• Therapeutic Cataract & Refractive
  • Lens Technology
  • Glasses
  • Ptosis
  • Comprehensive Eye Exams
  • AMD
  • COVID-19
  • DME
  • Ocular Surface Disease
  • Optic Relief
  • Geographic Atrophy
  • Cornea
  • Conjunctivitis
  • LASIK
  • Myopia
  • Presbyopia
  • Allergy
  • Nutrition
  • Pediatrics
  • Retina
  • Cataract
  • Contact Lenses
  • Lid and Lash
  • Dry Eye
  • Glaucoma
  • Refractive Surgery
  • Comanagement
  • Blepharitis
  • OCT
  • Patient Care
  • Diabetic Eye Disease
  • Technology

Utilizing the immune system to combat dry eye disease

Publication
Article
Optometry Times JournalJune digital edition 2024
Volume 16
Issue 06

Exploring strategies for effective management of a multifaceted condition.

Orange shield of blue backdrop with bacterial illustrations Image Credit: AdobeStock/Jennifer Toomey/MJH Life Sciences

Image Credit: AdobeStock/Jennifer Toomey/MJH Life Sciences

Dry eye disease (DED) is a chronic and often progressive condition that can be a source of frustration for patients and clinicians.1 Finding the right combination of treatments that effectively alleviate symptoms can be demanding, and the chronic nature of the condition typically requires ongoing management to maintain ocular health and comfort.

Managing DED presents unique challenges due to its diverse presentation and multifaceted etiology, which complicate both diagnosis and treatment strategies. With symptoms spanning mild to severe dryness, irritation, redness, light sensitivity, and vision loss, DED imposes a substantial burden on the quality of life of individuals affected.2 A study in 2017 estimated there were 16 million patients diagnosed with DED in the US alone—a number likely to be higher today, given the progressively aging population.3

Inflammation in DED

In recent years, a definition of DED has emerged that facilitates the discussion and management of DED.4 In a pivotal report published in 2017, the Tear Film and Ocular Surface Society Dry Eye Workshop II (TFOS DEWS II) group defined dry eye as “a multifactorial disease of the ocular surface characterized by a loss of homeostasis of the tear film, and accompanied by ocular symptoms, in which tear film instability and hyperosmolarity, ocular surface inflammation and damage, and neurosensory abnormalities play etiological roles.”5 This definition of DED underscores the crucial role that inflammation plays in the disease’s pathogenesis, highlighting the immune system as an appealing target for therapeutic intervention.

Inflammation in DED manifests as a persistent activation of the innate and adaptive immune systems, perpetuating ocular surface damage and disease progression.6 Although the exact mechanisms remain unclear, exposure to environmental stress, intrinsic dysfunction of immunoregulatory pathways, desensitization of corneal nerves, and changes in tear fluid composition are considered important drivers of ocular surface inflammation.7 These triggers promote the accumulation of immune cells at the ocular surface, where they release cytokines and induce epithelial damage.

Although inflammation is a physiological process that facilitates a return to homeostasis in normal circumstances, dysregulation of the inflammatory response can lead to a vicious cycle that perpetuates the disruption of the immune system and exacerbates damage to the ocular surface.7 In 2 clinical studies that evaluated large populations of subjects with DED, elevated levels of the inflammatory cytokine metalloproteinase-9 (MMP-9) was detected in more than 80% of subjects.8,9 To combat DED-associated inflammation, various immunosuppressive therapies have been explored to disrupt the inflammatory cycle.

Current treatments of DED

In recent years, the FDA has approved several novel pharmacological agents for DED, marking significant progress in the treatment landscape. These treatments encompass a broad spectrum of mechanisms, including a corticosteroid with enhanced safety (loteprednol etabonate [Eysuvis] in 2020),10 a nasal spray that stimulates basal tear production (varenicline solution [Tyrvaya] in 2021),11 a water-free cyclosporine 0.1% ophthalmic solution (Vevye in 2023) approved to treat signs and symptoms of DED,12 and an ophthalmic solution that reduces tear evaporation (perfluorohexyloctane [Miebo] in 2023).13 These additions complement existing treatments, including cyclosporine 0.05% (Restasis), cyclosporine 0.09% (Cequa), and lifitegrast (Xiidra).

Among the FDA-approved therapies for DED, cyclosporine, lifitegrast, and loteprednol etabonate are medications designed to target inflammation and modulate the immune system. As the first FDA-approved medication for the treatment of DED, cyclosporine is an effective immunosuppressive agent that inhibits IL-2 expression and T-cell activation.14 Lifitegrast, the second anti-inflammatory agent approved by the FDA, is a competitive antagonist of specific leukocyte integrins that are associated with T-cell activation, cytokine release, and ocular inflammation.15

In addition to cyclosporine and lifitegrast, clinicians routinely prescribe immunosuppressive corticosteroids to treat DED, despite heightened risks of ocular infection, glaucoma, and cataracts.16 Among corticosteroids, loteprednol etabonate is the preferred choice, as it is designed to rapidly metabolize into inactive metabolites after exerting its effect, thereby minimizing the risk of adverse effects.17

The approval of these therapies represents meaningful advances in our approach to DED management. However, a considerable number of patients continue to suffer from the condition despite extended pharmacological intervention. Some of these patients may exhibit advanced ocular surface damages that remain inadequately addressed by first-line therapies, whereas others may experience neurotrophic keratitis (NK)–related nerve desensitization that requires alternative interventions.

Treating advanced ocular surface damage in DED with amniotic membranes

In addition to the aforementioned therapies, comprehensive DED management also consists of artificial tears, proper eyelid hygiene, warm compresses, and nutritional supplements such as ω-3 and ω-6 fatty acids.18 For more advanced cases, treatments may consist of scleral lenses, autologous serum, punctal plugs, cenegermin (Oxervate), punctal plugs, and amniotic membranes (AMs). Despite the broad array of treatments available for DED, there is still a pressing need for innovative therapeutic approaches due to the highly diverse disease etiologies and challenges in disease management.

For patients with extensive epithelial damage or NK, cenegermin and AMs can be effective options that promote wound healing and reduce inflammation. Cenegermin is a recombinant form of human nerve growth factor (NGF) structurally identical to natural NGF.19,20 Studies have shown that NGF helps corneal epithelial cells and corneal nerves survive.21-23

AMs can play an integral role in repairing ocular surface damage and rehabilitation, as they have unique anti-inflammatory, antifibrotic, antiangiogenic, and prohealing properties for acute and chronic ocular surface disease.24 AMs are generally prepared for ophthalmic use via dehydration (various companies, such as IOP Ophthalmics and BioDOptix) or cryopreservation (BioTissue). Although there are several differences between dehydrated and cryopreserved AMs, these are due to the preservation process.

The dehydrated AM preservation process is performed with heat or chemicals and is indicated for wound covering but should not be used for active ocular infection. The cryopreservation technique retains the extracellular matrix components, such as heavy-chain hyaluronic acids, growth factors, fibronectin, and collagen, all of which promote anti-inflammatory effects and healing. Cryopreserved AM (CAM) consists of a highly biocompatible natural scaffold derived from the placenta that contains a rich source of regenerative stem cells, anti-inflammatory cytokines, and growth factors.25 For advanced ocular surface damage, either due to dry eye or NK, the appropriate AM should be considered for treatment.

The application of CAM on the corneal surface suppresses inflammation, reduces scarring or fibrosis, and accelerates epithelial wound healing and corneal nerve regeneration.26 These effects were demonstrated in the DREAM study, in which patients with severe refractory DED showed significant improvements on the ocular surface and reductions in DEWS scores after approximately 5 days of CAM therapy.27 Remarkably, 2 days of CAM therapy are sufficient to improve DEWS scores, corneal staining, visual symptoms, and ocular discomfort for up to 3 months.28

Mesenchymal stem cells in the treatments of DED

In recent years, mesenchymal stromal cells (MSCs) and MSC-derived exosomes have received considerable research attention in the treatment of DED due to their anti-inflammatory, tissue repair, and immune regulatory effects.29 MSCs are pluripotent cells isolated from donor tissues and can interact directly with target tissue through intercellular contact or indirectly via paracrine secretion mechanisms. When placed in an environment with proinflammatory factors, MSCs differentiate into an immunosuppressive phenotype that regulates various immune cells and exerts anti-inflammatory effects.

In the past decade, a number of preclinical studies in animal models of DED found adipose-derived MSCs reduced inflammation and neovascularization and improved epithelial cell marker expressions.29,30 In a clinical study of patients with severe DED secondary to Sjögren syndrome, an injection of allogenic adipose-derived MSCs into the lacrimal gland demonstrated promising clinical results.31 A single injection of MSCs significantly reduced Ocular Surface Disease Index (OSDI) scores and improved noninvasive tear breakup time.32

Although MSCs hold promise as a viable treatment option for DED, there is a considerable risk of allograft and cell rejection associated with MSC transplantation. In contrast, MSC-derived exosomes offer a cell-free therapy option that avoids these potential risks.

Preclinical studies of MSC-derived exosomes found that treatment decreased cytokine levels, promoted corneal epithelial repair, and increased tear secretion in a murine model of dry eye.33 In a group of eyes with refractory graft-vs-host disease, exosomes substantially reduced corneal staining, improved tear breakup time, increased tear secretion, and lowered OSDI scores.34 These effects were attributed to the presence of miR-204, a microRNA which was shown to reprogram proinflammatory macrophages toward an immunosuppressive state.

For MSC transplantation, additional research is warranted to minimize the risk of rejection. Meanwhile, MSC-derived exosomes face considerable difficulty in purification and characterization, which hinder the academic and clinical studies needed to bring these therapies to market.35

Targeting the immune system in practice

Recognizing the central role of inflammation in DED, many contemporary treatments aim to modulate the immune response, thereby disrupting the inflammatory cascade and facilitating epithelial healing. Although these anti-inflammatory therapies effectively alleviate symptoms and enhance the quality of life for a substantial number of patients, a significant portion still experience suboptimal outcomes, underscoring the need for innovative treatment approaches.

Many practitioners can be reluctant to manage DED due to the multifaceted etiologies and diverse sequelae. Yet, the expanding landscape of available therapeutics and their varied mechanisms present a compelling opportunity to effectively manage even the most refractory cases. With novel therapies such as varenicline, perfluorohexyloctane, and AM at our disposal, we are entering an exciting era in the treatment of ocular surface diseases, including DED and NK.

References:
  1. Mukamal R. Why is dry eye so difficult to treat? American Academy of Ophthalmology. January 19, 2024. Accessed March 3, 2024. https://www.aao.org/eye-health/glasses-contacts/fix-dry-eye-treatment-eyedrops
  2. Shiraishi A, Sakane Y. Assessment of dry eye symptoms: current trends and issues of dry eye questionnaires in Japan. Invest Ophthalmol Vis Sci. 2018;59(14):DES23-DES28. doi:10.1167/iovs.18-24570
  3. Farrand KF, Fridman M, Stillman IÖ, Schaumberg DA. Prevalence of diagnosed dry eye disease in the United States among adults aged 18 years and older. Am J Ophthalmol. 2017;182:90-98. doi:10.1016/j.ajo.2017.06.033
  4. Tsubota K, Pflugfelder SC, Liu Z, et al. Defining dry eye from a clinical perspective. Int J Mol Sci. 2020;21(23):9271. doi:10.3390/ijms21239271
  5. Craig JP, Nichols KK, Akpek EK, et al. TFOS DEWS II definition and classification report. Ocul Surf. 2017;15(3):276-283. doi:10.1016/j.jtos.2017.05.008
  6. Kaur RP, Gurnani B, Kaur K. Intricate insights into immune response in dry eye disease. Indian J Ophthalmol. 2023;71(4):1248-1255. doi:10.4103/IJO.IJO_481_23
  7. Rao SK, Mohan R, Gokhale N, Matalia H, Mehta P. Inflammation and dry eye disease—where are we? Int J Ophthalmol. 2022;15(5):820-827. doi:10.18240/ijo.2022.05.20
  8. Sambursky R, Davitt WF III, Latkany R, et al. Sensitivity and specificity of a point-of-care matrix metalloproteinase 9 immunoassay for diagnosing inflammation related to dry eye. JAMA Ophthalmol. 2013;131(1):24-28. doi:10.1001/jamaophthalmol.2013.561
  9. Sambursky R, Davitt WF III, Friedberg M, Tauber S. Prospective, multicenter, clinical evaluation of point-of-care matrix metalloproteinase-9 test for confirming dry eye disease. Cornea. 2014;33(8):812-818. doi:10.1097/ICO.0000000000000175
  10. Beckman K, Katz J, Majmudar P, Rostov A. Loteprednol etabonate for the treatment of dry eye disease. J Ocul Pharmacol Ther. 2020;36(7):497-511. doi:10.1089/jop.2020.0014
  11. Frampton JE. Varenicline solution nasal spray: a review in dry eye disease. Drugs. 2022;82(14):1481-1488. doi:10.1007/s40265-022-01782-4
  12. Hutton D. FDA approves Novaliq’s cyclosporine ophthalmic solution for treatment of dry eye disease. Ophthalmology Times. June 8, 2023. Accessed March 4, 2024. https://www.ophthalmologytimes.com/view/fda-approves-novaliq-s-cyclosporine-ophthalmic-solution-for-treatment-of-dry-eye-disease
  13. Sheppard JD, Evans DG, Protzko EE. A review of the first anti-evaporative prescription treatment for dry eye disease: perfluorohexyloctane ophthalmic solution. Am J Manag Care. Published online November 3, 2023. Accessed November 21, 2023. https://www.ajmc.com/view/a-review-of-the-first-anti-evaporative-prescription-treatment-for-dry-eye-disease-perfluorohexyloctane-ophthalmic-solution
  14. Russell G, Graveley R, Seid J, al-Humidan AK, Skjodt H. Mechanisms of action of cyclosporine and effects on connective tissues. Semin Arthritis Rheum. 1992;21(6 suppl 3):16-22. doi:10.1016/0049-0172(92)90009-3
  15. Abidi A, Shukla P, Ahmad A. Lifitegrast: a novel drug for treatment of dry eye disease. J Pharmacol Pharmacother. 2016;7(4):194-198. doi:10.4103/0976-500X.195920
  16. Lemp MA. Management of dry eye. Am J Manag Care. Published online April 15, 2008. Accessed March 4, 2024. https://www.ajmc.com/view/apr08-3142ps088-s101
  17. Venkateswaran N, Bian Y, Gupta PK. Practical guidance for the use of loteprednol etabonate ophthalmic suspension 0.25% in the management of dry eye disease. Clin Ophthalmol. 2022;16:349-355. doi:10.2147/OPTH.S323301
  18. Mondal H, Kim HJ, Mohanto N, Jee JP. A review on dry eye disease treatment: recent progress, diagnostics, and future perspectives. Pharmaceutics. 2023;15(3):990. doi:10.3390/pharmaceutics15030990
  19. Oxervate. Prescribing information. Dompé U.S. Inc.; 2019. Accessed June 4, 2024. https://oxervate.com/wp-content/uploads/2023/10/2023-OXERVATE-Prescribing-Information_10.30.23.pdf
  20. Bonini S, Rama P, Olzi D, Lambiase A. Neurotrophic keratitis. Eye (Lond). 2003;17(8):989-995. doi:10.1038/sj.eye.6700616
  21. Mastropasqua L, Massaro-Giordano G, Nubile M, Sacchetti M. Understanding the pathogenesis of neurotrophic keratitis: the role of corneal nerves. J Cell Physiol. 2017;232(4):717-724. doi:10.1002/jcp.25623
  22. Müller LJ, Marfurt CF, Kruse F, Tervo TM. Corneal nerves: structure, contents and function. Exp Eye Res. 2003;76(5):521-542. doi:10.1016/s0014-4835(03)00050-2
  23. Sacchetti M, Lambiase A. Neurotrophic factors and corneal nerve regeneration. Neural Regen Res. 2017;12(8):1220-1224. doi:10.4103/1673-5374.213534
  24. Tseng SC. HC-HA/PTX3 purified from amniotic membrane as novel regenerative matrix: insight into relationship between inflammation and regeneration. Invest Ophthalmol Vis Sci. 2016;57(5):ORSFh1-ORSFh8. doi:10.1167/iovs.15-17637
  25. Elkhenany H, El-Derby A, Abd Elkodous M, Salah RA, Lotfy A, El-Badri N. Applications of the amniotic membrane in tissue engineering and regeneration: the hundred-year challenge. Stem Cell Res Ther. 2022;13(1):8. doi:10.1186/s13287-021-02684-0
  26. John T, Tighe S, Sheha H, et al. Corneal nerve regeneration after self-retained cryopreserved amniotic membrane in dry eye disease. J Ophthalmol. 2017;2017:6404918. doi:10.1155/2017/6404918
  27. McDonald MB, Sheha H, Tighe S, et al. Treatment outcomes in the DRy Eye Amniotic Membrane (DREAM) study. Clin Ophthalmol. 2018;12:677-681. doi:10.2147/OPTH.S162203
  28. McDonald M, Janik SB, Bowden FW, et al. Association of treatment duration and clinical outcomes in dry eye treatment with sutureless cryopreserved amniotic membrane. Clin Ophthalmol. 2023;17:2697-2703. doi:10.2147/OPTH.S423040
  29. Jiang Y, Lin S, Gao Y. Mesenchymal stromal cell–based therapy for dry eye: current status and future perspectives. Cell Transplant. 2022;31:9636897221133818. doi:10.1177/09636897221133818
  30. Galindo S, Herreras JM, López-Paniagua M, et al. Therapeutic effect of human adipose tissue–derived mesenchymal stem cells in experimental corneal failure due to limbal stem cell niche damage. Stem Cells. 2017;35(10):2160-2174. doi:10.1002/stem.2672
  31. Møller-Hansen M, Larsen AC, Wiencke AK, et al. Allogeneic mesenchymal stem cell therapy for dry eye disease in patients with Sjögren’s syndrome: a randomized clinical trial. Ocul Surf. 2024;31:1-8. doi:10.1016/j.jtos.2023.11.007
  32. Møller-Hansen M. Mesenchymal stem cell therapy in aqueous deficient dry eye disease. Acta Ophthalmol. 2023;101 suppl 277:3-27. doi:10.1111/aos.15739
  33. Wang G, Li H, Long H, Gong X, Hu S, Gong C. Exosomes derived from mouse adipose–derived mesenchymal stem cells alleviate benzalkonium chloride–induced mouse dry eye model via inhibiting NLRP3 inflammasome. Ophthalmic Res. 2022;65(1):40-51. doi:10.1159/000519458
  34. Zhou T, He C, Lai P, et al. miR-204–containing exosomes ameliorate GVHD-associated dry eye disease. Sci Adv. 2022;8(2):eabj9617. doi:10.1126/sciadv.abj9617
  35. Wu KY, Ahmad H, Lin G, Carbonneau M, Tran SD. Mesenchymal stem cell–derived exosomes in ophthalmology: a comprehensive review. Pharmaceutics. 2023;15(4):1167. doi:10.3390/pharmaceutics15041167
Related Videos
Tracy Lynn Schroeder Swartz O.D., M.S., FAAO, overviews her talk on comanagement
Drs Erin Draper and Chris Cuk outline the important role optometrists have in spotting neurodegenerative disease in patients
Susan Gromacki, OD, MS, FAAO, FSLS
Jade Coats, OD, overviews a lecture on ocular pain and patient care
Jade Coats, OD, outlines two poster presentations she gave on a novel lipid-containing eye drop at the AOA Optometry's Meeting
Adam Alexander, OD, chats with Optometry Times about his AOA e-poster presentation on Miebo
Megan Cavet, PhD
Nazlee Zebardast, MD, MSc, overviews her ARVO 2024 presentations on glaucoma and polygenic risk scores
© 2024 MJH Life Sciences

All rights reserved.