, Song JS, et al. Comparison of ocular surface mucin expression after topical ophthalmic drug administration in dry eye-induced mouse model. J Ocul Pharmacol Ther. (2018) 34:612–20. doi: 10.1089/jop.2018.0005

PubMed Abstract | Crossref Full Text | Google Scholar

13. de Paiva CS, Pflugfelder SC, Ng SM, and Akpek EK. Topical cyclosporine A therapy for dry eye syndrome. Cochrane Database Syst Rev. (2019) 9:CD010051. doi: 10.1002/14651858.CD010051.pub2

PubMed Abstract | Crossref Full Text | Google Scholar

14. Yoshida A, Fujihara T, and Nakata K. Cyclosporin A increases tear fluid secretion via release of sensory neurotransmitters and muscarinic pathway in mice. Exp Eye Res. (1999) 68:541–6. doi: 10.1006/exer.1998.0619

PubMed Abstract | Crossref Full Text | Google Scholar

15. Mullick R, Annavajjhala S, Thakur P, Mohapatra A, Shetty R, and D'Souza S. Efficacy of topical cyclosporine 0.05% and osmoprotective lubricating eye drops in treating dry eye disease and inflammation. Indian J Ophthalmol. (2021) 69:3473–7. doi: 10.4103/ijo.IJO_3822_20

PubMed Abstract | Crossref Full Text | Google Scholar

16. Pflugfelder SC, Farley W, Luo L, Chen LZ, de Paiva CS, Olmos LC, et al. Matrix metalloproteinase-9 knockout confers resistance to corneal epithelial barrier disruption in experimental dry eye. Am J Pathol. (2005) 166:61–71. doi: 10.1016/S0002-9440(10)62232-8

PubMed Abstract | Crossref Full Text | Google Scholar

17. De Paiva CS, Corrales RM, Villarreal AL, Farley WJ, Li DQ, Stern ME, et al. Corticosteroid and doxycycline suppress MMP-9 and inflammatory cytokine expression, MAPK activation in the corneal epithelium in experimental dry eye. Exp Eye Res. (2006) 83:526–35. doi: 10.1016/j.exer.2006.02.004

PubMed Abstract | Crossref Full Text | Google Scholar

18. Phillips TE, McHugh J, and Moore CP. Cyclosporine has a direct effect on the differentiation of a mucin-secreting cell line. J Cell Physiol. (2000) 184:400–8. doi: 10.1002/1097-4652(200009)184:3<400::AID-JCP15>3.0.CO;2-W

PubMed Abstract | Crossref Full Text | Google Scholar

19. Iaccheri B, Torroni G, Cagini C, Fiore T, Cerquaglia A, Lupidi M, et al. Corneal confocal scanning laser microscopy in patients with dry eye disease treated with topical cyclosporine. Eye (Lond). (2017) 31:788–94. doi: 10.1038/eye.2017.3

PubMed Abstract | Crossref Full Text | Google Scholar

20. Levy O, Labbe A, Borderie V, Hamiche T, Dupas B, Laroche L, et al. Increased corneal sub-basal nerve density in patients with Sjogren syndrome treated with topical cyclosporine A. Clin Exp Ophthalmol. (2017) 45:455–63. doi: 10.1111/ceo.12898

PubMed Abstract | Crossref Full Text | Google Scholar

21. Periman LM, Mah FS, and Karpecki PM. A review of the mechanism of action of cyclosporine A: the role of cyclosporine A in dry eye disease and recent formulation developments. Clin Ophthalmol. (2020) 14:4187–200. doi: 10.2147/OPTH.S279051

PubMed Abstract | Crossref Full Text | Google Scholar

22. Coursey TG, Wassel RA, Quiambao AB, and Farjo RA. Once-daily cyclosporine-A-miDROPS for treatment of dry eye disease. Transl Vis Sci Technol. (2018) 7:24. doi: 10.1167/tvst.7.5.24

PubMed Abstract | Crossref Full Text | Google Scholar

23. Chen D, Zhang S, Bian A, Hong J, Deng Y, Zhang M, et al. Efficacy and safety of 0.05% cyclosporine ophthalmic emulsion in treatment of Chinese patients with moderate to severe dry eye disease: A 12-week, multicenter, randomized, double-masked, placebo-controlled phase III clinical study. Med (Baltimore). (2019) 98:e16710. doi: 10.1097/MD.0000000000016710

PubMed Abstract | Crossref Full Text | Google Scholar

24. Daull P, Lallemand F, and Garrigue JS. Benefits of cetalkonium chloride cationic oil-in-water nanoemulsions for topical ophthalmic drug delivery. J Pharm Pharmacol. (2014) 66:531–41. doi: 10.1111/jphp.12075

PubMed Abstract | Crossref Full Text | Google Scholar

25. Terreni E, Chetoni P, Burgalassi S, Tampucci S, Zucchetti E, Chipala E, et al. A hybrid ocular delivery system of cyclosporine-A comprising nanomicelle-laden polymeric inserts with improved efficacy and tolerability. Biomater Sci. (2021) 9:8235–48. doi: 10.1039/D1BM01453F

PubMed Abstract | Crossref Full Text | Google Scholar

26. Lu Y, Yue Z, Xie J, Wang W, Zhu H, Zhang E, et al. Micelles with ultralow critical micelle concentration as carriers for drug delivery. Nat BioMed Eng. (2018) 2:318–25. doi: 10.1038/s41551-018-0234-x

PubMed Abstract | Crossref Full Text | Google Scholar

27. Lubtow MM, Marciniak H, Schmiedel A, Roos M, Lambert C, and Luxenhofer R. Ultra-high to ultra-low drug-loaded micelles: probing host-guest interactions by fluorescence spectroscopy. Chemistry. (2019) 25:12601–10. doi: 10.1002/chem.201902619

PubMed Abstract | Crossref Full Text | Google Scholar

28. Chaudhari P, Birangal S, Mavlankar N, Pal A, Mallela LS, Roy S, et al. Oil-free eye drops containing Cyclosporine A/cyclodextrin/PVA supramolecular complex as a treatment modality for dry eye disease. Carbohydr Polym. (2022) 297:120007. doi: 10.1016/j.carbpol.2022.120007

PubMed Abstract | Crossref Full Text | Google Scholar

29. Sheppard JD, Wirta DL, McLaurin E, Boehmer BE, Ciolino JB, Meides AS, et al. A water-free 0.1% Cyclosporine A solution for treatment of dry eye disease: results of the randomized phase 2B/3 ESSENCE study. Cornea. (2021) 40:1290–7. doi: 10.1097/ICO.0000000000002633

PubMed Abstract | Crossref Full Text | Google Scholar

30. Akpek EK, Wirta DL, Downing JE, Tauber J, Sheppard JD, Ciolino JB, et al. Efficacy and safety of a water-free topical cyclosporine, 0.1%, solution for the treatment of moderate to severe dry eye disease: the ESSENCE-2 randomized clinical trial. JAMA Ophthalmol. (2023) 141:459–66. doi: 10.1001/jamaophthalmol.2023.0709

PubMed Abstract | Crossref Full Text | Google Scholar

31. Lynch CR, Kondiah PPD, Choonara YE, du Toit LC, Ally N, and Pillay V. Hydrogel biomaterials for application in ocular drug delivery. Front Bioeng Biotechnol. (2020) 8:228. doi: 10.3389/fbioe.2020.00228

PubMed Abstract | Crossref Full Text | Google Scholar

32. Zhao L, Song J, Du Y, Ren C, Guo B, and Bi H. Therapeutic applications of contact lens-based drug delivery systems in ophthalmic diseases. Drug Deliv. (2023) 30:2219419. doi: 10.1080/10717544.2023.2219419

PubMed Abstract | Crossref Full Text | Google Scholar

33. Qin R, Guo Y, Ren H, Liu Y, Su H, Chu X, et al. Instant adhesion of amyloid-like nanofilms with wet surfaces. ACS Cent Sci. (2022) 8:705–17. doi: 10.1021/acscentsci.2c00151

PubMed Abstract | Crossref Full Text | Google Scholar

34. Zhang A, Sun R, Ran M, Deng Y, Ge Y, Zhu Y, et al. A novel eyes topical drug delivery system: csA-LNC for the treatment of DED. Pharm Res. (2020) 37:146. doi: 10.1007/s11095-020-02872-2

PubMed Abstract | Crossref Full Text | Google Scholar

35. Bang SP, Yeon CY, Adhikari N, Neupane S, Kim H, Lee DC, et al. Cyclosporine A eyedrops with self-nanoemulsifying drug delivery systems have improved physicochemical properties and efficacy against dry eye disease in a murine dry eye model. PloS One. (2019) 14:e0224805. doi: 10.1371/journal.pone.0224805

PubMed Abstract | Crossref Full Text | Google Scholar

36. Bhujel B, Oh SH, Kim CM, Yoon YJ, Chung HS, Ye EA, et al. Current advances in regenerative strategies for dry eye diseases: A comprehensive review. Bioeng (Basel). (2023) 11. doi: 10.3390/bioengineering11010039

PubMed Abstract | Crossref Full Text | Google Scholar

37. Toyos M, Gupta PK, Mitchell B, and Karpecki P. The effect of OTX-101 on tear production in patients with severe tear-deficient dry eye disease: A pooled analysis of phase 2b/3 and phase 3 studies. Curr Eye Res. (2022) 47:220–4. doi: 10.1080/02713683.2021.1966477

PubMed Abstract | Crossref Full Text | Google Scholar

38. Kim J, Moon TK, Yoon HJ, Ji YS, and Yoon KC. Efficacy of switching from cyclosporine A 0.05% Anionic emulsion to cyclosporine A 0.1% Cationic emulsion in patients with dry eye associated with sjogren's syndrome. J Ocul Pharmacol Ther. (2021) 37:472–8. doi: 10.1089/jop.2020.0146

PubMed Abstract | Crossref Full Text | Google Scholar

39. Stevenson D, Tauber J, and Reis BL. Efficacy and safety of cyclosporin A ophthalmic emulsion in the treatment of moderate-to-severe dry eye disease: a dose-ranging, randomized trial. Cyclosporin A Phase 2 Study Group Ophthalmol. (2000) 107:967–74. doi: 10.1016/s0161-6420(00)00035-x

PubMed Abstract | Crossref Full Text | Google Scholar

40. Lighthizer N, Schwertz BK, Chester T, Longo R, Riley P, Mody L, et al. TearCare system versus cyclosporine ophthalmic emulsion for the treatment of moderate-to-severe meibomian gland disease associated dry eye disease in the United States: a cost-utility analysis. Expert Rev Pharmacoecon Outcomes Res. (2025), 1–9. doi: 10.1080/14737167.2025.2537850

PubMed Abstract | Crossref Full Text | Google Scholar

41. Salam AA, Sen S, Lomi N, Gupta N, Vanathi M, and Tandon R. Comparative evaluation of aqueous solution and oil emulsion formulations of 0.05% cyclosporine eye drops in dry eye disease - A randomized clinical trial. Indian J Ophthalmol. (2025) 73:577–81. doi: 10.4103/IJO.IJO_1850_24

PubMed Abstract | Crossref Full Text | Google Scholar

42. Yoon SH, Kim EC, You IC, Choi CY, Kim JY, Song JS, et al. Clinical efficacy of 0.1% cyclosporine A in dry eye patients with inadequate responses to 0.05% cyclosporine A: a switching, prospective, open-label, multicenter study. BMC Ophthalmol. (2025) 25:37. doi: 10.1186/s12886-025-03862-x

PubMed Abstract | Crossref Full Text | Google Scholar

43. Yoon HJ, Jin R, Yoon HS, Choi JS, Kim Y, Pan SH, et al. Bacillus-derived manganese superoxide dismutase relieves ocular-surface inflammation and damage by reducing oxidative stress and apoptosis in dry eye. Invest Ophthalmol Vis Sci. (2023) 64:30. doi: 10.1167/iovs.64.12.30

PubMed Abstract | Crossref Full Text | Google Scholar

44. Chen M, Gong L, Sun X, Xie H, Zhang Y, Zou L, et al. A comparison of cyclosporine 0.05% ophthalmic emulsion versus vehicle in Chinese patients with moderate to severe dry eye disease: an eight-week, multicenter, randomized, double-blind, parallel-group trial. J Ocul Pharmacol Ther. (2010) 26:361–6. doi: 10.1089/jop.2009.0145

PubMed Abstract | Crossref Full Text | Google Scholar

45. Kim HY, Lee JE, Oh HN, Song JW, Han SY, and Lee JS. Clinical efficacy of combined topical 0.05% cyclosporine A and 0.1% sodium hyaluronate in the dry eyes with meibomian gland dysfunction. Int J Ophthalmol. (2018) 11:593–600. doi: 10.18240/ijo.2018.04.09

PubMed Abstract | Crossref Full Text | Google Scholar

46. Rhim JW, Eom Y, Yoon EG, Park SY, Choi Y, Song JS, et al. Efficacy of a 0.05% cyclosporine a topical nanoemulsion in dry eyes with obstructive meibomian gland dysfunction. Jpn J Ophthalmol. (2022) 66:254–63. doi: 10.1007/s10384-022-00906-3

PubMed Abstract | Crossref Full Text | Google Scholar

47. Namdev V, Kaur M, Sharma VK, Mulay A, Raj R, and Titiyal JS. Current paradigms in refractive surgery. Med J Armed Forces India. (2024) 80:497–504. doi: 10.1016/j.mjafi.2024.08.003

PubMed Abstract | Crossref Full Text | Google Scholar

48. Lee JH, Song IS, Kim KL, and Yoon SY. Effectiveness and Optical Quality of Topical 3.0% Diquafosol versus 0.05% Cyclosporine A in Dry Eye Patients following Cataract Surgery. J Ophthalmol. (2016) 2016:8150757. doi: 10.1155/2016/8150757

PubMed Abstract | Crossref Full Text | Google Scholar

49. Lelli GJ Jr., Musch DC, Gupta A, Farjo QA, Nairus TM, and Mian SI. Ophthalmic cyclosporine use in ocular GVHD. Cornea. (2006) 25:635–8. doi: 10.1097/01.ico.0000208818.47861.1d

PubMed Abstract | Crossref Full Text | Google Scholar

50. Chun YH, Beak JU, Kim HS, and Na KS. Topical cyclosporine pretreatment of ocular surface in allogeneic hematopoietic stem cell transplant recipients. J Ocul Pharmacol Ther. (2018) 34:628–32. doi: 10.1089/jop.2018.0006

PubMed Abstract | Crossref Full Text | Google Scholar

51. Zhu W, Kong X, Xu J, and Sun X. Effects of long-term antiglaucoma eye drops on conjunctival structures: an in vivo confocal microscopy study. J Ophthalmol. (2015) 2015:165475. doi: 10.1155/2015/165475

PubMed Abstract | Crossref Full Text | Google Scholar

52. Yu Y, Chen D, Li Y, Yang W, Tu J, and Shen Y. Improving the topical ocular pharmacokinetics of lyophilized cyclosporine A-loaded micelles: formulation, in vitro and in vivo studies. Drug Deliv. (2018) 25:888–99. doi: 10.1080/10717544.2018.1458923

PubMed Abstract | Crossref Full Text | Google Scholar

53. Nagai N and Otake H. Novel drug delivery systems for the management of dry eye. Adv Drug Delivery Rev. (2022) 191:114582. doi: 10.1016/j.addr.2022.114582

PubMed Abstract | Crossref Full Text | Google Scholar

54. Lei HL, Ku WC, Sun MH, Chen KJ, Lai JY, and Sun CC. Cyclosporine a eye drop-induced elongated eyelashes: a case report. Case Rep Ophthalmol. (2011) 2:398–400. doi: 10.1159/000335281

PubMed Abstract | Crossref Full Text | Google Scholar

55. Rhee MK and Mah FS. Clinical utility of cyclosporine (CsA) ophthalmic emulsion 0.05% for symptomatic relief in people with chronic dry eye: a review of the literature. Clin Ophthalmol. (2017) 11:1157–66. doi: 10.2147/OPTH.S113437

PubMed Abstract | Crossref Full Text | Google Scholar

56. Ramos-Casals M, Tzioufas AG, Stone JH, Siso A, and Bosch X. Treatment of primary Sjogren syndrome: a systematic review. JAMA. (2010) 304:452–60. doi: 10.1001/jama.2010.1014

PubMed Abstract | Crossref Full Text | Google Scholar

57. Brito-Zeron P, Retamozo S, Kostov B, Baldini C, Bootsma H, De Vita S, et al. Efficacy and safety of topical and systemic medications: a systematic literature review informing the EULAR recommendations for the management of Sjogren's syndrome. RMD Open. (2019) 5:e001064. doi: 10.1136/rmdopen-2019-001064

PubMed Abstract | Crossref Full Text | Google Scholar

58. Daull P, Lallemand F, Philips B, Lambert G, Buggage R, and Garrigue JS. Distribution of cyclosporine A in ocular tissues after topical administration of cyclosporine A cationic emulsions to pigmented rabbits. Cornea. (2013) 32:345–54. doi: 10.1097/ICO.0b013e31825e83f4

PubMed Abstract | Crossref Full Text | Google Scholar

59. Weiss SL and Kramer WG. Ocular distribution of cyclosporine following topical administration of OTX-101 in New Zealand white rabbits. J Ocul Pharmacol Ther. (2019) 35:395–402. doi: 10.1089/jop.2018.0106

PubMed Abstract | Crossref Full Text | Google Scholar

60. Preeti, Sambhakar S, Malik R, Bhatia S, Al Harrasi A, Rani C, et al. Nanoemulsion: an emerging novel technology for improving the bioavailability of drugs. Scientifica (Cairo). (2023) 2023:6640103. doi: 10.1155/2023/6640103

PubMed Abstract | Crossref Full Text | Google Scholar

61. He H, Li J, Meng H, He Y, Pei X, and Zhang Z. Recent advances in cationic nanoemulsions for drug delivery: preparation, properties, and applications. Curr Pharm Des. (2025) 31:2267–81. doi: 10.2174/0113816128357859250121120216

PubMed Abstract | Crossref Full Text | Google Scholar

62. Kishore A, Mahor AK, Singh NK, Singh PP, Rathore P, and Bansal KK. Oil-in-water nanoemulsions for glaucoma treatment: An insight into the latest trends. Bioimpacts. (2025) 15:30224. doi: 10.34172/bi.2024.30224

PubMed Abstract | Crossref Full Text | Google Scholar

63. Straathof K, Anoop P, Allwood Z, Silva J, Nikolajeva O, Chiesa R, et al. Long-term outcome following cyclosporine-related neurotoxicity in paediatric allogeneic haematopoietic stem cell transplantation. Bone Marrow Transplant. (2017) 52:159–62. doi: 10.1038/bmt.2016.232

PubMed Abstract | Crossref Full Text | Google Scholar

64. Singla S, Sarkar L, and Joshi M. Comparison of topical cyclosporine alone and topical loteprednol with cyclosporine in moderate dry eye in Indian population: A prospective study. Taiwan J Ophthalmol. (2019) 9:173–8. doi: 10.4103/tjo.tjo_15_18

PubMed Abstract | Crossref Full Text | Google Scholar

65. Lollett IV and Galor A. Dry eye syndrome: developments and lifitegrast in perspective. Clin Ophthalmol. (2018) 12:125–39. doi: 10.2147/OPTH.S126668

PubMed Abstract | Crossref Full Text | Google Scholar

66. Liu SH, Saldanha IJ, Abraham AG, Rittiphairoj T, Hauswirth S, Gregory D, et al. Topical corticosteroids for dry eye. Cochrane Database Syst Rev. (2022) 10:CD015070. doi: 10.1002/14651858.CD015070.pub2

PubMed Abstract | Crossref Full Text | Google Scholar

67. ElShaer A, Ghatora B, Mustafa S, and Alany RG. Contact lenses as drug reservoirs & delivery systems: the successes & challenges. Ther Deliv. (2014) 5:1085–100. doi: 10.4155/tde.14.73

PubMed Abstract | Crossref Full Text | Google Scholar

68. Ryu KJ, Kim S, Kim MK, Paik HJ, and Kim DH. Short-term therapeutic effects of topical corticosteroids on refractory dry eye disease: clinical usefulness of matrix metalloproteinase 9 testing as a response prediction marker. Clin Ophthalmol. (2021) 15:759–67. doi: 10.2147/OPTH.S300047

PubMed Abstract | Crossref Full Text | Google Scholar

69. Pucker AD, Ng SM, and Nichols JJ. Over the counter (OTC) artificial tear drops for dry eye syndrome. Cochrane Database Syst Rev. (2016) 2:CD009729. doi: 10.1002/14651858.CD009729.pub2

PubMed Abstract | Crossref Full Text | Google Scholar

70. Li K, Guo B, Gu J, Ta N, Gu J, Yu H, et al. Emerging advances in drug delivery systems (DDSs) for optimizing cancer complications. Mater Today Bio. (2025) 30:101375. doi: 10.1016/j.mtbio.2024.101375

PubMed Abstract | Crossref Full Text | Google Scholar

Keywords: dry eye disease, cyclosporine A, inflammation, immune modulation, review

Citation: Bian X, Ma J, Liu Y, Feng Y, Liu Z, Zhang B and Huang B (2025) Cyclosporine a in the treatment of dry eye disease: a narrative review. Front. Ophthalmol. 5:1700163. doi: 10.3389/fopht.2025.1700163

Received: 13 September 2025; Accepted: 05 November 2025; Revised: 05 November 2025;
Published: 19 November 2025.

Edited by:

Driss Zoukhri, Tufts University, United States

Reviewed by:

Shree Rath, All India Institute of Medical Sciences Bhubaneswar, India
Laura Valencia Nieto, Complutense University of Madrid, Spain

Copyright © 2025 Bian, Ma, Liu, Feng, Liu, Zhang and Huang. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Zhiqiang Liu, NTMyNjQxNDJAcXEuY29t; Bozhou Zhang, emhhbmdib3pob3VAY2hhb2p1ZXllLmNvbQ==; Baoyu Huang, aHVhbmdiYW95dTIwMjVAMTYzLmNvbQ==

These authors have contributed equally to this work

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.