296
Views
1
CrossRef citations to date
0
Altmetric
REVIEW

Glaucoma and Dry Eye Disease: Opportunity to Assess and Treat

, & ORCID Icon
Pages 3063-3076 | Received 11 May 2023, Accepted 07 Sep 2023, Published online: 17 Oct 2023

Abstract

Dry eye disease (DED) has been found to occur at a higher prevalence in individuals with glaucoma than in individuals without glaucoma. The relationship between glaucoma and DED may be, in part, a result of glaucoma therapy. Greater number of antiglaucoma medications used and greater number of antiglaucoma eyedrops instilled per day have been associated with ocular surface disease in patients with glaucoma. Use of antiglaucoma medication has also been associated with higher levels of ocular surface inflammatory markers and ocular surface alterations. There is evidence to suggest that antiglaucoma medications with preservatives and, to some extent, antiglaucoma medication formulations without preservatives may contribute to ocular surface signs and symptoms. Trabeculectomy for glaucoma has also been associated with ocular surface signs related to DED; however, there may be benefits of trabeculectomy and other procedures for glaucoma due to reduced use of antiglaucoma medications. Patients with glaucoma with ocular surface disease have been found to have greater ocular surface symptoms, poorer vision-related quality of life, and poorer antiglaucoma medication adherence compared with patients with glaucoma without ocular surface disease. Because of the potential negative impact of DED on patients with glaucoma, patients with glaucoma may benefit from evaluation for DED. Management of DED in patients with glaucoma may include modifications to antiglaucoma medications and use of treatments for DED.

Introduction

Glaucoma is characterized by progressive deterioration of the optic nerve and vision loss.Citation1 It is the leading cause of irreversible blindness worldwide.Citation1 Deterioration of the optic nerve in glaucoma is often associated with elevated intraocular pressure (IOP), and treatments for glaucoma aim to slow the progression of the disease by lowering IOP.Citation1 Available treatments include topical medications, laser, or incisional surgeries.Citation1 Early diagnosis, ongoing treatment, and treatment adherence are critical to slowing disease progression and preventing blindness.Citation1–4

Dry eye disease (DED) is an ocular surface disease, characterized by hyperosmolarity and tear film instability, that can cause discomfort and fluctuating vision and can potentially lead to ocular surface damage.Citation5 DED can be detected with tear-film and ocular-surface measures, including tear film breakup time, osmolarity, matrix metalloproteinase-9 (MMP-9) testing, and ocular surface staining.Citation6 Symptoms of DED negatively affect quality of life, and the associated visual disturbances from DED may affect activities such as reading and driving.Citation7 DED occurs at an increased prevalence in patients with glaucoma and has frequently been associated with the use of antiglaucoma medications.Citation8–13 Because of the negative impact of DED on the ocular surface and quality of life, it is important to understand the potential reasons for the association between glaucoma and DED and ways to manage DED in patients with glaucoma. This review describes the relationship between glaucoma and DED, the impact of DED on patients with glaucoma, and management strategies for DED in patients with glaucoma.

Methods

A PubMed database search was conducted to review the literature on DED and glaucoma. Search terms included combinations of “dry eye”, “ocular surface”, “tear film”, and “meibomian gland” with “glaucoma” and combinations of “glaucoma drainage implant”, “laser”, “minimally invasive glaucoma surgery”, and “surgery” with “glaucoma” and “dry eye” or “ocular surface”. Articles were included if the subject matter was relevant to discussing the association between DED and glaucoma, with studies in humans given priority. Review articles, editorials, articles not in English, economic analyses, and case studies were excluded.

Relationship Between Glaucoma and Dry Eye Disease

The estimated prevalence of ocular surface signs and/or symptoms in individuals with glaucoma varies across studies.Citation10,Citation14–18 In a large register study, 52.6% (10,338/19,665) of those with glaucoma had a diagnosis of DED.Citation10 However, given the wide range of estimates of DED in the general population (5% to 50%),Citation7 these estimates are difficult to interpret. Studies that have included a control group have found that DED occurs at a higher rate in individuals with glaucoma than in individuals without glaucoma. In a national survey, DED was identified in 16.5% (104/629) of those with glaucoma compared with 5.6% (386/6934) of those without glaucoma.Citation8 Conversely, among individuals with DED, glaucoma was identified in 21.2% (104/490), compared with 7.4% (525/7073) of those without DED.Citation8 Although the prevalence of both DED and glaucoma increases with age, age does not fully explain the relationship between these diseases, because a higher prevalence of ocular surface signs and symptoms has been found among individuals with glaucoma when compared with age-matched controls and when age has been included as a covariate in the analysis.Citation19 Similar to findings in the general population,Citation7 DED among individuals with glaucoma has been found to be more prevalent at older agesCitation10,Citation11 and more prevalent in women (56.9%) than in men (45.7%).Citation10

When ocular surface symptoms and signs have been examined, individuals with glaucoma have been found to have higher rates of abnormalities compared with controls.Citation19–23 Among 211 patients with glaucoma, 41.7% (88/211) were found to have ocular surface symptoms, as measured by the Ocular Surface Disease Index (OSDI), compared with 25.5% (13/51) of controls without glaucoma.Citation19 Findings with the OSDI may need to be interpreted with caution because the vision-related component of the OSDI may be influenced by vision changes related to glaucoma as opposed to ocular surface disease.Citation23 Ocular surface staining has also been found to be more prevalent in individuals with glaucoma.Citation19,Citation20,Citation24–26 Ghosh et al found corneal staining (grade 2–3) in 51.3% (154/300) of those with glaucoma and 17.0% (17/100) of controls.Citation25 Tear breakup time has also been found to be shorter in individuals with glaucoma.Citation20,Citation26,Citation27 Furthermore, total tear breakup area has been found to be greater, and the growth rate of the dry area has been found to be steeper in those with glaucoma.Citation27 Tear meniscus height and Schirmer scores have also been found to be lower in eyes with glaucoma compared with controls.Citation24,Citation28 Compared with fellow eyes, eyes with glaucoma have been found to have greater tear film osmolarity, greater conjunctival hyperemia, and greater eyelid margin abnormality.Citation28 In addition to ocular surface signs, individuals with glaucoma have been found to have greater meibomian gland loss and poorer meibum scores than controls.Citation29–32 The tear film lipid layer—which plays an important role in tear film stability—is made up of meibum.Citation33,Citation34 Thus, meibomian gland dysfunction may lead to deficiencies in the tear film lipid layer and increased evaporation of the tear film.Citation34,Citation35

Relationship Between Glaucoma Therapy and Dry Eye Disease

Topical Antiglaucoma Medication and Dry Eye Disease

The relationship between glaucoma and DED may result from, at least in part, the use of topical antiglaucoma medications. Notably, Kuppens et al found that basal tear turnover in individuals with untreated primary open-angle glaucoma was lower than that of individuals without glaucoma, suggesting that glaucoma may in itself lead to DED;Citation36 however, the role of antiglaucoma medications in DED has been suggested by several studies.Citation9–13 Antiglaucoma medications consist of an active component and an excipient, which may include a preservative. Several types of active components exist, including prostaglandin analogues, β-blockers, ⍺-2 adrenergic agonists, carbonic anhydrase inhibitors, pilocarpine, rho-associated kinase inhibitors, and combination medications.Citation9,Citation37,Citation38

Several studies suggest that a greater use of antiglaucoma medications is associated with an increased risk of DED. In a large study of patients with glaucoma (N=10,325), the odds of DED were found to increase with the number of medications used (OR=1.23 with 2 medications; OR=1.63 with 3 medications; OR=2.60 with 4 medications).Citation9 Similarly, in a separate study, ocular surface disease prevalence was found to increase with the number of antiglaucoma medications, the number of drops instilled per day, and history of treatment changes due to ocular intolerance.Citation12 Several studies have found an association between specific ocular surface signs and antiglaucoma medications.Citation15,Citation25,Citation26,Citation39,Citation40 Greater number of antiglaucoma medications and duration of therapy were found to be predictors of abnormal ocular surface staining, Schirmer test <5 mm, and tear breakup time of ≤ 5 seconds.Citation25 Greater number of antiglaucoma drops instilled per day was also found to be associated with abnormal corneal staining and shorter tear breakup time.Citation15,Citation26,Citation40 Longitudinal studies of antiglaucoma medications in treatment-naïve patients with glaucoma have also shown an association between antiglaucoma medication and ocular surface signs.Citation41–43 In newly diagnosed individuals with glaucoma, treatment with antiglaucoma medication was associated with a decrease in tear breakup time from 11.7 seconds at baseline to 8.3 seconds at 3 months.Citation41 In another study, treatment with antiglaucoma medication was associated with decreased Schirmer scores, increased prevalence of tear film breakup time of <10 seconds, and increased ocular surface staining over 4 months.Citation42

The Role of Preservatives and Topical Antiglaucoma Medication Formulations

Several studies have found an association between ocular surface signs and the use of preserved antiglaucoma medication (summarized in ).Citation17,Citation19,Citation44–46 Preservatives such as benzalkonium chloride (BAK) are added to antiglaucoma medications for their antimicrobial actions in multidose eyedrop containers, which are susceptible to contamination.Citation47 Using multivariate analysis, Rossi et al found that the number of glaucoma medications, prolonged use of glaucoma medications, and exposure to at least 2000 µg of BAK were predictors of ocular surface disease (mean daily BAK was 6 µg).Citation45 Similarly, Lee et al found that a greater amount of BAK instilled per day was correlated with worse corneal epithelial punctate erosion and shorter tear breakup time.Citation48 Other studies have compared ocular surface signs in patients treated with preserved antiglaucoma medications and in patients treated with preservative-free medications. Villani et al found that individuals treated with antiglaucoma medications with BAK had shorter tear breakup times than those treated with preservative-free antiglaucoma medications, and lower Schirmer scores than those treated with preservative-free antiglaucoma medications or with antiglaucoma medications preserved with polyquad.Citation39 Similarly, Zaleska-Żmijewska et al found a higher prevalence of tear breakup time of <5 seconds among patients receiving antiglaucoma medication with BAK (50%) than among those receiving preservative-free antiglaucoma medications (10%).Citation46 In a study that examined the relationship between DED and specific antiglaucoma medications, the odds of DED were found to increase with the use of each type of medication examined (prostaglandin analogue, β-blocker, carbonic anhydrase inhibitor, pilocarpine, and combination medications) except for ⍺-agonists.Citation9 It was suggested that the lack of an association between DED and ⍺-agonists may have been related to the use of Purite® as a preservative for ⍺-agonists as opposed to BAK or benzododecinium bromide.Citation9

Table 1 Studies Examining the Ocular Surface Effects of Preservatives in Glaucoma Medications

In addition to the preservatives in antiglaucoma medication, there is evidence to suggest that antiglaucoma medications may contribute to DED even in the absence of preservatives. One study found greater corneal epithelial punctate erosion in patients who used β-blockers compared to those who used non-β-blocker antiglaucoma medications when adjusting for amount of BAK instilled per day, suggesting that β-blockers may contribute to corneal epithelial punctate erosion independently of preservatives.Citation48 Furthermore, in a study that examined patients treated with preservative-free antiglaucoma monotherapy (tafluprost 0.0015% or timolol maleate 0.1%) for at least 36 months, patients treated with preservative-free timolol had shorter tear breakup times and patients treated with either preservative-free timolol or preservative-free tafluprost had higher OSDI scores than age- and sex-matched controls.Citation49 These findings suggest that in addition to antiglaucoma medications with preservatives, formulations without preservatives may contribute to ocular surface signs and symptoms.

Topical Antiglaucoma Medication and Meibomian Gland Dysfunction

Meibomian gland loss and meibum scores have also been associated with antiglaucoma medications, which is relevant given that meibomian gland dysfunction may contribute to DED.Citation33 Ha et al found that patients treated with preservative-containing antiglaucoma medications had greater meibomian gland loss and poorer meibum scores than patients treated with a preservative-free antiglaucoma medication after 12 months.Citation50 However, patients treated with preservative-free drops also showed worse meibomian gland measures than normal controls, suggesting that antiglaucoma medications with or without preservatives have a negative effect on meibomian glands, but preservative use was worse for meibomian glands.Citation50 In a separate study, patients using prostaglandin analogues were found to have a higher prevalence of meibomian gland dysfunction compared to patients using non-prostaglandin analogue antiglaucoma medication.Citation51

Topical Antiglaucoma Medications and Ocular Surface Inflammation

The association between antiglaucoma therapy and DED suggests that patients with glaucoma may benefit from evaluation of the ocular surface. Addressing DED in patients with glaucoma is important because DED can lead to ocular surface inflammation and damage.Citation5,Citation33 There is evidence indicating that eyes receiving topical antiglaucoma medication have increased ocular surface expression of inflammatory markers.Citation22,Citation52–56 This may be because of antiglaucoma medication, the DED, or a combination of these. Benitez-del-Castillo et al found higher expression of interleukin (IL)-6 in the tears of eyes receiving antiglaucoma medication compared with the tears of control eyes.Citation22 Furthermore, higher expression of IL-1β was found in eyes receiving antiglaucoma medication with preservatives compared with eyes receiving antiglaucoma medication without preservatives.Citation22 Increased expression of IL-6, IL-8, and IL-1β in tears of eyes receiving preserved antiglaucoma medication was also found in a study in which patients were randomized to antiglaucoma medications with preservatives or without preservatives.Citation53 To investigate involvement of T helper (Th) 1 and Th2 pathways in eyes receiving antiglaucoma medication, Baudouin et al examined expression of chemokine receptors CCR5 and CCR4 as markers of these pathways, respectively, in eyes treated with antiglaucoma medication for more than 1 year.Citation57 Increased expression of both CCR5 and CCR4 was found in the conjunctival epithelium, suggesting involvement of both allergic and Th1 mechanisms in the ocular surface of patients with glaucoma.Citation57 In addition to cytokines and chemokines, a higher prevalence of elevated MMP-9 levels has been found in eyes treated with antiglaucoma medications with BAK (47%) compared with untreated eyes suspected of having glaucoma (17%) and eyes treated with preservative-free antiglaucoma medication (17%), suggesting inflammation or ocular surface damage in eyes receiving antiglaucoma medications with BAK.Citation46 Interestingly, the tears of eyes receiving antiglaucoma medication have been found to have higher levels of IL-6, tumor necrosis factor-⍺, and vascular endothelial growth factor and lower levels of IL-4 than those of eyes with DED without glaucoma, suggesting activity of different inflammatory signaling pathways between the two groups.Citation22

Topical Antiglaucoma Medications and Ocular Surface Alterations

In addition to inflammatory markers, studies have found anatomical differences in the ocular surface of eyes receiving antiglaucoma therapy beyond what has been observed through ocular surface staining. Kamath et al found that after 1 year of treatment with antiglaucoma medication, the prevalence of goblet cell density <50 cells/HPF increased from 2.2% at baseline to 32%, the prevalence of inflammatory cells increased from 9% to 37.5%, and the prevalence of squamous metaplasia increased from 10.2% to 45%.Citation54 Saini et al found that the density of basal epithelial cells in the central cornea was increased in individuals on antiglaucoma therapy compared with controls, and the number, length, and density of the central-corneal sub-basal nerve fiber layer was reduced.Citation58 The corneal nerve plexus is important for the health of the cornea, and damage to it can lead to a variety of ocular-surface-related issues.Citation58 A negative association of the central-corneal sub-basal nerve fiber layer density with corneal staining score and OSDI was found, as well as a positive association between sub-basal nerve fiber layer density and tear breakup time.Citation58 In addition to reduced sub-basal nerve fiber density, greater nerve tortuosity has been found with use of antiglaucoma medications. Villani et al found increased nerve length, nerve tortuosity, and dendritic cell density at the sub-basal nerve plexus in patients receiving antiglaucoma medication compared with controls, which was suggested to result from neuroinflammatory processes.Citation39 The effect of topical application of BAK on corneal nerves has been examined in animal models. In one study of mouse eyes, BAK-treated corneas showed significantly reduced nerve fiber density.Citation59 Other findings from this study included reduction in aqueous tear production, increased inflammatory cell infiltration, and corneal fluorescein staining.Citation59 Antiglaucoma medication formulations without preservatives may also contribute to anatomical alterations. Rolle et al found greater basal epithelial cell density, greater stromal reflectivity, a lower number of sub-basal nerves, and greater sub-basal nerve tortuosity in patients treated with preservative-free timolol or preservative-free tafluprost compared to controls.Citation49 Overall, these studies indicate that there can be ocular surface alterations in patients treated with antiglaucoma medications.

Procedures for Glaucoma and Dry Eye Disease

Procedures for glaucoma, including surgery and laser procedures, may reduce the burden of antiglaucoma medications. However, similar to antiglaucoma medications, certain procedures may be associated with ocular surface signs and symptoms.Citation60,Citation61 Trabeculectomy is a filtering surgery that involves the creation of a drainage path from the anterior chamber to the subconjunctival space.Citation62 Following trabeculectomy, Zhong et al found reductions in tear breakup time and increases in corneal staining, which did not fully recover to baseline at 3 months postsurgery.Citation61 Ocular surface signs following trabeculectomy may be related to the presence of the filtering bleb.Citation61,Citation63 Patients with glaucoma with filtering blebs not using topical medications were found to have higher rates of DED, shorter tear breakup times, and greater corneal staining compared with individuals without glaucoma.Citation63,Citation64 Shorter tear breakup times, greater corneal staining, and presence of DED were all associated with higher blebs, suggesting that the bleb may interfere with blinking and spreading of the tear film.Citation64 There is also evidence to suggest that trabeculectomy may contribute to loss of meibomian glands. Sagara et al found greater loss of meibomian glands in the upper-eyelid area that contacted the bleb compared with the area that did not contact the bleb in patients with glaucoma who underwent trabeculectomy with mitomycin C.Citation65 In contrast, other studies have found evidence of improvements in the ocular surface following trabeculectomy,Citation66,Citation67 which have been postulated to be due to discontinuation of antiglaucoma medications and use of postoperative steroids.Citation67 In a study that compared trabeculectomy-treated eyes with fellow eyes treated with antiglaucoma medication, trabeculectomy-treated eyes were found to have longer tear breakup times and less corneal staining compared to fellow eyes.Citation68 Together these findings suggest that trabeculectomy may negatively affect the ocular surface, but the effects may be less than the negative effects of antiglaucoma medications.

In addition to trabeculectomy, other surgeries and procedures exist including laser trabeculoplasty, placement of tube shunts, and minimally invasive glaucoma surgery (MIGS).Citation69 To the authors’ knowledge, a negative association between these glaucoma procedures and the ocular surface has not been demonstrated. Importantly, patients may continue to require glaucoma medications after surgical procedures.Citation69

Impact of Dry Eye Disease on Patients with Glaucoma

Impact of DED on Quality of Life

The evaluation of DED in patients with glaucoma is important because of the negative impact that DED may have on quality of life. Rossi et al found that patients with glaucoma with ocular surface disease had poorer nonvisual-symptoms scores (eg, burning, tearing, dryness, itching as measured with the Glaucoma Symptom Scale [GSS]) and poorer vision-related quality of life (as measured by the National Eye Institute-Visual Function Questionnaire [NEI-VFQ 25]) compared with patients with glaucoma without ocular surface disease.Citation16,Citation45 Other studies have found a relationship between antiglaucoma medications and quality of life. Camp et al found that patients on antiglaucoma medications had poorer dry eye–related emotional well-being scores (as measured with the Impact of Dry Eye on Everyday Life [IDEEL] questionnaire) compared with patients not on antiglaucoma medications.Citation70 Furthermore, use of a greater number of antiglaucoma medications was associated with poorer dry eye–related emotional well-being scores.Citation70 Similarly, Rossi et al found that a greater number of antiglaucoma drops instilled per day was associated with poorer vision-related quality of life (NEI-VFQ 25), GSS total scores, and GSS symptom scores.Citation71 However, this study failed to find a difference in quality of life between individuals with glaucoma and DED and individuals with glaucoma without DED.Citation71 Poorer quality of life has also been associated with the use of preservatives in antiglaucoma medications. Kumar et al found that patients receiving antiglaucoma medication with BAK had poorer Glaucoma Quality of Life-15 (GQL-15) scores and higher OSDI scores compared with patients receiving antiglaucoma medication without BAK and controls, suggesting poorer visual disability and ocular surface symptoms in the patients receiving antiglaucoma medication with BAK.Citation72 No significant difference in the GQL-15 or OSDI was found between the patients receiving antiglaucoma medication without BAK and controls.Citation72 There is evidence to suggest that reducing antiglaucoma medication has a positive effect on quality of life. In a post hoc analysis examining an implanted trabecular microbypass stent (iStent inject® W) plus cataract surgery, the prevalence of patients who showed a response in vision-related quality of life was greater for patients who received the stent compared to patients who received cataract surgery alone.Citation73 Furthermore, those who showed a response in vision-related quality of life were more likely to be antiglaucoma medication–free at the end of the study, suggesting improved vision-related quality of life with reduced antiglaucoma medication use.Citation73

Impact of DED on Glaucoma Management

In addition to the negative effects on quality of life, DED in patients with glaucoma may negatively affect adherence to antiglaucoma medications. Stringham et al found that the rate of topical antiglaucoma treatment compliance was lower among patients with glaucoma with dry eye symptoms (63%) than among patients with glaucoma without dry eye symptoms (89%).Citation74 Poorer antiglaucoma medication compliance has also been associated with greater meibomian gland loss, poorer meibum expression, and greater lid margin abnormality.Citation30 Since meibomian gland dysfunction is associated with DED,Citation33 these results further suggest a role of DED in antiglaucoma medication compliance. In another study, patients with glaucoma who experienced treatment adverse effects and those who reported being unhappy with their treatment more often experienced glaucoma disease progression.Citation75 These findings suggest the importance of treating DED in patients with glaucoma to facilitate treatment adherence.

There is some evidence to suggest that treatment of DED may improve management of glaucoma. In 2 case series, treatment changes aimed at addressing ocular surface disease in patients with glaucoma resulted in improvements in both ocular surface signs and in IOP.Citation76,Citation77 Although these findings require further investigation, they suggest that addressing ocular surface disease in patients with glaucoma may help improve IOP in some patients.Citation76,Citation77 Treatment of ocular surface disease in patients with glaucoma has also been found to result in improvement in optical coherence tomography signal quality, which is important for monitoring changes in glaucoma progression.Citation78 Treatment of DED may also be important for the outcome of trabeculectomy because trabeculectomy failure has been associated with higher levels of presurgical conjunctival inflammatory cells.Citation79

Clinical Perspective: Managing Dry Eye Disease in Patients with Glaucoma

Assessment of DED in Patients with Glaucoma

Because of the increased risk of DED in patients with glaucoma, patients with glaucoma may benefit from evaluation for DED. An initial assessment for DED should be conducted before starting glaucoma medications to facilitate interpretations of subsequent assessments for DED. Questionnaires such as the OSDI and the Dry Eye Questionnaire – 5 item (DEQ-5) may be helpful for detecting DED.Citation6 The DEQ-5 may be more appropriate for patients with glaucoma than the OSDI because the DEQ-5 comprises questions on ocular surface symptoms without questions on visual function, which could be influenced by visual field loss related to glaucoma.Citation6,Citation23 Ideally, ocular surface signs should also be assessed in addition to symptoms because patients with DED may have signs without symptoms, which may warrant treatment to prevent further manifestations of DED.Citation6 Clinical tests that can be used in the diagnosis of DED include tear breakup time, osmolarity testing, ocular surface staining, and MMP-9 testing.Citation6 Additional tests to consider for further characterizing the subtype of DED as primarily aqueous deficient (occurring with decreased lacrimal secretion), primarily evaporative (occurring with an increased rate of tear film evaporation and normal lacrimal secretion), or a mixed subtype include evaluation of tear volume, meibography, and meibomian expressibility.Citation6,Citation33

Treatment of DED in Patients with Glaucoma

To address DED in patients with glaucoma, antiglaucoma medications may need to be adjusted. Switching to antiglaucoma medications without preservatives has been found to result in improvements in ocular surface signs and symptoms.Citation80–82 Furthermore, preservative-free antiglaucoma medications have been associated with better treatment adherence than antiglaucoma medications with preservatives.Citation83 If preservative-free medications are not an option, combination medications may be considered to reduce the number of drops for those patients who require more than 1 active molecule to control IOP.Citation84 Switching the active component may also be important in some cases because patients may have allergies or sensitivities to specific medications.Citation57 Furthermore, switching from a generic to a brand name for predictability of drop characteristics may also be considered.Citation85 A sustained-release implant for delivering bimatoprost is also available and may be considered as an alternative to topical application of antiglaucoma medication, although it is currently indicated for a single administration only and is therefore not an option for long-term treatment; moreover, it is not an option for all glaucoma patients.Citation86–88 In addition to switching medications or drug delivery methods, laser, MIGS, and traditional incisional procedures may also be considered to reduce the burden of antiglaucoma medications.Citation69 For example, MIGS has been found to result in reduced antiglaucoma medication use and improved ocular surface signs and symptoms.Citation89,Citation90 Furthermore, selective laser trabeculoplasty may be considered as first-line therapy for glaucoma in place of antiglaucoma medication and has been associated with lower rates of eyelid erythema and conjunctival hyperemia compared with antiglaucoma medication.Citation91,Citation92 It should be noted, however, that for some patients there are significant barriers to such interventions. For example, some patients or surgeons may not be comfortable with the risks of surgery, whereas for other patients, surgery may not be an option for their type of glaucoma.Citation69,Citation91 Moreover, costs for certain procedures may be prohibitive.Citation69

In addition to modifications to antiglaucoma medications, treatment for DED should be considered to restore the homeostasis of the ocular surface.Citation93 A variety of treatments are available for DED, the choice of which depends on the severity and subtype of DED.Citation93 Topical treatments for DED that may be considered for either evaporative or aqueous-deficient DED include ocular lubricants and topical anti-inflammatory treatments.Citation94 Ocular lubricants come in a variety of formulations, some of which have lipids to supplement the lipid layer of the tear film,Citation95 and some of which are preservative-free.Citation96 Topical anti-inflammatory treatments include corticosteroids (for short-term treatment, as long-term use is associated with numerous complicationsCitation97), cyclosporine A, and lifitegrast.Citation97 For patients with evaporative DED related to meibomian gland dysfunction, topical or oral antibiotics may be considered as well as procedures such as therapeutic meibomian gland expression, thermal pulsation, intraductal probing, and intense pulsed light.Citation98–102 Notably, risks of dermatologic side effects with intense pulsed light may be greater for darker skin types.Citation103 A recently approved perfluorohexyloctane ophthalmic solution (MieboTM) has shown promise for the treatment of DED associated with meibomian gland dysfunction, and although it is not contraindicated for glaucoma patients, its safety and efficacy have yet to be evaluated in patients concomitantly administering IOP-lowering drugs.Citation104–107 Therapies to promote tear retention or tear production for aqueous-deficient DED include punctal plugs and varenicline solution nasal spray, respectively.Citation108,Citation109 Several treatments for DED have been associated with improvements in OSDI and/or ocular surface signs for patients receiving antiglaucoma medications, including certain ocular lubricants, topical immunomodulatory treatments, and punctal plugs.Citation110–116 For patients who do not respond to a given treatment, additional or alternative treatments may be considered.Citation93 Because patients with glaucoma typically already have a medication burden, patient education on the importance of DED medications for addressing symptoms may help improve compliance with DED medications. provides a summary of assessment and treatment considerations for addressing DED in patients with glaucoma.

Table 2 Considerations for the Assessment and Management of DED in Patients with Glaucoma

Conclusion

DED occurs at a higher prevalence in individuals with glaucoma than in individuals without glaucoma, which may be related to therapy for glaucoma. Assessment and treatment of DED in patients with glaucoma is important to prevent ocular surface damage, improve quality of life, and facilitate treatment adherence to antiglaucoma medications. As such, patients with glaucoma may benefit from evaluation for DED. Management of DED may include modifications to antiglaucoma medications and use of treatments for DED.

Abbreviations

BAK, benzalkonium chloride; DED, dry eye disease; DEQ-5, Dry Eye Questionnaire – 5 item; GQL-15, Glaucoma Quality of Life-15; IDEEL, Impact of Dry Eye on Everyday Life; IL, interleukin; IOP, intraocular pressure; MGD, meibomian gland dysfunction; MIGS, minimally invasive glaucoma surgery; MMP-9, matrix metalloproteinase 9; NEI-VFQ 25, National Eye Institute-Visual Function Questionnaire; OR, odds ratio; OSDI, Ocular Surface Disease Index; Th, T helper.

Author Contributions

All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.

Disclosure

Lisa M Nijm, MD, JD, has been a consultant/advisor for Aerie Pharmaceuticals, Alcon Laboratories, Allergan, Bausch + Lomb, Bruder Healthcare Company, Carl Zeiss Meditec, CooperVision, Dompe, Horizon, Johnson & Johnson Vision, Novartis, Ocular Therapeutix, Rayner, Oyster Point, Sun Ophthalmics, TearLab Corporation, Thea Pharmaceuticals; has received lecture fees and/or has been on a speaker bureau for Allergan, Bausch + Lomb, Oyster Point, Sun Ophthalmics; and holds equity/stock in TearLab Corporation. Justin Schweitzer, OD, has received relevant honoraria for consulting and speaking on the subject matter from Allergan, Glaukos, Novartis Pharmaceuticals Corporation, Sun Ophthalmics, and Quidel. Jennifer Gould Blackmore, OD, has received relevant honoraria for consulting and speaking on the subject matter from Aerie Pharmaceuticals, Bausch + Lomb, and Allergan. The authors report no other conflicts of interest in this work.

Acknowledgment

Medical writing support was provided by Ann E. Todd (IMPRINT Science, New York, NY) and was funded by Novartis Pharmaceuticals Corporation. Inder Paul Singh contributed to conceptualizing and critically reviewing this work. This manuscript was developed in accordance with Good Publication Practice (GPP3) guidelines. Authors had full control of the content and made the final decision on all aspects of this publication.

Additional information

Funding

This work was funded by Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA.

References

  • Weinreb RN, Aung T, Medeiros FA. The pathophysiology and treatment of glaucoma: a review. JAMA. 2014;311(18):1901–1911 doi:10.1001/jama.2014.3192.
  • Heijl A, Leske MC, Bengtsson B, et al. Reduction of intraocular pressure and glaucoma progression: results from the Early Manifest Glaucoma Trial. Arch Ophthalmol. 2002;120(10):1268–1279. doi:10.1001/archopht.120.10.1268
  • Robin A, Grover DS. Compliance and adherence in glaucoma management. Indian J Ophthalmol. 2011;59(1):S93–96. doi:10.4103/0301-4738.73693
  • Sleath B, Blalock S, Covert D, et al. The relationship between glaucoma medication adherence, eye drop technique, and visual field defect severity. Ophthalmology. 2011;118(12):2398–2402. doi:10.1016/j.ophtha.2011.05.013
  • 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
  • Wolffsohn JS, Arita R, Chalmers R, et al. TFOS DEWS II diagnostic methodology report. Ocul Surf. 2017;15(3):539–574. doi:10.1016/j.jtos.2017.05.001
  • Stapleton F, Alves M, Bunya VY, et al. TFOS DEWS II epidemiology report. Ocul Surf. 2017;15(3):334–365. doi:10.1016/j.jtos.2017.05.003
  • Schmier JK, Covert DW. Characteristics of respondents with glaucoma and dry eye in a national panel survey. Clin Ophthalmol. 2009;3:645–650. doi:10.2147/OPTH.S8241
  • Chen HY, Lin CL, Tsai YY, Kao CH. Association between glaucoma medication usage and dry eye in Taiwan. Optom Vis Sci. 2015;92(9):e227–232. doi:10.1097/OPX.0000000000000667
  • Erb C, Gast U, Schremmer D. German register for glaucoma patients with dry eye. I. Basic outcome with respect to dry eye. Graefes Arch Clin Exp Ophthalmol. 2008;246(11):1593–1601. doi:10.1007/s00417-008-0881-9
  • Kobia-Acquah E, Gyekye GA, Antwi-Adjei EK, et al. Assessment of ocular surface disease in glaucoma patients in Ghana. J Glaucoma. 2021;30(2):180–186. doi:10.1097/IJG.0000000000001713
  • Baudouin C, Renard JP, Nordmann JP, et al. Prevalence and risk factors for ocular surface disease among patients treated over the long term for glaucoma or ocular hypertension. Eur J Ophthalmol. 2013;23(1):47–54. doi:10.5301/ejo.5000181
  • Tirpack AR, Vanner E, Parrish JM, Galor A, Hua HU, Wellik SR. Dry eye symptoms and ocular pain in veterans with glaucoma. J Clin Med. 2019;8(7):1076. doi:10.3390/jcm8071076
  • Costa VP, Marcon IM, Galvão Filho RP, Malta RFS. The prevalence of ocular surface complaints in Brazilian patients with glaucoma or ocular hypertension. Arq Bras Oftalmol. 2013;76(4):221–225. doi:10.1590/S0004-27492013000400006
  • Ruangvaravate N, Prabhasawat P, Vachirasakchai V, Tantimala R. High prevalence of ocular surface disease among glaucoma patients in Thailand. J Ocul Pharmacol Ther. 2018;34(5):387–394. doi:10.1089/jop.2017.0104
  • Rossi GCM, Pasinetti GM, Scudeller L, Bianchi PE. Ocular surface disease and glaucoma: how to evaluate impact on quality of life. J Ocul Pharmacol Ther. 2013;29(4):390–394. doi:10.1089/jop.2011.0159
  • Leung EW, Medeiros FA, Weinreb RN. Prevalence of ocular surface disease in glaucoma patients. J Glaucoma. 2008;17(5):350–355. doi:10.1097/IJG.0b013e31815c5f4f
  • Stalmans I, Lemij H, Clarke J, Baudouin C. GOAL study group. Signs and symptoms of ocular surface disease: the reasons for patient dissatisfaction with glaucoma treatments. Clin Ophthalmol. 2020;14:3675–3680. doi:10.2147/OPTH.S269586
  • Pérez-Bartolomé F, Martínez-de-la-Casa JM, Arriola-Villalobos P, Fernández-Pérez C, Polo V, García-Feijoó J. Ocular surface disease in patients under topical treatment for glaucoma. Eur J Ophthalmol. 2017;27(6):694–704. doi:10.5301/ejo.5000977
  • Saade CE, Lari HB, Berezina TL, Fechtner RD, Khouri AS. Topical glaucoma therapy and ocular surface disease: a prospective, controlled cohort study. Can J Ophthalmol. 2015;50(2):132–136. doi:10.1016/j.jcjo.2014.11.006
  • Jandroković S, Suić SP, Kordić R, Kuzman T, Petricek I. Tear film status in glaucoma patients. Coll Antropol. 2013;37(1):137–140.
  • Benitez-del-Castillo J, Cantu-Dibildox J, Sanz-González SM, Zanón-Moreno V, Pinazo-Duran MD. Cytokine expression in tears of patients with glaucoma or dry eye disease: a prospective, observational cohort study. Eur J Ophthalmol. 2019;29(4):437–443. doi:10.1177/1120672118795399
  • Mathews PM, Ramulu PY, Friedman DS, Utine CA, Akpek EK. Evaluation of ocular surface disease in patients with glaucoma. Ophthalmology. 2013;120(11):2241–2248. doi:10.1016/j.ophtha.2013.03.045
  • Ramli N, Supramaniam G, Samsudin A, Juana A, Zahari M, Choo MM. Ocular surface disease in glaucoma: effect of polypharmacy and preservatives. Optom Vis Sci. 2015;92(9):e222–226. doi:10.1097/OPX.0000000000000542
  • Ghosh S, O’Hare F, Lamoureux E, Vajpayee RB, Crowston JG. Prevalence of signs and symptoms of ocular surface disease in individuals treated and not treated with glaucoma medication. Clin Exp Ophthalmol. 2012;40(7):675–681. doi:10.1111/j.1442-9071.2012.02781.x
  • Sahlu M, Giorgis AT. Dry eye disease among glaucoma patients on topical hypotensive medications, in a tertiary hospital, Ethiopia. BMC Ophthalmol. 2021;21(1):155. doi:10.1186/s12886-021-01917-3
  • Guarnieri A, Carnero E, Bleau AM, López de Aguileta Castaño N, Llorente Ortega M, Moreno-Montañés J. Ocular surface analysis and automatic non-invasive assessment of tear film breakup location, extension and progression in patients with glaucoma. BMC Ophthalmol. 2020;20(1):12. doi:10.1186/s12886-019-1279-7
  • Wong ABC, Wang MTM, Liu K, Prime ZJ, Danesh-Meyer HV, Craig JP. Exploring topical anti-glaucoma medication effects on the ocular surface in the context of the current understanding of dry eye. Ocul Surf. 2018;16(3):289–293. doi:10.1016/j.jtos.2018.03.002
  • Arita R, Itoh K, Maeda S, et al. Comparison of the long-term effects of various topical antiglaucoma medications on meibomian glands. Cornea. 2012;31(11):1229–1234. doi:10.1097/ICO.0b013e31823f8e7d
  • Lee TH, Sung MS, Heo H, Park SW. Association between meibomian gland dysfunction and compliance of topical prostaglandin analogs in patients with normal tension glaucoma. PLoS One. 2018;13(1):e0191398. doi:10.1371/journal.pone.0191398
  • Albakeri A, Taşkıran Çömez A. Assessment of meibomian glands with topography in patients using unilateral antiglaucoma drops. Eur Eye Res. 2022;2(1):1–8. doi:10.14744/eer.2022.54264
  • Soriano D, Ferrandez B, Mateo A, Polo V, Garcia-Martin E. Meibomian gland changes in open-angle glaucoma users treated with topical medication. Optom Vis Sci. 2021;98(10):1177–1182. doi:10.1097/OPX.0000000000001782
  • Bron AJ, de Paiva CS, Chauhan SK, et al. TFOS DEWS II pathophysiology report. Ocul Surf. 2017;15(3):438–510. doi:10.1016/j.jtos.2017.05.011
  • Tomlinson A, Bron AJ, Korb DR, et al. The international workshop on meibomian gland dysfunction: report of the diagnosis subcommittee. Invest Ophthalmol Vis Sci. 2011;52(4):2006–2049. doi:10.1167/iovs.10-6997f
  • Willcox MDP, Argueso P, Georgiev GA, et al. TFOS DEWS II tear film report. Ocul Surf. 2017;15(3):366–403. doi:10.1016/j.jtos.2017.03.006
  • Kuppens EV, van Best JA, Sterk CC, de Keizer RJ. Decreased basal tear turnover in patients with untreated primary open-angle glaucoma. Am J Ophthalmol. 1995;120(1):41–46. doi:10.1016/S0002-9394(14)73757-2
  • Schuster AK, Erb C, Hoffmann EM, Dietlein T, Pfeiffer N. The diagnosis and treatment of glaucoma. Dtsch Arztebl Int. 2020;117(13):225–234. doi:10.3238/arztebl.2020.0225
  • Hoy SM. Netarsudil ophthalmic solution 0.02%: first global approval. Drugs. 2018;78(3):389–396. doi:10.1007/s40265-018-0877-7
  • Villani E, Sacchi M, Magnani F, et al. The ocular surface in medically controlled glaucoma: an in vivo confocal study. Invest Ophthalmol Vis Sci. 2016;57(3):1003–1010. doi:10.1167/iovs.15-17455
  • Cvenkel B, Ŝtunf Ŝ, Srebotnik Kirbiŝ I, Strojan Fležar M. Symptoms and signs of ocular surface disease related to topical medication in patients with glaucoma. Clin Ophthalmol. 2015;9:625–631. doi:10.2147/OPTH.S81247
  • Tomić M, Kaštelan S, Soldo KM, Salopek-Rabatić J. Influence of BAK-preserved prostaglandin analog treatment on the ocular surface health in patients with newly diagnosed primary open-angle glaucoma. Biomed Res Int. 2013;2013:603782. doi:10.1155/2013/603782
  • Su CC, Lee YC, Lee PRC. Assessment of ocular surface disease in glaucoma patients with benzalkonium chloride-preserved latanoprost eye drops: a short-term longitudinal study. Graefes Arch Clin Exp Ophthalmol. 2021;259(5):1243–1251. doi:10.1007/s00417-020-05067-y
  • Firat PG, Samdanci E, Doganay S, Cavdar M, Sahin N, Gunduz A. Short-term effect of topical brinzolamide-timolol fixed combination on ocular surface of glaucoma patients. Int J Ophthalmol. 2012;5(6):714–718. doi:10.3980/j.issn.2222-3959.2012.06.12
  • Labbé A, Terry O, Brasnu E, Van Went C, Baudouin C. Tear film osmolarity in patients treated for glaucoma or ocular hypertension. Cornea. 2012;31(9):994–999. doi:10.1097/ICO.0b013e31823f8cb6
  • Rossi GCM, Pasinetti GM, Scudeller L, Raimondi M, Lanteri S, Bianchi PE. Risk factors to develop ocular surface disease in treated glaucoma or ocular hypertension patients. Eur J Ophthalmol. 2013;23(3):296–302. doi:10.5301/ejo.5000220
  • Zaleska-Żmijewska A, Strzemecka E, Wawrzyniak ZM, Szaflik JP. Extracellular MMP-9-based assessment of ocular surface inflammation in patients with primary open-angle glaucoma. J Ophthalmol. 2019;2019:1240537. doi:10.1155/2019/1240537
  • Baudouin C, Labbé A, Liang H, Pauly A, Brignole-Baudouin F. Preservatives in eyedrops: the good, the bad and the ugly. Prog Retin Eye Res. 2010;29(4):312–334. doi:10.1016/j.preteyeres.2010.03.001
  • Lee S, Kim MK, Choi HJ, Wee WR, Kim DM. Comparative cross-sectional analysis of the effects of topical antiglaucoma drugs on the ocular surface. Adv Ther. 2013;30(4):420–429. doi:10.1007/s12325-013-0021-8
  • Rolle T, Spinetta R, Nuzzi R. Long term safety and tolerability of Tafluprost 0.0015% vs Timolol 0.1% preservative-free in ocular hypertensive and in primary open-angle glaucoma patients: a cross sectional study. BMC Ophthalmol. 2017;17(1):136. doi:10.1186/s12886-017-0534-z
  • Ha JY, Sung MS, Park SW. Effects of preservative on the meibomian gland in glaucoma patients treated with prostaglandin analogues. Chonnam Med J. 2019;55(3):156–162. doi:10.4068/cmj.2019.55.3.156
  • Mocan MC, Uzunosmanoglu E, Kocabeyoglu S, Karakaya J, Irkec M. The association of chronic topical prostaglandin analog use with meibomian gland dysfunction. J Glaucoma. 2016;25(9):770–774. doi:10.1097/IJG.0000000000000495
  • Kim DW, Seo JH, Lim SH. Evaluation of ocular surface disease in elderly patients with glaucoma: expression of matrix metalloproteinase-9 in tears. Eye. 2021;35(3):892–900. doi:10.1038/s41433-020-0993-y
  • Mohammed I, Kulkarni B, Faraj LA, Abbas A, Dua HS, King AJ. Profiling ocular surface responses to preserved and non-preserved topical glaucoma medications: a 2-year randomized evaluation study. Clin Exp Ophthalmol. 2020;48(7):973–982. doi:10.1111/ceo.13814
  • Kamath AP, Satyanarayana S, Rodrigues F. Ocular surface changes in primary open angle glaucoma with long term topical anti glaucoma medication. Med J Armed Forces India. 2007;63(4):341–345. doi:10.1016/S0377-1237(07)80011-6
  • Martinez-de-la-Casa JM, Perez-Bartolome F, Urcelay E, et al. Tear cytokine profile of glaucoma patients treated with preservative-free or preserved latanoprost. Ocul Surf. 2017;15(4):723–729. doi:10.1016/j.jtos.2017.03.004
  • Wong TT, Zhou L, Li J, et al. Proteomic profiling of inflammatory signaling molecules in the tears of patients on chronic glaucoma medication. Invest Ophthalmol Vis Sci. 2011;52(10):7385–7391. doi:10.1167/iovs.10-6532
  • Baudouin C, Liang H, Hamard P, et al. The ocular surface of glaucoma patients treated over the long term expresses inflammatory markers related to both T-helper 1 and T-helper 2 pathways. Ophthalmology. 2008;115(1):109–115. doi:10.1016/j.ophtha.2007.01.036
  • Saini M, Vanathi M, Dada T, Agarwal T, Dhiman R, Khokhar S. Ocular surface evaluation in eyes with chronic glaucoma on long term topical antiglaucoma therapy. Int J Ophthalmol. 2017;10(6):931–938. doi:10.18240/ijo.2017.06.16
  • Sarkar J, Chaudhary S, Namavari A, et al. Corneal neurotoxicity due to topical benzalkonium chloride. Invest Ophthalmol Vis Sci. 2012;53(4):1792–1802. doi:10.1167/iovs.11-8775
  • Lee SY, Wong TT, Chua J, Boo C, Soh YF, Tong L. Effect of chronic anti-glaucoma medications and trabeculectomy on tear osmolarity. Eye. 2013;27(10):1142–1150. doi:10.1038/eye.2013.144
  • Zhong S, Zhou H, Chen X, Zhang W, Yi L. Influence of glaucoma surgery on the ocular surface using oculus keratograph. Int Ophthalmol. 2019;39(4):745–752. doi:10.1007/s10792-018-0869-3
  • Carnevale C, Riva I, Roberti G, et al. Confocal microscopy and anterior segment optical coherence tomography imaging of the ocular surface and bleb morphology in medically and surgically treated glaucoma patients: a review. Pharmaceuticals. 2021;14(6): 581. doi:10.3390/ph14060581
  • Neves Mendes CR, Hida RY, Kasahara N. Ocular surface changes in eyes with glaucoma filtering blebs. Curr Eye Res. 2012;37(4):309–311. doi:10.3109/02713683.2011.635400
  • Ji H, Zhu Y, Zhang Y, Li Z, Ge J, Zhuo Y. Dry eye disease in patients with functioning filtering blebs after trabeculectomy. PLoS One. 2016;11(3):e0152696. doi:10.1371/journal.pone.0152696
  • Sagara H, Sekiryu T, Noji H, Ogasawara M, Sugano Y, Horikiri H. Meibomian gland loss due to trabeculectomy. Jpn J Ophthalmol. 2014;58(4):334–341. doi:10.1007/s10384-014-0324-6
  • Tong L, Hou AH, Wong TT. Altered expression level of inflammation-related genes and long-term changes in ocular surface after trabeculectomy, a prospective cohort study. Ocul Surf. 2018;16(4):441–447. doi:10.1016/j.jtos.2018.06.005
  • Agnifili L, Brescia L, Oddone F, et al. The ocular surface after successful glaucoma filtration surgery: a clinical, in vivo confocal microscopy, and immune-cytology study. Sci Rep. 2019;9(1):11299. doi:10.1038/s41598-019-47823-z
  • Romano D, De Ruvo V, Fogagnolo P, Farci R, Rossetti LM. Inter-eye comparison of the ocular surface of glaucoma patients receiving surgical and medical treatments. J Clin Med. 2022;11(5):1238. doi:10.3390/jcm11051238
  • Megevand SG, Bron AM. Personalising surgical treatments for glaucoma patients. Prog Retin Eye Res. 2021;81:100879. doi:10.1016/j.preteyeres.2020.100879
  • Camp A, Wellik SR, Tzu JH, et al. Dry eye specific quality of life in veterans using glaucoma drops. Cont Lens Anterior Eye. 2015;38(3):220–225. doi:10.1016/j.clae.2015.02.001
  • Rossi GCM, Tinelli C, Pasinetti GM, Milano G, Bianchi PE. Dry eye syndrome-related quality of life in glaucoma patients. Eur J Ophthalmol. 2009;19(4):572–579. doi:10.1177/112067210901900409
  • Kumar S, Singh T, Ichhpujani P, Vohra S, Thakur S. Correlation of ocular surface disease and quality of life in Indian glaucoma patients: BAC-preserved versus BAC-free travoprost. Turk J Ophthalmol. 2020;50(2):75–81. doi:10.4274/tjo.galenos.2019.29000
  • Samuelson TW, Singh IP, Williamson BK, et al. Quality of life in primary open-angle glaucoma and cataract: an analysis of VFQ-25 and OSDI from the iStent inject® pivotal trial. Am J Ophthalmol. 2021;229:220–229. doi:10.1016/j.ajo.2021.03.007
  • Stringham J, Ashkenazy N, Galor A, Wellik SR. Barriers to glaucoma medication compliance among veterans: dry eye symptoms and anxiety disorders. Eye Contact Lens. 2018;44(1):50–54. doi:10.1097/ICL.0000000000000301
  • Denis P, Lafuma A, Berdeaux G. Medical outcomes of glaucoma therapy from a nationwide representative survey. Clin Drug Investig. 2004;24(6):343–352. doi:10.2165/00044011-200424060-00004
  • Dubrulle P, Labbe A, Brasnu E, et al. Influence of treating ocular surface disease on intraocular pressure in glaucoma patients intolerant to their topical treatments: a report of 10 cases. J Glaucoma. 2018;27(12):1105–1111. doi:10.1097/IJG.0000000000001041
  • Batra R, Tailor R, Mohamed S. Ocular surface disease exacerbated glaucoma: optimizing the ocular surface improves intraocular pressure control. J Glaucoma. 2014;23(1):56–60. doi:10.1097/IJG.0b013e318264cd68
  • Oktay Ö, Dursun Ö, Yılmaz A. The effects of ocular surface disease on optical coherence tomography test results in patients with glaucoma. Eur J Ophthalmol. 2021;31(6):2997–3002. doi:10.1177/1120672121991395
  • Broadway DC, Grierson I, O’Brien C, Hitchings RA. Adverse effects of topical antiglaucoma medication. II. The outcome of filtration surgery. Arch Ophthalmol. 1994;112(11):1446–1454. doi:10.1001/archopht.1994.01090230060021
  • Uusitalo H, Egorov E, Kaarniranta K, Astakhov Y, Ropo A. Benefits of switching from latanoprost to preservative-free tafluprost eye drops: a meta-analysis of two Phase IIIb clinical trials. Clin Ophthalmol. 2016;10:445–454. doi:10.2147/OPTH.S91402
  • El Ameen A, Vandermeer G, Khanna RK, Pisella PJ. Objective ocular surface tolerance in patients with glaucoma treated with topical preserved or unpreserved prostaglandin analogues. Eur J Ophthalmol. 2019;29(6):645–653. doi:10.1177/1120672118805877
  • Hommer A, Schmidl D, Kromus M, et al. Effect of changing from preserved prostaglandins to preservative-free tafluprost in patients with glaucoma on tear film thickness. Eur J Ophthalmol. 2018;28(4):385–392. doi:10.1177/1120672117753703
  • Kim DW, Shin J, Lee CK, Kim M, Lee S, Rho S. Comparison of ocular surface assessment and adherence between preserved and preservative-free latanoprost in glaucoma: a parallel-grouped randomized trial. Sci Rep. 2021;11(1):14971. doi:10.1038/s41598-021-94574-x
  • Nijm LM, De Benito-Llopis L, Rossi GC, Vajaranant TS, Coroneo MT. Understanding the dual dilemma of dry eye and glaucoma: an international review. Asia Pac J Ophthalmol. 2020;9(6):481–490. doi:10.1097/APO.0000000000000327
  • Mammo ZN, Flanagan JG, James DF, Trope GE. Generic versus brand-name North American topical glaucoma drops. Can J Ophthalmol. 2012;47(1):55–61. doi:10.1016/j.jcjo.2011.12.004
  • Craven ER, Walters T, Christie WC, et al. 24-month phase I/II clinical trial of bimatoprost sustained-release implant (bimatoprost SR) in glaucoma patients. Drugs. 2020;80(2):167–179. doi:10.1007/s40265-019-01248-0
  • Medeiros FA, Walters TR, Kolko M, et al. Phase 3, randomized, 20-month study of bimatoprost implant in open-angle glaucoma and ocular hypertension (ARTEMIS 1). Ophthalmology. 2020;127(12):1627–1641. doi:10.1016/j.ophtha.2020.06.018
  • Shirley M. Bimatoprost implant: first approval. Drugs Aging. 2020;37(6):457–462. doi:10.1007/s40266-020-00769-8
  • Schweitzer JA, Hauser WH, Ibach M, et al. Prospective interventional cohort study of ocular surface disease changes in eyes after trabecular micro-bypass stent(s) implantation (iStent or iStent inject) with phacoemulsification. Ophthalmol Ther. 2020;9(4):941–953. doi:10.1007/s40123-020-00290-6
  • Kashiwagi K, Matsubara M. Reduction in ocular hypotensive eyedrops by ab interno trabeculotomy improves not only ocular surface condition but also quality of vision. J Ophthalmol. 2018;2018:8165476. doi:10.1155/2018/8165476
  • Gazzard G, Konstantakopoulou E, Garway-Heath D, et al. Selective laser trabeculoplasty versus eye drops for first-line treatment of ocular hypertension and glaucoma (LiGHT): a multicentre randomised controlled trial. Lancet. 2019;393(10180):1505–1516. doi:10.1016/S0140-6736(18)32213-X
  • Ang GS, Fenwick EK, Constantinou M, et al. Selective laser trabeculoplasty versus topical medication as initial glaucoma treatment: the glaucoma initial treatment study randomised clinical trial. Br J Ophthalmol. 2020;104(6):813–821. doi:10.1136/bjophthalmol-2018-313396
  • Jones L, Downie LE, Korb D, et al. TFOS DEWS II management and therapy report. Ocul Surf. 2017;15(3):575–628. doi:10.1016/j.jtos.2017.05.006
  • Milner MS, Beckman KA, Luchs JI, et al. Dysfunctional tear syndrome: dry eye disease and associated tear film disorders - new strategies for diagnosis and treatment. Curr Opin Ophthalmol. 2017;27(1):3–47. doi:10.1097/01.icu.0000512373.81749.b7
  • Craig JP, Muntz A, Wang MTM, et al. Developing evidence-based guidance for the treatment of dry eye disease with artificial tear supplements: a six-month multicentre, double-masked randomised controlled trial. Ocul Surf. 2021;20:62–69. doi:10.1016/j.jtos.2020.12.006
  • Moshirfar M, Pierson K, Hanamaikai K, Santiago-Caban L, Muthappan V, Passi SF. Artificial tears potpourri: a literature review. Clin Ophthalmol. 2014;8:1419–1433. doi:10.2147/OPTH.S65263
  • 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
  • Kaiserman I, Rabina G, Mimouni M, et al. The effect of therapeutic meibomian glands expression on evaporative dry eye: a prospective randomized controlled trial. Curr Eye Res. 2021;46(2):195–201. doi:10.1080/02713683.2020.1789663
  • Hu J, Zhu S, Liu X. Efficacy and safety of a vectored thermal pulsation system (Lipiflow®) in the treatment of meibomian gland dysfunction: a systematic review and meta-analysis. Graefes Arch Clin Exp Ophthalmol. 2022;260(1):25–39. doi:10.1007/s00417-021-05363-1
  • Kheirkhah A, Kobashi H, Girgis J, Jamali A, Ciolino JB, Hamrah P. A randomized, sham-controlled trial of intraductal meibomian gland probing with or without topical antibiotic/steroid for obstructive meibomian gland dysfunction. Ocul Surf. 2020;18(4):852–856. doi:10.1016/j.jtos.2020.08.008
  • Piyacomn Y, Kasetsuwan N, Reinprayoon U, Satitpitakul V, Tesapirat L. Efficacy and safety of intense pulsed light in patients with meibomian gland dysfunction—a randomized, double-masked, sham-controlled clinical trial. Cornea. 2020;39(3):325–332. doi:10.1097/ICO.0000000000002204
  • Luchs J. Efficacy of topical azithromycin ophthalmic solution 1% in the treatment of posterior blepharitis. Adv Ther. 2008;25(9):858–870. doi:10.1007/s12325-008-0096-9
  • Thaysen-Petersen D, Erlendsson AM, Nash JF, et al. Side effects from intense pulsed light: importance of skin pigmentation, fluence level and ultraviolet radiation-A randomized controlled trial. Lasers Surg Med. 2017;49(1):88–96. doi:10.1002/lsm.22566
  • Tauber J, Berdy GJ, Wirta DL, Krosser S, Vittitow JL; GOBI Study Group. NOV03 for dry eye disease associated with meibomian gland dysfunction: results of the randomized phase 3 GOBI study. Ophthalmology. 2023;130(5):516–524. doi:10.1016/j.ophtha.2022.12.021
  • Sheppard JD, Kurata F, Epitropoulos AT, Krosser S, Vittitow JL; MOJAVE Study Group. NOV03 for signs and symptoms of dry eye disease associated with meibomian gland dysfunction: the randomized phase 3 MOJAVE study. Am J Ophthalmol. 2023;252:265–274. doi:10.1016/j.ajo.2023.03.008
  • Bausch + Lomb and Novaliq Announce FDA Approval of MIEBO™ (perfluorohexyloctane ophthalmic solution) for the treatment of the signs and symptoms of dry eye disease; 2023. Available from: https://ir.bausch.com/press-releases/bausch-lomb-and-novaliq-announce-fda-approval-miebotm-perfluorohexyloctane. Accessed August 5, 2023.
  • MIEBO™ (perfluorohexyloctane ophthalmic solution). Prescribing information. Bausch + Lomb and Novaliq; 2023. Available from: https://www.bausch.com/globalassets/pdf/packageinserts/pharma/miebo-package-insert.pdf. Accessed August 5, 2023.
  • Ervin AM, Wojciechowski R, Schein O. Punctal occlusion for dry eye syndrome. Cochrane Database Syst Rev. 2010;9:CD006775 doi:10.1002/14651858.CD006775.pub2.
  • Wirta D, Vollmer P, Paauw J, et al. Efficacy and safety of OC-01 (varenicline solution) nasal spray on signs and symptoms of dry eye disease: the ONSET-2 phase 3 randomized trial. Ophthalmology. 2022;129(4):379–387. doi:10.1016/j.ophtha.2021.11.004
  • Monaco G, Cacioppo V, Consonni D, Troiano P. Effects of osmoprotection on symptoms, ocular surface damage, and tear film modifications caused by glaucoma therapy. Eur J Ophthalmol. 2011;21(3):243–250. doi:10.5301/EJO.2010.5730
  • So HR, Park HYL, Chung SH, Kim HS, Byun YS. Effect of autologous serum eyedrops on ocular surface disease caused by preserved glaucoma eyedrops. J Clin Med. 2020;9(12):3904. doi:10.3390/jcm9123904
  • Saini M, Dhiman R, Dada T, Tandon R, Vanathi M. Topical cyclosporine to control ocular surface disease in patients with chronic glaucoma after long-term usage of topical ocular hypotensive medications. Eye. 2015;29(6):808–814. doi:10.1038/eye.2015.40
  • Liu X, Yu FF, Zhong YM, Guo XX, Mao Z. Therapeutic effects of sodium hyaluronate on ocular surface damage induced by benzalkonium chloride preserved anti-glaucoma medications. Chin Med J. 2015;128(18):2444–2449. doi:10.4103/0366-6999.164927
  • Prabhasawat P, Ruangvaravate N, Tesavibul N, Thewthong M. Effect of 0.3% hydroxypropyl methylcellulose/dextran versus 0.18% sodium hyaluronate in the treatment of ocular surface disease in glaucoma patients: a randomized, double-blind, and controlled study. J Ocul Pharmacol Ther. 2015;31(6):323–329. doi:10.1089/jop.2014.0115
  • Chen M, Yung Choi S. Preliminary outcomes of temporary collagen punctal plugs for patients with dry eye and glaucoma. Med Hypothesis Discov Innov Ophthalmol. 2020;9(1):56–60.
  • Mylla Boso AL, Gasperi E, Fernandes L, Costa VP, Alves M. Impact of ocular surface disease treatment in patients with glaucoma. Clin Ophthalmol. 2020;14:103–111. doi:10.2147/OPTH.S229815