Skip to main content
Erschienen in: Ophthalmology and Therapy 6/2023

Open Access 19.10.2023 | COMMENTARY

Challenging the "Topical Medications-First” Approach to Glaucoma: A Treatment Paradigm in Evolution

verfasst von: Nathan M. Radcliffe, Manjool Shah, Thomas W. Samuelson

Erschienen in: Ophthalmology and Therapy | Ausgabe 6/2023

Abstract

Topical glaucoma medications are effective and safe, but they have numerous well-documented limitations that diminish their long-term utility and sustainability. These limitations can include high rates of nonadherence (with associated glaucoma progression), concerning side effects, inconsistent circadian intraocular pressure (IOP) control, complex dosing regimens, difficulty with self-administration, costs, and decreased quality of life. Despite these limitations, topical medications traditionally have been first-line in the glaucoma treatment algorithm, as no other minimally invasive treatment alternatives existed. In recent years, however, novel interventional therapies—including sustained-release drug-delivery platforms, selective laser trabeculoplasty, and micro-invasive glaucoma surgery procedures—have made it possible to intervene earlier without relying on topical medications. As a result, the topical medication-first treatment approach is being reevaluated in an overall shift toward earlier more proactive interventions.
Key Summary Points
Topical medications traditionally have been first-line in the glaucoma treatment paradigm. However, their usage is limited by a host of widespread and impactful downsides, including nonadherence (with associated glaucoma progression), side effects, inconsistent circadian intraocular pressure (IOP) control, complex dosing regimens, difficulty with self-administration, costs, and decreased quality of life.
In the past, the limitations of topical medications were accepted due to the lack of viable alternatives, but this may now no longer be the case.
Novel minimally invasive interventions, including sustained-release drug-delivery platforms, selective laser trabeculoplasty, and micro-invasive glaucoma surgery, are being used earlier in the treatment cascade, including as the first-line therapy.
This shift toward earlier intervention has led to an overall reevaluation of the glaucoma treatment paradigm.
In the coming years, it may be possible and desirable for the topical medication-first approach to be replaced by earlier more interventional therapies that are more effective and less adversely impactful on patients, doctors, and society.

Medications First: Reexamining an Aging Glaucoma Treatment Paradigm

For more than 150 years—since the discovery of physostigmine isolated from the Calabar bean [1]—topical medical therapy has been the preferred first-line treatment for primary open-angle glaucoma. Other drugs followed, including pilocarpine and epinephrine, and the era of modern glaucoma pharmacology commenced with the development of timolol in the 1970s [2]. The 1990s saw a rapid expansion of medical options for glaucoma, including the first prostaglandin analogue (latanoprost), the first topical carbonic anhydrase inhibitor (dorzolamide), and the first alpha adrenergic agonist (brimonidine) [2]. The first-line utilization of medications has been validated in numerous multicentered clinical trials designed to evaluate its efficacy and safety in the primary treatment of open-angle glaucoma (OAG) or ocular hypertension (OHT) [37]. As recently as 2020, the American Academy of Ophthalmology recommended topical medications as the preferred first-line therapy for newly diagnosed POAG or OHT over other treatment modalities for most patients [8].
Topical medical therapy for the treatment of glaucoma is generally considered safe and effective as commonly utilized medications are required to undergo a series of clinical trials to achieve regulatory approval around the world. There is no question that the approved compounds considered foundational therapy are highly effective in reducing intraocular pressure to control glaucoma progression when appropriately instilled. However, topical medication use has critical limitations, among them high rates of therapeutic nonadherence that increase the risk of disease progression, side effects that can be sight-threatening, suboptimal circadian efficacy effects, the need for complex regimens of multiple drugs administered multiple times per day, difficulty with self-administration of topical medications, and others. Despite these significant and well-documented limitations, topical medical therapy remains the established first-line treatment for primary open-angle glaucoma (POAG) and ocular hypertension (OHT) because no meaningful safe and effective alternative therapies existed.
Given the well-documented limitations of topical therapy, coupled with significant advances in alternate approaches to glaucoma management, reliance on a medication-first approach may no longer be necessary. The 21st century has seen significant development of innovative interventional glaucoma therapies that do not require topical self-dosing one or more times daily and that obviate patient adherence. These include sustained-release drug-delivery platforms, selective laser trabeculoplasty (SLT), and minimally invasive glaucoma surgeries (MIGS), all of which lower IOP and reduce or eliminate the need for chronic medical therapy. The widespread availability of these therapies, coupled with high-quality evidence supporting their efficacy and safety, prompt a reexamination of the traditional topical medication-first approach and consideration of a more proactive, targeted, and earlier patient-centric interventional approach to glaucoma management.

Limitations of Topical Medical Therapy

Nonadherence

Rates of nonadherence to glaucoma therapy range from 30% to 80% in published studies [913] and adherence worsens over time [14]. In turn, topical medication nonadherence increases the risk of glaucoma progression [9, 10, 14, 15], which in turn leads to more invasive interventions at increased risk and cost. Underlying causes and contributors to nonadherence are multiple and complex and have been extensively reviewed [1618]. Unfortunately, intentional nonadherence—purposefully not taking medications as prescribed—can be difficult to detect in the clinical setting since patients may take their medications only in the days leading up to a scheduled office visit. Unintentional nonadherence can often be more easily detected and remedied.
Common causes of unintentional nonadherence include cognitive factors, physical factors, regimen complexity, and difficulty with drop administration [19]. Cognitive factors contributing to glaucoma medication nonadherence can range from forgetfulness [20, 21] to dementia [22, 23], which are particularly relevant given the demographics of the glaucoma population. For example, Alzheimer’s disease is a leading cause of dementia in older individuals and is more common in glaucoma patients than in the general population [2426]. Physical factors may include hand pain and/or weakness attributable to arthritis [23] or tremor [27], both of which make self-dosing with topical ophthalmic therapy difficult [2830]. The complexity of the medical regimen also affects adherence, with nonadherence being more likely as regimen complexity increases [31]; this can affect a substantial proportion of patients (40–50%) who require a multi-medication regimen for disease control [3, 3236].
Difficulty with self-dosing can also adversely affect adherence. Correctly administering eye drops is a complex, multistep process involving hygiene, dexterity, proprioception, coordination, and visual acuity. Most patients never receive any training in proper instillation techniques and instead are left to learn on their own [37, 38]. Perhaps not surprisingly, most patients make one or more mistakes when self-dosing [37, 3941], which consequently increase the risk of glaucoma progression [42] as well as dosing-related adverse events [43, 44]. Examples of common mistakes during eye drop instillation include difficulty aiming the bottle over the eye [30], difficulty squeezing the bottle [30], contaminating the dropper bottle through contact with the lid or ocular surface [37, 38, 40, 41, 4548], and either dispensing more than one drop [40, 4750] or missing the eye entirely [38, 40, 45, 47].
Engaging newly diagnosed glaucoma patients to accept responsibility for self-administering topical medical treatment can be challenging. In its mild and moderate stages, glaucoma is typically asymptomatic. There are no bothersome symptoms that therapy relieves to motivate adherence. Often, the only symptoms that medically well-controlled glaucoma patients are likely to experience in their lifetimes are the side effects of topical medical therapy. Even if glaucoma therapy did provide the incentive of symptomatic relief or some other immediate tangible benefit, it is uncertain that adherence would improve: for example, only 33% of patients with cluster headache (also called suicide headache due to their intensity) adhere to preventive therapy [51], and nearly 30% of patients who have received a kidney transplant fail to adhere to their anti-rejection drug regimen even while knowing that such behavior increases the risk of organ rejection [52]. In fact, rather than providing motivation to adhere, topical medical therapy for glaucoma is associated with numerous safety and tolerability issues that provide substantial deterrence to adherence, as discussed below.
Therapeutic nonadherence in glaucoma increases the risk of disease progression [9, 10, 14, 15] and can lead to a vicious cycle. Nonadherence can lead to disease progression, prompting the addition of more medications, which can further worsen adherence, leading to more progression and the need for more invasive therapies such as incisional surgery. Glaucoma progression is not uncommon among treated patients but can be prevented with effective therapy. In the era before topical beta-blockers, the 20-year cumulative risk of progression from glaucoma diagnosis to blindness in at least one eye was 27% among treated glaucoma patients [53]. After the development of beta-blockers but before prostaglandin analogues, the 20-year risk of blindness in the same population dropped by half to 13.5% [54]. In another study, the 15-year risk of blindness in at least one eye was 14.6% and nonadherence with therapy was a significant risk factor [55]. In a Swedish study, the 10- and 20-year risks of blindness from treated glaucoma in at least one eye were 26.5 and 38.1%, respectively [56]; these higher estimates may be related to the high prevalence of exfoliation glaucoma in Sweden [57], which generally has a more severe course than POAG. These and other studies of progression to blindness in eyes with treated glaucoma [58] underscore the need for a new approach to glaucoma therapy.

Side Effects

Perhaps the most obvious contributor to nonadherence with topical medical therapy is that all medications have side effects, and these side effects are common. For example, while latanoprost is widely considered to be among the safest of topical glaucoma medications, the rate of non-serious ocular adverse events with latanoprost in its US phase 3 registry trial was 10% and the rate of ocular events not graded as adverse events (examples include burning/stinging, tearing, foreign body sensation, etc.) was 48% [59]. Thus, more than 50% of patients receiving latanoprost exhibited one or more unwanted ocular symptoms while on therapy. Additionally, in the United Kingdom Glaucoma Treatment Study (UKGTS), latanoprost only slowed visual field progression by 58% more than placebo, indicating that even first-line standard of care therapy has limitations [60].
In general, the side effects associated with topical glaucoma medications fall into one of three categories: those related to mild or transient discomfort or disturbance, those associated with cosmetic alterations, and those that are safety-related. Numerous topical medications are associated with transient bothersome side effects that may not represent true safety issues. For instance, dorzolamide and the dorzolamide-timolol fixed combination often sting upon instillation because they are formulated as acidic solutions (pH ~ 5.6 to 5.7) to solubilize the carbonic anhydrase inhibitor (CAI) component [61, 62]. The insoluble CAI brinzolamide, in contrast, is formulated as a suspension, avoiding the low-pH stinging but instead causing transient formulation-related blurring of vision that is severe enough to impair driving [63]. Both CAI options are known to cause taste perturbations. Other drugs have other characteristics that are similarly undesirable. For example, 18% of patients using brimonidine tartrate 0.2% will develop a type IV hypersensitivity allergy within 12 months that will cause significant irritation and require cessation of the medication [64]. While these events pose no health-related safety concerns, they can incentivize adverse behaviors such as skipping doses or immediate post-dose use of artificial tears (thereby washing out the active drug before optimal absorption) that reduce therapeutic efficacy.
Side effects that alter patients’ appearance can be more problematic. The prostaglandin analogues—the most commonly used class of topical glaucoma medications—are well known to cause numerous cosmetic alterations. These include common side effects such as conjunctival hyperemia as well as lengthening, thickening, and darkening of eye lashes; and less common effects such as growth of vellus hairs and hyperpigmentation of periorbital skin as well as permanent iris hyperpigmentation [65]. These undesirable cosmetic issues can be more obvious and more unacceptable to patients with unilateral use, rendering the drug class relatively contraindicated for just one eye. As these drugs became available, patients had to tolerate these cosmetic side effects to reap their therapeutic efficacy. New and emerging therapies offer freedom from these adverse cosmetic events. Glaucoma patients today do not have to choose between how they look and how they see.
Some side effects can represent true safety issues. In the first 7 years after the commercialization of topical timolol, before its systemic safety issues were fully recognized, the topical glaucoma drug was suspected as the cause of 32 deaths reported to the United States Food and Drug Administration (FDA) [66], and topical beta-blockers have a substantial list of contraindications related to their systemic side effects [67]. Topical medical therapy was also associated with a higher risk of cataract surgery in both the Ocular Hypertension Treatment Study (OHTS, versus no treatment) [68] and the Laser in Glaucoma and Ocular Hypertension Trial (LiGHT, versus SLT) [69]. Topical prostaglandin analogues have been associated with several safety issues, including intraocular inflammation [70, 71], cystoid macular edema [71], and periorbital fat atrophy [7274]. The latter is part of a syndrome called prostaglandin-associated periorbitopathy (PAP) characterized by dermatochalasia, blepharoptosis, deepening of orbital sulci, and flattening of lower eyelid fat pads [75]. PAP has also been associated with topical prostaglandin analogues commercialized for over-the-counter use as lash lengtheners [76]. Initially considered a cosmetic side effect, PAP has come to be considered a safety issue: its presence compromises outcomes of trabeculectomy [77] and affects surgical planning for oculoplastics procedures [78].
Perhaps the most impactful adverse effect of topical glaucoma therapy is its association with ocular surface toxicity. Multiple prospective studies have found that the prevalence of ocular surface disease (OSD) in patients using topical glaucoma medications ranges from 30% to 70% [7985] and is much higher than in the general population (5–30% [86]). Commercial formulations of topical glaucoma medications contain both active and inactive ingredients, and while the active ingredients can have adverse effects on ocular surface health, most of the damage to the ocular surface is attributable to inactive ingredients, specifically preservatives, most commonly the preservative benzalkonium chloride (BAK).
BAK is present in about 70% of all ophthalmic formulations [87, 88] and its cytotoxic effects on ocular surface cells have been well characterized [8991]. In laboratory and animal studies, BAK has been demonstrated to injure and/or reduce survival of corneal [9295], conjunctival [93, 96], trabecular meshwork (TM) [97, 98], and ciliary epithelial [97] cells, to promote ocular tissue inflammation [95, 99], to delay corneal wound healing [100], to induce corneal epithelial cell apoptosis [101], and to alter gene expression in TM cells through DNA fragmentation and oxidative stress [98]. The clinical manifestations of BAK-induced ocular surface toxicity are numerous and include symptoms such as pain/discomfort [102, 103] and tearing [103] as well as signs including increased ocular surface staining [104106], worsened Schirmer test scores [103, 105], and decreased tear break-up time [104, 107, 108]. Together these result in a higher prevalence of punctate keratitis [107, 109] and overall worsening of Ocular Surface Disease Index scores [104, 110]. These adverse events are dose-dependent, worsening with increasing exposure to BAK via multiple medications, multiple drops per day, and duration of therapy [7985]. BAK-related ocular toxicity is associated with diminished quality of life [111, 112]. Perhaps most insidiously of all, the ocular surface damage caused by long-term use of BAK-preserved medications reduces the success of subsequent glaucoma filtration surgery [88, 113, 114]. Negative effects of first-line therapy on the outcomes of subsequent interventions alone warrant a reevaluation of the current medications-first approach, as this could be a set-up for long-term treatment failure in some patients.

Efficacy

Topical medical therapy has demonstrated effectiveness in reducing or preventing progression in multiple landmark clinical trials [3, 5, 6]. However, topical medical therapy—even when increased to multiple medications to achieve target pressures—does not always prevent progression. In OHTS, 4.4% of topical medication-treated patients developed POAG within 5 years [3]. In the Collaborative Initial Glaucoma Treatment Study (CIGTS), 25% of topical medication-treated patients progressed within 8 years [6]. In the LiGHT trial, 27% of topical medication-treated patients progressed within 6 years [69]. Coupled with the population data on blindness risks among treated glaucoma patients described above, it is clear that a substantial proportion of glaucoma patients will require more than topical medical therapy for disease control in their lifetime.
As discussed above, nonadherence is one of the factors contributing to the failure of medical therapy to control glaucoma. As former Surgeon General C. Everett Koop, MD, once said, “Drugs don’t work in patients who don’t take them.” [115]. However, even topical medications that are taken as directed have shortcomings in terms of the IOP control they deliver. Many topical medications exhibit peak and trough efficacy related to their pharmacokinetics and dosing schedules. For example, topical timolol dosed twice daily exhibits a roughly 33% loss of effect between 1-h peak and 12-h trough [116]. In its phase 3 registry trials, brimonidine demonstrated an approximately 50% loss of effect from peak to trough with three-times-daily dosing [117]. Also, both timolol and brimonidine have been shown to lower IOP poorly or not at all in the nocturnal period [118, 119]. This is problematic given that IOP tends to be highest at night when blood pressure is at its lowest [120, 121], and the nocturnal period has been identified as a time when glaucoma progression is most likely to occur. These shortcomings of topical medical therapy may partially explain the blindness rates among treated patients described above.
An additional limitation of topical medication efficacy is that many patients require IOP reduction beyond the capacity of single-agent therapy. In OHTS, for example, only 60% of patients achieved target IOP (20% reduction from baseline) using a single medication at 5 years [3]. In CIGTS, only 56% of patients were still at target IOP (determined using a study-specific formula) using one medication only 3 months into the study [34]. In LiGHT, only 43% of medication-first eyes were still at target IOP (20–30% depending on diagnosis and stage of disease) using a single medication at 6 years [69]. Unfortunately, the need for multiple medications for IOP control exposes patients to additive side effect profiles and reduces adherence [31]. Furthermore, as the number of medications required for IOP control increases, the law of diminishing returns comes into play: the medication selected as fourth-line therapy is utilized late in the treatment cascade because its efficacy and/or safety is inferior to the three drugs used before it, and thus it is unlikely to deliver IOP control when better medications have failed to do so. In fact, multiple studies have shown that the addition of a second, third, or fourth medication typically results in only a 10–15% incremental IOP reduction [35, 122, 123], and the addition of a 3rd or 4th medication carries only a 14% chance of successfully controlling IOP after 12 months [35].

Cost

Topical glaucoma medications are expensive. In 2017, Medicare spent USD $1.09 billion on prostaglandin analogues alone [124]. This does not include medications from other drug classes, drugs covered by non-Medicare insurers, nor uncovered medications purchased out-of-pocket by patients. The overall cost to the US healthcare system attributable directly to glaucoma care exceeds USD $9 billion annually [125]. There are also indirect costs to medical therapy borne by physician, the patient, and the patient’s caregiver(s). Physician costs include the time spent educating patients about side effects, teaching proper drop instillation, and prescribing/refilling medications, none of which are billable services. Patient costs include the time spent instilling drops (which can be 5–10 min several times per day for patients on multiple medications), driving to/from the pharmacy to get the medications, and coordinating complex dosing regimens and medication availability. These time costs often also extend to caregivers who may invest time in assisting with daily topical medication dosing, retrieving medications from the pharmacy, and accompanying patients with transportation to doctor visits. The combined nonmedical out-of-pocket costs to patients and caregivers for expenses such as travel and lost wages has been estimated at approximately $100 per patient per year [126]. If insurers decline to cover a prescribed medication (a common occurrence with new medications not yet added to insurers’ formularies), there are additional time costs to both patients and physicians when patients call the office seeking prior authorizations or alternate medications. In addition, cost can affect adherence. For example, before the implementation of the Medicare Part D drug benefit, 8.2% of beneficiaries reported skipping doses of glaucoma medications due to cost; after implementation, this dropped to 2.8% [127]. All treatment options have associated costs. Emerging evidence suggests that more interventional approaches to glaucoma—such as SLT or MIGS—may be more cost-effective strategies for long-term glaucoma care [69, 128131].

Quality of Life

Quality of life is a complex construct defined by the World Health Organization as “an individual’s perception of their position in life in the context of the culture and value systems in which they live and in relation to their goals, expectations, standards and concerns.” [132]. Health-related quality of life is informed by perceived physical and mental health over time. The glaucoma disease itself affects quality of life both physically and psychologically. At the time of diagnosis, the fear of blindness has a significant detrimental effect on quality of life that dissipates over time as patients become acclimated to and educated about the disease and its prognosis [133]. As glaucoma progresses, deterioration of the visual field decreases quality of life by imposing a series of daily life challenges, including difficulty driving, reading, and recognizing faces [134]. Recent evidence suggests early impairment of vision-related quality of life may manifest even before measurable visual field loss has occurred [135].
The side effects of topical medical treatment for glaucoma can also impact quality of life, including both physical and psychological well-being. Tearing, redness, and burning/stinging associated with topical glaucoma therapy have been shown to decrease quality of life [136]. The development of ocular surface disease from topical glaucoma medications—occurring in 30–70% of patients [7985]—has also been demonstrated to reduce quality of life [111, 112]. Difficulty self-administering drops also decreases quality of life [137]. Additionally, the treatment burden includes the time to instill drops—which can take 5–10 min multiple times per day if spaced correctly for patients on multiple medications—and the need for dosing away from the privacy of home (at work, at social events, etc.), all of which can affect treatment satisfaction and quality of life.
It is therefore surprising that well-designed studies evaluating quality of life have shown mixed outcomes. For example, in the pivotal trial of the trabecular micro-bypass iStent inject, greater reduction of medication dependence in the iStent inject group versus cataract surgery alone resulted in greater improvement in quality of life using several validated instruments [138]. In another study, improvement in quality of life after cataract surgery combined with various MIGS procedures was related to medication reductions [139, 140]. In another, combined cataract surgery with a trabecular micro-bypass significantly reduced topical medication use and improved ocular surface health, a key determinant of quality of life in glaucoma patients [111, 112]. In contrast, several of the landmark glaucoma trials—including OHTS [141], the Early Manifest Glaucoma Trial (EMGT) [142], the UKGTS [143], and the LiGHT study [69]—have not consistently shown significant adverse effects of topical medical therapy on quality of life.
It is unlikely that topical medical therapy for glaucoma has no adverse effect on quality of life. Rather, it is likely that the instruments used in these studies are not sensitive or specific enough to detect these differences. Most of the instruments used in these studies were designed to assess the effect of general health, or vision function, or the presence of glaucoma, on quality of life. In each of these studies, patients in all treatment arms were balanced by randomization in terms of diagnosis, visual function status, and general health status, so it is unsurprising that instruments designed to measure effects of these variables found no differences between the balanced treatment groups in these studies. Most instruments are not designed to measure the effects of glaucoma treatment on quality of life [144]. Given the interest in early use of sustained-release drug-delivery platforms, SLT, and minimally invasive glaucoma surgery to reduce or eliminate the need for topical medical therapy, there remains significant unmet need for an instrument that assesses the effect of glaucoma treatment on quality of life, as such data will inform clinicians and payors regarding the relative utility of these treatments in improving patient well-being, which will be essential for effecting paradigm change.

Discussion: Beyond Medications: A Modern Interventional Glaucoma Treatment Paradigm

There are considerable limitations of topical medical therapy outlined above, many of which can directly or indirectly increase the risk of disease progression and vision loss, and reduce patient quality of life. Given these limitations, now may be the appropriate time to reexamine the current conventional step-wise treatment approach, in which topical medications are often automatically and universally used as first-line therapy. By broadening the glaucoma treatment algorithm to include various less-invasive surgical interventions in addition to topical medications, doctors and patients are better equipped to design patient-centric treatment plans that span all disease severities and individual needs.
Indeed, the glaucoma treatment landscape has modernized and evolved, providing an opportunity to be more interventional in our approach–utilizing advanced therapies such as SLT, MIGS, and sustained-release drug-delivery systems—all with the common goal of preserving vision and improving patient quality of life. Although change can often be incremental and difficult to detect, there is growing evidence that the glaucoma treatment paradigm is evolving away from topical medications as first-line therapy. Following years of increasing rates of topical medication prescriptions for glaucoma in Australia [145], new data show prescription-writing on the decline in favor of SLT and MIGS procedures [146]. Likewise, based on data emerging from the LiGHT trial of first-line SLT versus topical medications for newly diagnosed glaucoma, the United Kingdom’s National Institute for Care and Health Excellence has recommended that SLT be the preferred first-line treatment for patients in the National Health Service [147]. Similarly, the European Glaucoma Society and the American Academy of Ophthalmology have recently updated their glaucoma management guidelines to recommend SLT as first-line therapy [8, 148]. Furthermore, a multicenter trial funded by the US National Institutes of Health/National Eye Institute—the Clarifying the Optimal Application of SLT Therapy (COAST) trial—is currently investigating the role of annual low-energy SLT compared to standard-energy SLT performed as needed in keeping newly diagnosed OAG and OHTN patients off medications for as long as possible [149]. From an economic perspective, compared to topical medical therapy, SLT has been shown to be more cost-effective [69, 130, 150152].
Similar to laser procedures, the advent of the MIGS family of glaucoma procedures has opened the door for earlier surgical intervention. Designed as safer and more physiologic alternatives to traditional trabeculectomy and tube-shunt procedures, these procedures collectively offer more modest IOP reductions for patients who would benefit from surgery but whose treatment goals may not justify the risk of traditional surgery [153155]. Accordingly, the notion of employing surgery earlier in the treatment cascade, rather than saving it as a last resort, has been raised [156, 157]. Given that the trabecular meshwork is the primary site of histopathological changes related to elevated IOP [158], targeting the meshwork early in the disease process is a logical approach, and several studies have suggested a beneficial disease-modifying effect of early intervention that may not be fully explained by IOP reduction alone [159, 160]. Various MIGS procedures combined with cataract surgery have been shown to be cost-effective compared to cataract surgery alone [128, 129, 161], and also compared to topical medication therapy. Relatedly, there have been calls to unbundle some glaucoma procedures from the often-required concomitant cataract surgery and allow them as standalone procedures so patients who might benefit have more than a one-moment-in-time opportunity to receive them at the time of cataract surgery [156].
Following upon SLT and MIGS, SRDD’s are the newest entry into the glaucoma treatment space. As a class they aim to provide the benefits of medication without the drawbacks of topical administration or reliance on patient adherence. SRDD’s include FDA-approved bimatoprost sustained-release (Durysta, Allergan), as well as several implants in various stages of development and with different routes of administration. These SRDD’s include, for example: ocular surface devices (e.g., gel-forming eyedrops, drug-delivery devices, medicated contact lenses, ocular ring inserts, collagen shields); punctal plug depots (e.g., travoprost ophthalmic insert, latanoprost punctal plug delivery system); subconjunctival injections (e.g., of latanoprost or dorzolamide-loaded polymer microparticles); and intracameral implants (providing sustained release of a medication such as travoprost, latanoprost, or omidenepag isopropyl [OMDI]). Although SRDD’s are the newest addition to the glaucoma treatment algorithm and many are still in development, they ultimately may offer additional topical-medication-free alternatives within the glaucoma treatment paradigm.
In summary, topical medications for glaucoma have a long history of effectiveness and safety, but newer treatment options may offer meaningful clinical benefits over topical medical therapy. The glaucoma treatment paradigm is in evolution, and novel interventional treatments—including sustained-release drug-delivery platforms, SLT, and MIGS procedures—are increasingly being considered earlier in the treatment cascade, including as first-line therapy. As data continue to emerge from clinical trials and clinical practice, the default topical medication-first approach to glaucoma care may be replaced by a more individualized, patient-centric approach that leverages the wider variety of interventional therapies now available—interventions that may be more effective, safer, and less adversely impactful on patients, doctors, and society at large.

Editorial Assistance

Editorial assistance (Dana M. Hornbeak, MD, MPH) was provided and funded by Glaukos Corporation.

Declarations

Conflict of Interest

Dr. Nathan Radcliffe: consultant/fees from Allergan, Alcon, Glaukos, Sight Science, New World Medical, Lumenis, Belkin, Bausch and Lomb, Thea, and Iridex. Dr. Manjool Shah: to be submitted separately. Dr. Thomas W. Samuelson: consultant for Aerie Pharmaceuticals, Alcon Surgical, Allergan, Bausch & Lomb/Valeant, BELKIN Vision, ELT Sight, Equinox, Glaukos, Imprimis, Irisvision, Ivantis, Johnson & Johnson Vision, Ocuphire, MicroOptx, New World Medical, PolyActiva, Ripple Therapeutics, Santen, Sight Science, TearClear, Vialase, and Zeiss Meditec; and stock options from Belkin Vision, Equinox, Ivantis, Ocuphire, Sight Science, Tear Clear, and Vialase.

Ethical Approval

This article is based on previously conducted studies and does not contain any new studies with human participants or animals performed by any of the authors.
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License, which permits any non-commercial use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by-nc/​4.​0/​.
Literatur
3.
Zurück zum Zitat Kass MA, Heuer DK, Higginbotham EJ, Johnson CA, Keltner JL, Miller JP, et al. The Ocular Hypertension Treatment Study: a randomized trial determines that topical ocular hypotensive medication delays or prevents the onset of primary open-angle glaucoma. Arch Ophthalmol. 2002;120(6):701–13.PubMedCrossRef Kass MA, Heuer DK, Higginbotham EJ, Johnson CA, Keltner JL, Miller JP, et al. The Ocular Hypertension Treatment Study: a randomized trial determines that topical ocular hypotensive medication delays or prevents the onset of primary open-angle glaucoma. Arch Ophthalmol. 2002;120(6):701–13.PubMedCrossRef
4.
Zurück zum Zitat Miglior S, Zeyen T, Pfeiffer N, Cunha-Vaz J, Torri V, Adamsons I. Results of the European Glaucoma Prevention Study. Ophthalmology. 2005;112(3):366–75.PubMedCrossRef Miglior S, Zeyen T, Pfeiffer N, Cunha-Vaz J, Torri V, Adamsons I. Results of the European Glaucoma Prevention Study. Ophthalmology. 2005;112(3):366–75.PubMedCrossRef
5.
Zurück zum Zitat Heijl A, Leske MC, Bengtsson B, Hyman L, Bengtsson B, Hussein M. Reduction of intraocular pressure and glaucoma progression: results from the Early Manifest Glaucoma Trial. Arch Ophthalmol. 2002;120(10):1268–79.PubMedCrossRef Heijl A, Leske MC, Bengtsson B, Hyman L, Bengtsson B, Hussein M. Reduction of intraocular pressure and glaucoma progression: results from the Early Manifest Glaucoma Trial. Arch Ophthalmol. 2002;120(10):1268–79.PubMedCrossRef
6.
Zurück zum Zitat Lichter PR, Musch DC, Gillespie BW, Guire KE, Janz NK, Wren PA, et al. Interim clinical outcomes in the Collaborative Initial Glaucoma Treatment Study comparing initial treatment randomized to medications or surgery. Ophthalmology. 2001;108(11):1943–53.PubMedCrossRef Lichter PR, Musch DC, Gillespie BW, Guire KE, Janz NK, Wren PA, et al. Interim clinical outcomes in the Collaborative Initial Glaucoma Treatment Study comparing initial treatment randomized to medications or surgery. Ophthalmology. 2001;108(11):1943–53.PubMedCrossRef
7.
Zurück zum Zitat Glaucoma Laser Trial Research Group. The glaucoma laser trial (GLT) and glaucoma laser trial follow-up study: 7. Results. Am J Ophthalmol. 1995;120(6):718–31.CrossRef Glaucoma Laser Trial Research Group. The glaucoma laser trial (GLT) and glaucoma laser trial follow-up study: 7. Results. Am J Ophthalmol. 1995;120(6):718–31.CrossRef
8.
Zurück zum Zitat American Academy of Ophthalmology. Primary open-angle glaucoma: preferred practice pattern. San Francisco: American Academy of Ophthalmology; 2020. American Academy of Ophthalmology. Primary open-angle glaucoma: preferred practice pattern. San Francisco: American Academy of Ophthalmology; 2020.
9.
Zurück zum Zitat Rossi GC, Pasinetti GM, Scudeller L, Radaelli R, Bianchi PE. Do adherence rates and glaucomatous visual field progression correlate? Eur J Ophthalmol. 2011;21(4):410–4.PubMedCrossRef Rossi GC, Pasinetti GM, Scudeller L, Radaelli R, Bianchi PE. Do adherence rates and glaucomatous visual field progression correlate? Eur J Ophthalmol. 2011;21(4):410–4.PubMedCrossRef
10.
Zurück zum Zitat Sleath B, Blalock S, Covert D, Stone JL, Skinner AC, Muir K, et al. The relationship between glaucoma medication adherence, eye drop technique, and visual field defect severity. Ophthalmology. 2011;118(12):2398–402.PubMedCrossRef Sleath B, Blalock S, Covert D, Stone JL, Skinner AC, Muir K, et al. The relationship between glaucoma medication adherence, eye drop technique, and visual field defect severity. Ophthalmology. 2011;118(12):2398–402.PubMedCrossRef
11.
Zurück zum Zitat Olthoff CM, Schouten JS, van de Borne BW, Webers CA. Noncompliance with ocular hypotensive treatment in patients with glaucoma or ocular hypertension an evidence-based review. Ophthalmology. 2005;112(6):953–61.PubMedCrossRef Olthoff CM, Schouten JS, van de Borne BW, Webers CA. Noncompliance with ocular hypotensive treatment in patients with glaucoma or ocular hypertension an evidence-based review. Ophthalmology. 2005;112(6):953–61.PubMedCrossRef
12.
Zurück zum Zitat Stewart WC, Chorak RP, Hunt HH, Sethuraman G. Factors associated with visual loss in patients with advanced glaucomatous changes in the optic nerve head. Am J Ophthalmol. 1993;116(2):176–81.PubMedCrossRef Stewart WC, Chorak RP, Hunt HH, Sethuraman G. Factors associated with visual loss in patients with advanced glaucomatous changes in the optic nerve head. Am J Ophthalmol. 1993;116(2):176–81.PubMedCrossRef
13.
Zurück zum Zitat Kass MA, Gordon M, Morley RE Jr, Meltzer DW, Goldberg JJ. Compliance with topical timolol treatment. Am J Ophthalmol. 1987;103(2):188–93.PubMedCrossRef Kass MA, Gordon M, Morley RE Jr, Meltzer DW, Goldberg JJ. Compliance with topical timolol treatment. Am J Ophthalmol. 1987;103(2):188–93.PubMedCrossRef
14.
Zurück zum Zitat Newman-Casey PA, Niziol LM, Gillespie BW, Janz NK, Lichter PR, Musch DC. The association between medication adherence and visual field progression in the collaborative initial glaucoma treatment study. Ophthalmology. 2020;127(4):477–83.PubMedCrossRef Newman-Casey PA, Niziol LM, Gillespie BW, Janz NK, Lichter PR, Musch DC. The association between medication adherence and visual field progression in the collaborative initial glaucoma treatment study. Ophthalmology. 2020;127(4):477–83.PubMedCrossRef
15.
Zurück zum Zitat Rajurkar K, Dubey S, Gupta PP, John D, Chauhan L. Compliance to topical anti-glaucoma medications among patients at a tertiary hospital in North India. J Curr Ophthalmol. 2018;30(2):125–9.PubMedPubMedCentralCrossRef Rajurkar K, Dubey S, Gupta PP, John D, Chauhan L. Compliance to topical anti-glaucoma medications among patients at a tertiary hospital in North India. J Curr Ophthalmol. 2018;30(2):125–9.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Tsai JC. A comprehensive perspective on patient adherence to topical glaucoma therapy. Ophthalmology. 2009;116(11 Suppl):S30–6.PubMedCrossRef Tsai JC. A comprehensive perspective on patient adherence to topical glaucoma therapy. Ophthalmology. 2009;116(11 Suppl):S30–6.PubMedCrossRef
17.
Zurück zum Zitat Tsai JC. Medication adherence in glaucoma: approaches for optimizing patient compliance. Curr Opin Ophthalmol. 2006;17(2):190–5.PubMed Tsai JC. Medication adherence in glaucoma: approaches for optimizing patient compliance. Curr Opin Ophthalmol. 2006;17(2):190–5.PubMed
18.
Zurück zum Zitat Budenz DL. A clinician’s guide to the assessment and management of nonadherence in glaucoma. Ophthalmology. 2009;116(11 Suppl):S43–7.PubMedCrossRef Budenz DL. A clinician’s guide to the assessment and management of nonadherence in glaucoma. Ophthalmology. 2009;116(11 Suppl):S43–7.PubMedCrossRef
19.
Zurück zum Zitat Hovanesian J, Singh IP, Bauskar A, Vantipalli S, Ozden RG, Goldstein MH. Identifying and addressing common contributors to nonadherence with ophthalmic medical therapy. Curr Opin Ophthalmol. 2023;34(Suppl 1):S1–13.PubMedCrossRef Hovanesian J, Singh IP, Bauskar A, Vantipalli S, Ozden RG, Goldstein MH. Identifying and addressing common contributors to nonadherence with ophthalmic medical therapy. Curr Opin Ophthalmol. 2023;34(Suppl 1):S1–13.PubMedCrossRef
20.
Zurück zum Zitat Poleon S, Racette L, Fifolt M, Schoenberger-Godwin YM, Abu SL, Twa MD. Patient and provider perspectives on glaucoma treatment adherence: a Delphi study in urban Alabama. Optom Vis Sci. 2021;98(9):1085–93.PubMedPubMedCentralCrossRef Poleon S, Racette L, Fifolt M, Schoenberger-Godwin YM, Abu SL, Twa MD. Patient and provider perspectives on glaucoma treatment adherence: a Delphi study in urban Alabama. Optom Vis Sci. 2021;98(9):1085–93.PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Newman-Casey PA, Robin AL, Blachley T, Farris K, Heisler M, Resnicow K, et al. The most common barriers to glaucoma medication adherence: a cross-sectional survey. Ophthalmology. 2015;122(7):1308–16.PubMedCrossRef Newman-Casey PA, Robin AL, Blachley T, Farris K, Heisler M, Resnicow K, et al. The most common barriers to glaucoma medication adherence: a cross-sectional survey. Ophthalmology. 2015;122(7):1308–16.PubMedCrossRef
22.
Zurück zum Zitat Yochim BP, Mueller AE, Kane KD, Kahook MY. Prevalence of cognitive impairment, depression, and anxiety symptoms among older adults with glaucoma. J Glaucoma. 2012;21(4):250–4.PubMedCrossRef Yochim BP, Mueller AE, Kane KD, Kahook MY. Prevalence of cognitive impairment, depression, and anxiety symptoms among older adults with glaucoma. J Glaucoma. 2012;21(4):250–4.PubMedCrossRef
23.
Zurück zum Zitat Asefzadeh B, Rett D, Pogoda TK, Selvin G, Cavallerano A. Glaucoma medication adherence in veterans and influence of coexisting chronic disease. J Glaucoma. 2014;23(4):240–5.PubMedCrossRef Asefzadeh B, Rett D, Pogoda TK, Selvin G, Cavallerano A. Glaucoma medication adherence in veterans and influence of coexisting chronic disease. J Glaucoma. 2014;23(4):240–5.PubMedCrossRef
24.
Zurück zum Zitat Rajan KB, Weuve J, Barnes LL, McAninch EA, Wilson RS, Evans DA. Population estimate of people with clinical Alzheimer’s disease and mild cognitive impairment in the United States (2020–2060). Alzheimers Dement. 2021;17:1966.PubMedPubMedCentralCrossRef Rajan KB, Weuve J, Barnes LL, McAninch EA, Wilson RS, Evans DA. Population estimate of people with clinical Alzheimer’s disease and mild cognitive impairment in the United States (2020–2060). Alzheimers Dement. 2021;17:1966.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Su CW, Lin CC, Kao CH, Chen HY. Association between glaucoma and the risk of dementia. Medicine (Baltimore). 2016;95(7): e2833.PubMedCrossRef Su CW, Lin CC, Kao CH, Chen HY. Association between glaucoma and the risk of dementia. Medicine (Baltimore). 2016;95(7): e2833.PubMedCrossRef
26.
Zurück zum Zitat Moon JY, Kim HJ, Park YH, Park TK, Park EC, Kim CY, et al. Association between open-angle glaucoma and the risks of Alzheimer’s and Parkinson’s diseases in South Korea: a 10-year nationwide cohort study. Sci Rep. 2018;8(1):11161.PubMedPubMedCentralCrossRef Moon JY, Kim HJ, Park YH, Park TK, Park EC, Kim CY, et al. Association between open-angle glaucoma and the risks of Alzheimer’s and Parkinson’s diseases in South Korea: a 10-year nationwide cohort study. Sci Rep. 2018;8(1):11161.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Handforth A, Parker GA. Conditions associated with essential tremor in veterans: a potential role for chronic stress. Tremor Other Hyperkinet Mov (N Y). 2018;8:517.PubMedCrossRef Handforth A, Parker GA. Conditions associated with essential tremor in veterans: a potential role for chronic stress. Tremor Other Hyperkinet Mov (N Y). 2018;8:517.PubMedCrossRef
28.
29.
Zurück zum Zitat Sayner R, Carpenter DM, Robin AL, Blalock SJ, Muir KW, Vitko M, et al. How glaucoma patient characteristics, self-efficacy and patient-provider communication are associated with eye drop technique. Int J Pharm Pract. 2016;24(2):78–85.PubMedCrossRef Sayner R, Carpenter DM, Robin AL, Blalock SJ, Muir KW, Vitko M, et al. How glaucoma patient characteristics, self-efficacy and patient-provider communication are associated with eye drop technique. Int J Pharm Pract. 2016;24(2):78–85.PubMedCrossRef
30.
Zurück zum Zitat Winfield AJ, Jessiman D, Williams A, Esakowitz L. A study of the causes of non-compliance by patients prescribed eyedrops. Br J Ophthalmol. 1990;74(8):477–80.PubMedPubMedCentralCrossRef Winfield AJ, Jessiman D, Williams A, Esakowitz L. A study of the causes of non-compliance by patients prescribed eyedrops. Br J Ophthalmol. 1990;74(8):477–80.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Robin AL, Covert D. Does adjunctive glaucoma therapy affect adherence to the initial primary therapy? Ophthalmology. 2005;112(5):863–8.PubMedCrossRef Robin AL, Covert D. Does adjunctive glaucoma therapy affect adherence to the initial primary therapy? Ophthalmology. 2005;112(5):863–8.PubMedCrossRef
32.
Zurück zum Zitat Gazzard G, Konstantakopoulou E, Garway-Heath D, Garg A, Vickerstaff V, Hunter R, 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):1505–16.PubMedPubMedCentralCrossRef Gazzard G, Konstantakopoulou E, Garway-Heath D, Garg A, Vickerstaff V, Hunter R, 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):1505–16.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Katz LJ, Steinmann WC, Kabir A, Molineaux J, Wizov SS, Marcellino G. Selective laser trabeculoplasty versus medical therapy as initial treatment of glaucoma: a prospective, randomized trial. J Glaucoma. 2012;21(7):460–8.PubMedCrossRef Katz LJ, Steinmann WC, Kabir A, Molineaux J, Wizov SS, Marcellino G. Selective laser trabeculoplasty versus medical therapy as initial treatment of glaucoma: a prospective, randomized trial. J Glaucoma. 2012;21(7):460–8.PubMedCrossRef
34.
Zurück zum Zitat Musch DC, Niziol LM, Gillespie BW, Lichter PR. Stepped medication use, associated symptoms, and treatment failure among participants in the medication arm of the collaborative initial glaucoma treatment study. ARVO; April 28–May 3, 2018; Honolulu, HA2018. Musch DC, Niziol LM, Gillespie BW, Lichter PR. Stepped medication use, associated symptoms, and treatment failure among participants in the medication arm of the collaborative initial glaucoma treatment study. ARVO; April 28–May 3, 2018; Honolulu, HA2018.
35.
Zurück zum Zitat Neelakantan A, Vaishnav HD, Iyer SA, Sherwood MB. Is addition of a third or fourth antiglaucoma medication effective? J Glaucoma. 2004;13(2):130–6.PubMedCrossRef Neelakantan A, Vaishnav HD, Iyer SA, Sherwood MB. Is addition of a third or fourth antiglaucoma medication effective? J Glaucoma. 2004;13(2):130–6.PubMedCrossRef
36.
Zurück zum Zitat Saini SD, Schoenfeld P, Kaulback K, Dubinsky MC. Effect of medication dosing frequency on adherence in chronic diseases. Am J Manag Care. 2009;15(6):e22-33.PubMed Saini SD, Schoenfeld P, Kaulback K, Dubinsky MC. Effect of medication dosing frequency on adherence in chronic diseases. Am J Manag Care. 2009;15(6):e22-33.PubMed
37.
Zurück zum Zitat Usgaonkar U, Zambaulicar V, Shetty A. Subjective and objective assessment of the eye drop instillation technique: a hospital-based cross-sectional study. Indian J Ophthalmol. 2021;69(10):2638–42.PubMedPubMedCentralCrossRef Usgaonkar U, Zambaulicar V, Shetty A. Subjective and objective assessment of the eye drop instillation technique: a hospital-based cross-sectional study. Indian J Ophthalmol. 2021;69(10):2638–42.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Tatham AJ, Sarodia U, Gatrad F, Awan A. Eye drop instillation technique in patients with glaucoma. Eye (Lond). 2013;27(11):1293–8.PubMedCrossRef Tatham AJ, Sarodia U, Gatrad F, Awan A. Eye drop instillation technique in patients with glaucoma. Eye (Lond). 2013;27(11):1293–8.PubMedCrossRef
39.
Zurück zum Zitat Stone JL, Robin AL, Novack GD, Covert DW, Cagle GD. An objective evaluation of eyedrop instillation in patients with glaucoma. Arch Ophthalmol. 2009;127(6):732–6.PubMedCrossRef Stone JL, Robin AL, Novack GD, Covert DW, Cagle GD. An objective evaluation of eyedrop instillation in patients with glaucoma. Arch Ophthalmol. 2009;127(6):732–6.PubMedCrossRef
40.
Zurück zum Zitat Gomes BF, Paredes AF, Madeira N, Moraes HV Jr, Santhiago MR. Assessment of eye drop instillation technique in glaucoma patients. Arq Bras Oftalmol. 2017;80(4):238–41.PubMedCrossRef Gomes BF, Paredes AF, Madeira N, Moraes HV Jr, Santhiago MR. Assessment of eye drop instillation technique in glaucoma patients. Arq Bras Oftalmol. 2017;80(4):238–41.PubMedCrossRef
41.
Zurück zum Zitat Gupta R, Patil B, Shah BM, Bali SJ, Mishra SK, Dada T. Evaluating eye drop instillation technique in glaucoma patients. J Glaucoma. 2012;21(3):189–92.PubMedCrossRef Gupta R, Patil B, Shah BM, Bali SJ, Mishra SK, Dada T. Evaluating eye drop instillation technique in glaucoma patients. J Glaucoma. 2012;21(3):189–92.PubMedCrossRef
42.
Zurück zum Zitat Rajanala AP, Prager AJ, Park MS, Tanna AP. Association of the effectiveness of eye drop self-instillation and glaucoma progression. J Glaucoma. 2022;31:156.PubMedCrossRef Rajanala AP, Prager AJ, Park MS, Tanna AP. Association of the effectiveness of eye drop self-instillation and glaucoma progression. J Glaucoma. 2022;31:156.PubMedCrossRef
43.
Zurück zum Zitat Dietlein TS, Jordan JF, Luke C, Schild A, Dinslage S, Krieglstein GK. Self-application of single-use eyedrop containers in an elderly population: comparisons with standard eyedrop bottle and with younger patients. Acta Ophthalmol. 2008;86(8):856–9.PubMedCrossRef Dietlein TS, Jordan JF, Luke C, Schild A, Dinslage S, Krieglstein GK. Self-application of single-use eyedrop containers in an elderly population: comparisons with standard eyedrop bottle and with younger patients. Acta Ophthalmol. 2008;86(8):856–9.PubMedCrossRef
44.
Zurück zum Zitat Taban M, Sarayba MA, Ignacio TS, Behrens A, McDonnell PJ. Ingress of India ink into the anterior chamber through sutureless clear corneal cataract wounds. Arch Ophthalmol. 2005;123(5):643–8.PubMedCrossRef Taban M, Sarayba MA, Ignacio TS, Behrens A, McDonnell PJ. Ingress of India ink into the anterior chamber through sutureless clear corneal cataract wounds. Arch Ophthalmol. 2005;123(5):643–8.PubMedCrossRef
45.
Zurück zum Zitat Schwartz GF, Hollander DA, Williams JM. Evaluation of eye drop administration technique in patients with glaucoma or ocular hypertension. Curr Med Res Opin. 2013;29(11):1515–22.PubMedCrossRef Schwartz GF, Hollander DA, Williams JM. Evaluation of eye drop administration technique in patients with glaucoma or ocular hypertension. Curr Med Res Opin. 2013;29(11):1515–22.PubMedCrossRef
46.
Zurück zum Zitat Naito T, Yoshikawa K, Namiguchi K, Mizoue S, Shiraishi A, Ichikawa Y, et al. Comparison of success rates in eye drop instillation between sitting position and supine position. PLoS ONE. 2018;13(9): e0204363.PubMedPubMedCentralCrossRef Naito T, Yoshikawa K, Namiguchi K, Mizoue S, Shiraishi A, Ichikawa Y, et al. Comparison of success rates in eye drop instillation between sitting position and supine position. PLoS ONE. 2018;13(9): e0204363.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Kashiwagi K, Matsuda Y, Ito Y, Kawate H, Sakamoto M, Obi S, et al. Investigation of visual and physical factors associated with inadequate instillation of eyedrops among patients with glaucoma. PLoS ONE. 2021;16(5): e0251699.PubMedPubMedCentralCrossRef Kashiwagi K, Matsuda Y, Ito Y, Kawate H, Sakamoto M, Obi S, et al. Investigation of visual and physical factors associated with inadequate instillation of eyedrops among patients with glaucoma. PLoS ONE. 2021;16(5): e0251699.PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Lazcano-Gomez G, Castillejos A, Kahook M, Jimenez-Roman J, Gonzalez-Salinas R. Videographic assessment of glaucoma drop instillation. J Curr Glaucoma Pract. 2015;9(2):47–50.PubMedPubMedCentralCrossRef Lazcano-Gomez G, Castillejos A, Kahook M, Jimenez-Roman J, Gonzalez-Salinas R. Videographic assessment of glaucoma drop instillation. J Curr Glaucoma Pract. 2015;9(2):47–50.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Liu Y, Murdoch A, Bassett K, Dharamsi S. Proficiency of eye drop instillation in postoperative cataract patients in Ghana. Clin Ophthalmol (Auckland, NZ). 2013;7:2099–105. Liu Y, Murdoch A, Bassett K, Dharamsi S. Proficiency of eye drop instillation in postoperative cataract patients in Ghana. Clin Ophthalmol (Auckland, NZ). 2013;7:2099–105.
50.
Zurück zum Zitat An JA, Kasner O, Samek DA, Levesque V. Evaluation of eyedrop administration by inexperienced patients after cataract surgery. J Cataract Refract Surg. 2014;40(11):1857–61.PubMedCrossRef An JA, Kasner O, Samek DA, Levesque V. Evaluation of eyedrop administration by inexperienced patients after cataract surgery. J Cataract Refract Surg. 2014;40(11):1857–61.PubMedCrossRef
51.
Zurück zum Zitat Rimmele F, Muller B, Becker-Hingst N, Wegener S, Rimmele S, Kropp P, et al. Medication adherence in patients with cluster headache and migraine: an online survey. Sci Rep. 2023;13(1):4546.PubMedPubMedCentralCrossRef Rimmele F, Muller B, Becker-Hingst N, Wegener S, Rimmele S, Kropp P, et al. Medication adherence in patients with cluster headache and migraine: an online survey. Sci Rep. 2023;13(1):4546.PubMedPubMedCentralCrossRef
52.
Zurück zum Zitat Denhaerynck K, Dobbels F, Cleemput I, Desmyttere A, Schafer-Keller P, Schaub S, et al. Prevalence, consequences, and determinants of nonadherence in adult renal transplant patients: a literature review. Transpl Int. 2005;18(10):1121–33.PubMedCrossRef Denhaerynck K, Dobbels F, Cleemput I, Desmyttere A, Schafer-Keller P, Schaub S, et al. Prevalence, consequences, and determinants of nonadherence in adult renal transplant patients: a literature review. Transpl Int. 2005;18(10):1121–33.PubMedCrossRef
53.
Zurück zum Zitat Hattenhauer MG, Johnson DH, Ing HH, Herman DC, Hodge DO, Yawn BP, et al. The probability of blindness from open-angle glaucoma. Ophthalmology. 1998;105(11):2099–104.PubMedCrossRef Hattenhauer MG, Johnson DH, Ing HH, Herman DC, Hodge DO, Yawn BP, et al. The probability of blindness from open-angle glaucoma. Ophthalmology. 1998;105(11):2099–104.PubMedCrossRef
54.
Zurück zum Zitat Malihi M, Moura Filho ER, Hodge DO, Sit AJ. Long-term trends in glaucoma-related blindness in Olmsted County, Minnesota. Ophthalmology. 2014;121(1):134–41.PubMedCrossRef Malihi M, Moura Filho ER, Hodge DO, Sit AJ. Long-term trends in glaucoma-related blindness in Olmsted County, Minnesota. Ophthalmology. 2014;121(1):134–41.PubMedCrossRef
55.
Zurück zum Zitat Chen PP. Blindness in patients with treated open-angle glaucoma. Ophthalmology. 2003;110(4):726–33. Chen PP. Blindness in patients with treated open-angle glaucoma. Ophthalmology. 2003;110(4):726–33.
56.
Zurück zum Zitat Peters D, Bengtsson B, Heijl A. Lifetime risk of blindness in open-angle glaucoma. Am J Ophthalmol. 2013;156(4):724–30.PubMedCrossRef Peters D, Bengtsson B, Heijl A. Lifetime risk of blindness in open-angle glaucoma. Am J Ophthalmol. 2013;156(4):724–30.PubMedCrossRef
57.
Zurück zum Zitat Ekstrom C. Incidence of open-angle glaucoma in central Sweden. Acta Ophthalmol. 2008;86(7):747–54.PubMedCrossRef Ekstrom C. Incidence of open-angle glaucoma in central Sweden. Acta Ophthalmol. 2008;86(7):747–54.PubMedCrossRef
58.
Zurück zum Zitat Mokhles P, Schouten JS, Beckers HJ, Azuara-Blanco A, Tuulonen A, Webers CA. A systematic review of end-of-life visual impairment in open-angle glaucoma: an epidemiological autopsy. J Glaucoma. 2016;25(7):623–8.PubMedCrossRef Mokhles P, Schouten JS, Beckers HJ, Azuara-Blanco A, Tuulonen A, Webers CA. A systematic review of end-of-life visual impairment in open-angle glaucoma: an epidemiological autopsy. J Glaucoma. 2016;25(7):623–8.PubMedCrossRef
59.
Zurück zum Zitat Camras CB. Comparison of latanoprost and timolol in patients with ocular hypertension and glaucoma: a six-month masked, multicenter trial in the United States. The United States Latanoprost Study Group. Ophthalmology. 1996;103(1):138–47.PubMedCrossRef Camras CB. Comparison of latanoprost and timolol in patients with ocular hypertension and glaucoma: a six-month masked, multicenter trial in the United States. The United States Latanoprost Study Group. Ophthalmology. 1996;103(1):138–47.PubMedCrossRef
60.
Zurück zum Zitat Founti P, Bunce C, Khawaja AP, Dore CJ, Mohamed-Noriega J, Garway-Heath DF, et al. Risk factors for visual field deterioration in the United Kingdom glaucoma treatment study. Ophthalmology. 2020;127(12):1642–51.PubMedCrossRef Founti P, Bunce C, Khawaja AP, Dore CJ, Mohamed-Noriega J, Garway-Heath DF, et al. Risk factors for visual field deterioration in the United Kingdom glaucoma treatment study. Ophthalmology. 2020;127(12):1642–51.PubMedCrossRef
61.
Zurück zum Zitat Merck & Co., Inc. TRUSOPT® (dorzolamide hydrochloride ophthalmic solution) 2%. Prescribing Information. Whitehouse Station: Merck & Co., Inc. ; 2014. Merck & Co., Inc. TRUSOPT® (dorzolamide hydrochloride ophthalmic solution) 2%. Prescribing Information. Whitehouse Station: Merck & Co., Inc. ; 2014.
62.
Zurück zum Zitat Merck & Co Inc. Cosopt (dorzolamide 2%/timolol 0.5% fixed combination) prescribing information. 2006. Merck & Co Inc. Cosopt (dorzolamide 2%/timolol 0.5% fixed combination) prescribing information. 2006.
63.
Zurück zum Zitat Alcon Laboratories, Inc. AZOPT® (brinzolamide ophthalmic suspension) 1%. Prescribing Information. Fort Worth: Alcon Laboratories, Inc. ; 2015. Alcon Laboratories, Inc. AZOPT® (brinzolamide ophthalmic suspension) 1%. Prescribing Information. Fort Worth: Alcon Laboratories, Inc. ; 2015.
64.
Zurück zum Zitat Motolko MA. Comparison of allergy rates in glaucoma patients receiving brimonidine 0.2% monotherapy versus fixed-combination brimonidine 0.2%-timolol 0.5% therapy. Curr Med Res Opin. 2008;24(9):2663–7.PubMedCrossRef Motolko MA. Comparison of allergy rates in glaucoma patients receiving brimonidine 0.2% monotherapy versus fixed-combination brimonidine 0.2%-timolol 0.5% therapy. Curr Med Res Opin. 2008;24(9):2663–7.PubMedCrossRef
65.
Zurück zum Zitat Hollo G. The side effects of the prostaglandin analogues. Expert Opin Drug Saf. 2007;6(1):45–52.PubMedCrossRef Hollo G. The side effects of the prostaglandin analogues. Expert Opin Drug Saf. 2007;6(1):45–52.PubMedCrossRef
66.
Zurück zum Zitat Nelson WL, Fraunfelder FT, Sills JM, Arrowsmith JB, Kuritsky JN. Adverse respiratory and cardiovascular events attributed to timolol ophthalmic solution, 1978–1985. Am J Ophthalmol. 1986;102(5):606–11.PubMedCrossRef Nelson WL, Fraunfelder FT, Sills JM, Arrowsmith JB, Kuritsky JN. Adverse respiratory and cardiovascular events attributed to timolol ophthalmic solution, 1978–1985. Am J Ophthalmol. 1986;102(5):606–11.PubMedCrossRef
67.
Zurück zum Zitat Bausch & Lomb, Inc. Timoptic 0.25% and 5% (timolol maleate ophthalmic solution). Prescribing Information. Bridgewater: Bausch & Lomb, Inc.; 2016. Bausch & Lomb, Inc. Timoptic 0.25% and 5% (timolol maleate ophthalmic solution). Prescribing Information. Bridgewater: Bausch & Lomb, Inc.; 2016.
68.
Zurück zum Zitat Herman DC, Gordon MO, Beiser JA, Chylack LT Jr, Lamping KA, Schein OD, et al. Topical ocular hypotensive medication and lens opacification: evidence from the ocular hypertension treatment study. Am J Ophthalmol. 2006;142(5):800–10.PubMedPubMedCentralCrossRef Herman DC, Gordon MO, Beiser JA, Chylack LT Jr, Lamping KA, Schein OD, et al. Topical ocular hypotensive medication and lens opacification: evidence from the ocular hypertension treatment study. Am J Ophthalmol. 2006;142(5):800–10.PubMedPubMedCentralCrossRef
69.
Zurück zum Zitat Gazzard G, Konstantakopoulou E, Garway-Heath D, Adeleke M, Vickerstaff V, Ambler G, et al. Laser in glaucoma and ocular hypertension (LiGHT) trial: six-year results of primary selective laser trabeculoplasty versus eye drops for the treatment of glaucoma and ocular hypertension. Ophthalmology. 2023;130(2):139–51.PubMedCrossRef Gazzard G, Konstantakopoulou E, Garway-Heath D, Adeleke M, Vickerstaff V, Ambler G, et al. Laser in glaucoma and ocular hypertension (LiGHT) trial: six-year results of primary selective laser trabeculoplasty versus eye drops for the treatment of glaucoma and ocular hypertension. Ophthalmology. 2023;130(2):139–51.PubMedCrossRef
70.
Zurück zum Zitat Fechtner RD, Khouri AS, Zimmerman TJ, Bullock J, Feldman R, Kulkarni P, et al. Anterior uveitis associated with latanoprost. Am J Ophthalmol. 1998;126(1):37–41.PubMedCrossRef Fechtner RD, Khouri AS, Zimmerman TJ, Bullock J, Feldman R, Kulkarni P, et al. Anterior uveitis associated with latanoprost. Am J Ophthalmol. 1998;126(1):37–41.PubMedCrossRef
71.
Zurück zum Zitat Warwar RE, Bullock JD, Ballal D. Cystoid macular edema and anterior uveitis associated with latanoprost use. Experience and incidence in a retrospective review of 94 patients. Ophthalmology. 1998;105(2):263–8.PubMedCrossRef Warwar RE, Bullock JD, Ballal D. Cystoid macular edema and anterior uveitis associated with latanoprost use. Experience and incidence in a retrospective review of 94 patients. Ophthalmology. 1998;105(2):263–8.PubMedCrossRef
72.
Zurück zum Zitat Peplinski LS, Albiani SK. Deepening of lid sulcus from topical bimatoprost therapy. Optom Vis Sci. 2004;81(8):574–7.PubMedCrossRef Peplinski LS, Albiani SK. Deepening of lid sulcus from topical bimatoprost therapy. Optom Vis Sci. 2004;81(8):574–7.PubMedCrossRef
73.
Zurück zum Zitat Tappeiner C, Perren B, Iliev ME, Frueh BE, Goldblum D. Orbital fat atrophy in glaucoma patients treated with topical bimatoprost—can bimatoprost cause enophthalmos? Klin Monbl Augenheilkd. 2008;225(5):443–5.PubMedCrossRef Tappeiner C, Perren B, Iliev ME, Frueh BE, Goldblum D. Orbital fat atrophy in glaucoma patients treated with topical bimatoprost—can bimatoprost cause enophthalmos? Klin Monbl Augenheilkd. 2008;225(5):443–5.PubMedCrossRef
74.
Zurück zum Zitat Filippopoulos T, Paula JS, Torun N, Hatton MP, Pasquale LR, Grosskreutz CL. Periorbital changes associated with topical bimatoprost. Ophthalm Plast Reconstr Surg. 2008;24(4):302–7.CrossRef Filippopoulos T, Paula JS, Torun N, Hatton MP, Pasquale LR, Grosskreutz CL. Periorbital changes associated with topical bimatoprost. Ophthalm Plast Reconstr Surg. 2008;24(4):302–7.CrossRef
75.
Zurück zum Zitat Tan J, Berke S. Latanoprost-induced prostaglandin-associated periorbitopathy. Optom Vis Sci. 2013;90(9):e245–7 (discussion 1029).PubMedCrossRef Tan J, Berke S. Latanoprost-induced prostaglandin-associated periorbitopathy. Optom Vis Sci. 2013;90(9):e245–7 (discussion 1029).PubMedCrossRef
76.
Zurück zum Zitat Jamison A, Okafor L, Ullrich K, Schiedler V, Malhotra R. Do prostaglandin analogue lash lengtheners cause eyelid fat and volume loss? Aesthet Surg J. 2022;42(11):1241–9.PubMedCrossRef Jamison A, Okafor L, Ullrich K, Schiedler V, Malhotra R. Do prostaglandin analogue lash lengtheners cause eyelid fat and volume loss? Aesthet Surg J. 2022;42(11):1241–9.PubMedCrossRef
77.
Zurück zum Zitat Ishida A, Miki T, Naito T, Ichioka S, Takayanagi Y, Tanito M. Surgical results of trabeculectomy among groups stratified by prostaglandin-associated periorbitopathy severity. Ophthalmology. 2023;130(3):297–303.PubMedCrossRef Ishida A, Miki T, Naito T, Ichioka S, Takayanagi Y, Tanito M. Surgical results of trabeculectomy among groups stratified by prostaglandin-associated periorbitopathy severity. Ophthalmology. 2023;130(3):297–303.PubMedCrossRef
78.
Zurück zum Zitat Tan P, Malhotra R. Oculoplastic considerations in patients with glaucoma. Surv Ophthalmol. 2016;61(6):718–25.PubMedCrossRef Tan P, Malhotra R. Oculoplastic considerations in patients with glaucoma. Surv Ophthalmol. 2016;61(6):718–25.PubMedCrossRef
79.
Zurück zum Zitat Labbe 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):994–9.PubMedCrossRef Labbe 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):994–9.PubMedCrossRef
80.
Zurück zum Zitat O’Hare F, Ghosh S, 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):675–81.PubMedCrossRef O’Hare F, Ghosh S, 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):675–81.PubMedCrossRef
81.
Zurück zum Zitat Valente C, Iester M, Corsi E, Rolando M. Symptoms and signs of tear film dysfunction in glaucomatous patients. J Ocul Pharmacol Ther. 2011;27(3):281–5.PubMedCrossRef Valente C, Iester M, Corsi E, Rolando M. Symptoms and signs of tear film dysfunction in glaucomatous patients. J Ocul Pharmacol Ther. 2011;27(3):281–5.PubMedCrossRef
82.
Zurück zum Zitat Leung EW, Medeiros FA, Weinreb RN. Prevalence of ocular surface disease in glaucoma patients. J Glaucoma. 2008;17(5):350–5.PubMedCrossRef Leung EW, Medeiros FA, Weinreb RN. Prevalence of ocular surface disease in glaucoma patients. J Glaucoma. 2008;17(5):350–5.PubMedCrossRef
83.
Zurück zum Zitat Fechtner RD, Godfrey DG, Budenz D, Stewart JA, Stewart WC, Jasek MC. Prevalence of ocular surface complaints in patients with glaucoma using topical intraocular pressure-lowering medications. Cornea. 2010;29(6):618–21.PubMedCrossRef Fechtner RD, Godfrey DG, Budenz D, Stewart JA, Stewart WC, Jasek MC. Prevalence of ocular surface complaints in patients with glaucoma using topical intraocular pressure-lowering medications. Cornea. 2010;29(6):618–21.PubMedCrossRef
84.
Zurück zum Zitat Rossi GC, 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.PubMedCrossRef Rossi GC, 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.PubMedCrossRef
85.
Zurück zum Zitat Skalicky SE, Goldberg I, McCluskey P. Ocular surface disease and quality of life in patients with glaucoma. Am J Ophthalmol. 2011. Skalicky SE, Goldberg I, McCluskey P. Ocular surface disease and quality of life in patients with glaucoma. Am J Ophthalmol. 2011.
86.
Zurück zum Zitat Stapleton F, Alves M, Bunya VY, Jalbert I, Lekhanont K, Malet F, et al. TFOS DEWS II epidemiology report. Ocul Surf. 2017;15(3):334–65.PubMedCrossRef Stapleton F, Alves M, Bunya VY, Jalbert I, Lekhanont K, Malet F, et al. TFOS DEWS II epidemiology report. Ocul Surf. 2017;15(3):334–65.PubMedCrossRef
87.
Zurück zum Zitat Freeman PD, Kahook MY. Preservatives in topical ophthalmic medications: historical and clinical perspectives. Expert Rev Ophthalmol. 2009;4:59–64.CrossRef Freeman PD, Kahook MY. Preservatives in topical ophthalmic medications: historical and clinical perspectives. Expert Rev Ophthalmol. 2009;4:59–64.CrossRef
88.
Zurück zum Zitat 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):1446–54.PubMedCrossRef 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):1446–54.PubMedCrossRef
89.
Zurück zum Zitat Goldstein MH, Silva FQ, Blender N, Tran T, Vantipalli S. Ocular benzalkonium chloride exposure: problems and solutions. Eye (Lond). 2022;36(2):361–8.PubMedCrossRef Goldstein MH, Silva FQ, Blender N, Tran T, Vantipalli S. Ocular benzalkonium chloride exposure: problems and solutions. Eye (Lond). 2022;36(2):361–8.PubMedCrossRef
90.
Zurück zum Zitat Baudouin C. Side effects of antiglaucomatous drugs on the ocular surface. Curr Opin Ophthalmol. 1996;7(2):80–6.PubMedCrossRef Baudouin C. Side effects of antiglaucomatous drugs on the ocular surface. Curr Opin Ophthalmol. 1996;7(2):80–6.PubMedCrossRef
91.
Zurück zum Zitat Baudouin C, Labbe 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):312–34.PubMedCrossRef Baudouin C, Labbe 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):312–34.PubMedCrossRef
92.
Zurück zum Zitat Kim JH, Kim EJ, Kim YH, Kim YI, Lee SH, Jung JC, et al. In vivo effects of preservative-free and preserved prostaglandin analogs: mouse ocular surface study. Korean J Ophthalmol. 2015;29(4):270–9.PubMedPubMedCentralCrossRef Kim JH, Kim EJ, Kim YH, Kim YI, Lee SH, Jung JC, et al. In vivo effects of preservative-free and preserved prostaglandin analogs: mouse ocular surface study. Korean J Ophthalmol. 2015;29(4):270–9.PubMedPubMedCentralCrossRef
93.
Zurück zum Zitat Ayaki M, Iwasawa A. Cytotoxicity of prostaglandin analog eye drops preserved with benzalkonium chloride in multiple corneoconjunctival cell lines. Clin Ophthalmol (Auckland, NZ). 2010;4:919–24.CrossRef Ayaki M, Iwasawa A. Cytotoxicity of prostaglandin analog eye drops preserved with benzalkonium chloride in multiple corneoconjunctival cell lines. Clin Ophthalmol (Auckland, NZ). 2010;4:919–24.CrossRef
94.
Zurück zum Zitat Guzman-Aranguez A, Calvo P, Ropero I, Pintor J. In vitro effects of preserved and unpreserved anti-allergic drugs on human corneal epithelial cells. J Ocul Pharmacol Ther. 2014;30(9):790–8.PubMedPubMedCentralCrossRef Guzman-Aranguez A, Calvo P, Ropero I, Pintor J. In vitro effects of preserved and unpreserved anti-allergic drugs on human corneal epithelial cells. J Ocul Pharmacol Ther. 2014;30(9):790–8.PubMedPubMedCentralCrossRef
95.
Zurück zum Zitat Kahook MY, Noecker RJ. Comparison of corneal and conjunctival changes after dosing of travoprost preserved with sofZia, latanoprost with 0.02% benzalkonium chloride, and preservative-free artificial tears. Cornea. 2008;27(3):339–43.PubMedCrossRef Kahook MY, Noecker RJ. Comparison of corneal and conjunctival changes after dosing of travoprost preserved with sofZia, latanoprost with 0.02% benzalkonium chloride, and preservative-free artificial tears. Cornea. 2008;27(3):339–43.PubMedCrossRef
96.
Zurück zum Zitat Ammar DA, Noecker RJ, Kahook MY. Effects of benzalkonium chloride-preserved, polyquad-preserved, and sofZia-preserved topical glaucoma medications on human ocular epithelial cells. Adv Ther. 2010;27(11):837–45.PubMedCrossRef Ammar DA, Noecker RJ, Kahook MY. Effects of benzalkonium chloride-preserved, polyquad-preserved, and sofZia-preserved topical glaucoma medications on human ocular epithelial cells. Adv Ther. 2010;27(11):837–45.PubMedCrossRef
97.
Zurück zum Zitat Ammar DA, Kahook MY. Effects of glaucoma medications and preservatives on cultured human trabecular meshwork and non-pigmented ciliary epithelial cell lines. Br J Ophthalmol. 2011;95(10):1466–9.PubMedCrossRef Ammar DA, Kahook MY. Effects of glaucoma medications and preservatives on cultured human trabecular meshwork and non-pigmented ciliary epithelial cell lines. Br J Ophthalmol. 2011;95(10):1466–9.PubMedCrossRef
98.
Zurück zum Zitat Izzotti A, La Maestra S, Micale RT, Longobardi MG, Sacca SC. Genomic and post-genomic effects of anti-glaucoma drugs preservatives in trabecular meshwork. Mutat Res. 2015;772:1–9.PubMedCrossRef Izzotti A, La Maestra S, Micale RT, Longobardi MG, Sacca SC. Genomic and post-genomic effects of anti-glaucoma drugs preservatives in trabecular meshwork. Mutat Res. 2015;772:1–9.PubMedCrossRef
99.
Zurück zum Zitat Liang H, Brignole-Baudouin F, Riancho L, Baudouin C. Reduced in vivo ocular surface toxicity with polyquad-preserved travoprost versus benzalkonium-preserved travoprost or latanoprost ophthalmic solutions. Ophthalm Res. 2012;48(2):89–101.CrossRef Liang H, Brignole-Baudouin F, Riancho L, Baudouin C. Reduced in vivo ocular surface toxicity with polyquad-preserved travoprost versus benzalkonium-preserved travoprost or latanoprost ophthalmic solutions. Ophthalm Res. 2012;48(2):89–101.CrossRef
100.
Zurück zum Zitat Nagai N, Murao T, Okamoto N, Ito Y. Comparison of corneal wound healing rates after instillation of commercially available latanoprost and travoprost in rat debrided corneal epithelium. J Oleo Sci. 2010;59(3):135–41.PubMedCrossRef Nagai N, Murao T, Okamoto N, Ito Y. Comparison of corneal wound healing rates after instillation of commercially available latanoprost and travoprost in rat debrided corneal epithelium. J Oleo Sci. 2010;59(3):135–41.PubMedCrossRef
101.
Zurück zum Zitat Pauly A, Brasnu E, Riancho L, Brignole-Baudouin F, Baudouin C. Multiple endpoint analysis of BAC-preserved and unpreserved antiallergic eye drops on a 3D-reconstituted corneal epithelial model. Mol Vis. 2011;17:745–55.PubMedPubMedCentral Pauly A, Brasnu E, Riancho L, Brignole-Baudouin F, Baudouin C. Multiple endpoint analysis of BAC-preserved and unpreserved antiallergic eye drops on a 3D-reconstituted corneal epithelial model. Mol Vis. 2011;17:745–55.PubMedPubMedCentral
102.
Zurück zum Zitat Jaenen N, Baudouin C, Pouliquen P, Manni G, Figueiredo A, Zeyen T. Ocular symptoms and signs with preserved and preservative-free glaucoma medications. Eur J Ophthalmol. 2007;17(3):341–9.PubMedCrossRef Jaenen N, Baudouin C, Pouliquen P, Manni G, Figueiredo A, Zeyen T. Ocular symptoms and signs with preserved and preservative-free glaucoma medications. Eur J Ophthalmol. 2007;17(3):341–9.PubMedCrossRef
103.
Zurück zum Zitat 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 (Auckland, NZ). 2016;10:445–54.CrossRef 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 (Auckland, NZ). 2016;10:445–54.CrossRef
104.
Zurück zum Zitat Horsley MB, Kahook MY. Effects of prostaglandin analog therapy on the ocular surface of glaucoma patients. Clin Ophthalmol (Auckland, NZ). 2009;3:291–5. Horsley MB, Kahook MY. Effects of prostaglandin analog therapy on the ocular surface of glaucoma patients. Clin Ophthalmol (Auckland, NZ). 2009;3:291–5.
105.
Zurück zum Zitat Hommer A, Kimmich F. Switching patients from preserved prostaglandin-analog monotherapy to preservative-free tafluprost. Clin Ophthalmol (Auckland, NZ). 2011;5:623–31. Hommer A, Kimmich F. Switching patients from preserved prostaglandin-analog monotherapy to preservative-free tafluprost. Clin Ophthalmol (Auckland, NZ). 2011;5:623–31.
106.
Zurück zum Zitat Lopes NLV, Gracitelli CPB, Chalita MR, de Faria NVL. Ocular surface evaluation after the substitution of benzalkonium chloride preserved prostaglandin eye drops by a preservative-free prostaglandin analogue. Med Hypoth Discov Innov Ophthalmol. 2019;8(1):52–6. Lopes NLV, Gracitelli CPB, Chalita MR, de Faria NVL. Ocular surface evaluation after the substitution of benzalkonium chloride preserved prostaglandin eye drops by a preservative-free prostaglandin analogue. Med Hypoth Discov Innov Ophthalmol. 2019;8(1):52–6.
107.
Zurück zum Zitat Rossi GC, Scudeller L, Rolle T, Pasinetti GM, Bianchi PE. From benzalkonium chloride-preserved Latanoprost to Polyquad-preserved Travoprost: a 6-month study on ocular surface safety and tolerability. Expert Opin Drug Saf. 2015;14(5):619–23.PubMedCrossRef Rossi GC, Scudeller L, Rolle T, Pasinetti GM, Bianchi PE. From benzalkonium chloride-preserved Latanoprost to Polyquad-preserved Travoprost: a 6-month study on ocular surface safety and tolerability. Expert Opin Drug Saf. 2015;14(5):619–23.PubMedCrossRef
108.
Zurück zum Zitat Tomic M, Kastelan S, Soldo KM, Salopek-Rabatic 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.PubMedPubMedCentralCrossRef Tomic M, Kastelan S, Soldo KM, Salopek-Rabatic 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.PubMedPubMedCentralCrossRef
109.
Zurück zum Zitat Aihara M, Ikeda Y, Mizoue S, Arakaki Y, Kita N, Kobayashi S, et al. Effect of switching to travoprost preserved with SofZia in glaucoma patients with chronic superficial punctate keratitis while receiving BAK-preserved latanoprost. J Glaucoma. 2016;25(6):e610–4.PubMedCrossRef Aihara M, Ikeda Y, Mizoue S, Arakaki Y, Kita N, Kobayashi S, et al. Effect of switching to travoprost preserved with SofZia in glaucoma patients with chronic superficial punctate keratitis while receiving BAK-preserved latanoprost. J Glaucoma. 2016;25(6):e610–4.PubMedCrossRef
110.
Zurück zum Zitat 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
111.
Zurück zum Zitat Rossi GC, Tinelli C, Pasinetti GM, Milano G, Bianchi PE. Dry eye syndrome-related quality of life in glaucoma patients. Eur J Ophthalmol. 2009;19(4):572–9.PubMedCrossRef Rossi GC, Tinelli C, Pasinetti GM, Milano G, Bianchi PE. Dry eye syndrome-related quality of life in glaucoma patients. Eur J Ophthalmol. 2009;19(4):572–9.PubMedCrossRef
112.
Zurück zum Zitat Rossi GC, 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):390–4.PubMedCrossRef Rossi GC, 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):390–4.PubMedCrossRef
113.
Zurück zum Zitat Chamard C, Larrieu S, Baudouin C, Bron A, Villain M, Daien V. Preservative-free versus preserved glaucoma eye drops and occurrence of glaucoma surgery. A retrospective study based on the French national health insurance information system, 2008–2016. Acta Ophthalmol. 2020;98(7):e876–81.PubMedCrossRef Chamard C, Larrieu S, Baudouin C, Bron A, Villain M, Daien V. Preservative-free versus preserved glaucoma eye drops and occurrence of glaucoma surgery. A retrospective study based on the French national health insurance information system, 2008–2016. Acta Ophthalmol. 2020;98(7):e876–81.PubMedCrossRef
114.
Zurück zum Zitat Boimer C, Birt CM. Preservative exposure and surgical outcomes in glaucoma patients: the PESO study. J Glaucoma. 2013;22(9):730–5.PubMedCrossRef Boimer C, Birt CM. Preservative exposure and surgical outcomes in glaucoma patients: the PESO study. J Glaucoma. 2013;22(9):730–5.PubMedCrossRef
115.
Zurück zum Zitat Lindenfeld J, Jessup M. “Drugs don’t work in patients who don’t take them” (C. Everett Koop, MD, US Surgeon General, 1985). Eur J Heart Fail. 2017;19(11):1412–3.PubMedCrossRef Lindenfeld J, Jessup M. “Drugs don’t work in patients who don’t take them” (C. Everett Koop, MD, US Surgeon General, 1985). Eur J Heart Fail. 2017;19(11):1412–3.PubMedCrossRef
116.
Zurück zum Zitat Zimmerman TJ, Kass MA, Yablonski ME, Becker B. Timolol maleate: efficacy and safety. Arch Ophthalmol. 1979;97(4):656–8.PubMedCrossRef Zimmerman TJ, Kass MA, Yablonski ME, Becker B. Timolol maleate: efficacy and safety. Arch Ophthalmol. 1979;97(4):656–8.PubMedCrossRef
117.
Zurück zum Zitat Katz LJ. Twelve-month evaluation of brimonidine-purite versus brimonidine in patients with glaucoma or ocular hypertension. J Glaucoma. 2002;11(2):119–26.PubMedCrossRef Katz LJ. Twelve-month evaluation of brimonidine-purite versus brimonidine in patients with glaucoma or ocular hypertension. J Glaucoma. 2002;11(2):119–26.PubMedCrossRef
118.
Zurück zum Zitat Liu JH, Kripke DF, Weinreb RN. Comparison of the nocturnal effects of once-daily timolol and latanoprost on intraocular pressure. Am J Ophthalmol. 2004;138(3):389–95.PubMedCrossRef Liu JH, Kripke DF, Weinreb RN. Comparison of the nocturnal effects of once-daily timolol and latanoprost on intraocular pressure. Am J Ophthalmol. 2004;138(3):389–95.PubMedCrossRef
119.
Zurück zum Zitat Liu JH, Medeiros FA, Slight JR, Weinreb RN. Diurnal and nocturnal effects of brimonidine monotherapy on intraocular pressure. Ophthalmology. 2010. Liu JH, Medeiros FA, Slight JR, Weinreb RN. Diurnal and nocturnal effects of brimonidine monotherapy on intraocular pressure. Ophthalmology. 2010.
120.
Zurück zum Zitat Liu JH, Zhang X, Kripke DF, Weinreb RN. Twenty-four-hour intraocular pressure pattern associated with early glaucomatous changes. Invest Ophthalmol Vis Sci. 2003;44(4):1586–90.PubMedCrossRef Liu JH, Zhang X, Kripke DF, Weinreb RN. Twenty-four-hour intraocular pressure pattern associated with early glaucomatous changes. Invest Ophthalmol Vis Sci. 2003;44(4):1586–90.PubMedCrossRef
121.
Zurück zum Zitat Ciulla L, Moorthy M, Mathew S, Siesky B, Verticchio Vercellin AC, Price D, et al. Circadian rhythm and glaucoma: what do we know? J Glaucoma. 2020;29(2):127–32.PubMedCrossRef Ciulla L, Moorthy M, Mathew S, Siesky B, Verticchio Vercellin AC, Price D, et al. Circadian rhythm and glaucoma: what do we know? J Glaucoma. 2020;29(2):127–32.PubMedCrossRef
122.
Zurück zum Zitat Jampel HD, Chon BH, Stamper R, Packer M, Han Y, Nguyen QH, et al. Effectiveness of intraocular pressure-lowering medication determined by washout. JAMA Ophthalmol. 2014;132(4):390–5.PubMedCrossRef Jampel HD, Chon BH, Stamper R, Packer M, Han Y, Nguyen QH, et al. Effectiveness of intraocular pressure-lowering medication determined by washout. JAMA Ophthalmol. 2014;132(4):390–5.PubMedCrossRef
123.
Zurück zum Zitat Johnson TV, Jampel HD. Intraocular pressure following prerandomization glaucoma medication washout in the HORIZON and COMPASS trials. Am J Ophthalmol. 2020;216:110–20.PubMedCrossRef Johnson TV, Jampel HD. Intraocular pressure following prerandomization glaucoma medication washout in the HORIZON and COMPASS trials. Am J Ophthalmol. 2020;216:110–20.PubMedCrossRef
124.
Zurück zum Zitat Bartlett VL, Liu P, Dhruva SS, Shah ND, Bollinger KE, Ross JS. Prostaglandin coverage and costs to Medicare and Medicare beneficiaries, 2009–2017. J Manag Care Spec Pharm. 2020;26(4):562–7.PubMed Bartlett VL, Liu P, Dhruva SS, Shah ND, Bollinger KE, Ross JS. Prostaglandin coverage and costs to Medicare and Medicare beneficiaries, 2009–2017. J Manag Care Spec Pharm. 2020;26(4):562–7.PubMed
125.
Zurück zum Zitat Rasendran C, Li A, Singh RP. Incremental health care expenditures associated with glaucoma in the united states: a propensity score-matched analysis. J Glaucoma. 2022;31(1):1–7.PubMedCrossRef Rasendran C, Li A, Singh RP. Incremental health care expenditures associated with glaucoma in the united states: a propensity score-matched analysis. J Glaucoma. 2022;31(1):1–7.PubMedCrossRef
126.
Zurück zum Zitat Schehlein EM, Im LT, Robin AL, Onukwugha E, Saeedi OJ. Nonmedical out-of-pocket patient and companion expenditures associated with glaucoma care. J Glaucoma. 2017;26(4):343–8.PubMedPubMedCentralCrossRef Schehlein EM, Im LT, Robin AL, Onukwugha E, Saeedi OJ. Nonmedical out-of-pocket patient and companion expenditures associated with glaucoma care. J Glaucoma. 2017;26(4):343–8.PubMedPubMedCentralCrossRef
127.
Zurück zum Zitat Blumberg DM, Prager AJ, Liebmann JM, Cioffi GA, De Moraes CG. Cost-related medication nonadherence and cost-saving behaviors among patients with glaucoma before and after the implementation of Medicare part D. JAMA Ophthalmol. 2015;133(9):985–96.PubMedCrossRef Blumberg DM, Prager AJ, Liebmann JM, Cioffi GA, De Moraes CG. Cost-related medication nonadherence and cost-saving behaviors among patients with glaucoma before and after the implementation of Medicare part D. JAMA Ophthalmol. 2015;133(9):985–96.PubMedCrossRef
128.
Zurück zum Zitat Sood S, Heilenbach N, Sanchez V, Glied S, Chen S, Al-Aswad LA. Cost-effectiveness analysis of minimally invasive trabecular meshwork stents with phacoemulsification. Ophthalmol Glaucoma. 2022;5(3):284–96.PubMedCrossRef Sood S, Heilenbach N, Sanchez V, Glied S, Chen S, Al-Aswad LA. Cost-effectiveness analysis of minimally invasive trabecular meshwork stents with phacoemulsification. Ophthalmol Glaucoma. 2022;5(3):284–96.PubMedCrossRef
129.
Zurück zum Zitat Ahmed IIK, Podbielski DW, Patel V, Falvey H, Murray J, Botteman M, et al. A Canadian cost-utility analysis of 2 trabecular microbypass stents at time of cataract surgery in patients with mild to moderate open-angle glaucoma. Ophthalmol Glaucoma. 2020;3(2):103–13.PubMedCrossRef Ahmed IIK, Podbielski DW, Patel V, Falvey H, Murray J, Botteman M, et al. A Canadian cost-utility analysis of 2 trabecular microbypass stents at time of cataract surgery in patients with mild to moderate open-angle glaucoma. Ophthalmol Glaucoma. 2020;3(2):103–13.PubMedCrossRef
130.
Zurück zum Zitat Ngan K, Fraser E, Buller S, Buller A. A cost minimisation analysis comparing iStent accompanying cataract surgery and selective laser trabeculoplasty versus topical glaucoma medications in a public healthcare setting in New Zealand. Graefes Arch Clin Exp Ophthalmol. 2018;256(11):2181–9.PubMedCrossRef Ngan K, Fraser E, Buller S, Buller A. A cost minimisation analysis comparing iStent accompanying cataract surgery and selective laser trabeculoplasty versus topical glaucoma medications in a public healthcare setting in New Zealand. Graefes Arch Clin Exp Ophthalmol. 2018;256(11):2181–9.PubMedCrossRef
131.
Zurück zum Zitat Stein JD, Kim DD, Peck WW, Giannetti SM, Hutton DW. Cost-effectiveness of medications compared with laser trabeculoplasty in patients with newly diagnosed open-angle glaucoma. Arch Ophthalmol. 2012;130(4):497–505.PubMedPubMedCentralCrossRef Stein JD, Kim DD, Peck WW, Giannetti SM, Hutton DW. Cost-effectiveness of medications compared with laser trabeculoplasty in patients with newly diagnosed open-angle glaucoma. Arch Ophthalmol. 2012;130(4):497–505.PubMedPubMedCentralCrossRef
133.
Zurück zum Zitat Janz NK, Wren PA, Lichter PR, Musch DC, Gillespie BW, Guire KE, et al. The Collaborative Initial Glaucoma Treatment Study: interim quality of life findings after initial medical or surgical treatment of glaucoma. Ophthalmology. 2001;108(11):1954–65.PubMedCrossRef Janz NK, Wren PA, Lichter PR, Musch DC, Gillespie BW, Guire KE, et al. The Collaborative Initial Glaucoma Treatment Study: interim quality of life findings after initial medical or surgical treatment of glaucoma. Ophthalmology. 2001;108(11):1954–65.PubMedCrossRef
134.
Zurück zum Zitat Latif K, Nishida T, Moghimi S, Weinreb RN. Quality of life in glaucoma. Graefes Arch Clin Exp Ophthalmol. 2023;261:3023.PubMedCrossRef Latif K, Nishida T, Moghimi S, Weinreb RN. Quality of life in glaucoma. Graefes Arch Clin Exp Ophthalmol. 2023;261:3023.PubMedCrossRef
135.
Zurück zum Zitat Nishida T, Moghimi S, Yamane MLM, Wu JH, Mohammadzadeh V, Kamalipour A, et al. Vision-related quality of life among healthy, preperimetric glaucoma, and perimetric glaucoma patients. Am J Ophthalmol. 2023;248:127–36.PubMedCrossRef Nishida T, Moghimi S, Yamane MLM, Wu JH, Mohammadzadeh V, Kamalipour A, et al. Vision-related quality of life among healthy, preperimetric glaucoma, and perimetric glaucoma patients. Am J Ophthalmol. 2023;248:127–36.PubMedCrossRef
136.
Zurück zum Zitat Nordmann JP, Auzanneau N, Ricard S, Berdeaux G. Vision related quality of life and topical glaucoma treatment side effects. Health Qual Life Outcomes. 2003;1(1):75.PubMedPubMedCentralCrossRef Nordmann JP, Auzanneau N, Ricard S, Berdeaux G. Vision related quality of life and topical glaucoma treatment side effects. Health Qual Life Outcomes. 2003;1(1):75.PubMedPubMedCentralCrossRef
137.
Zurück zum Zitat Balkrishnan R, Bond JB, Byerly WG, Camacho FT, Anderson RT. Medication-related predictors of health-related quality of life in glaucoma patients enrolled in a Medicare health maintenance organization. Am J Geriatr Pharmacother. 2003;1(2):75–81.PubMedCrossRef Balkrishnan R, Bond JB, Byerly WG, Camacho FT, Anderson RT. Medication-related predictors of health-related quality of life in glaucoma patients enrolled in a Medicare health maintenance organization. Am J Geriatr Pharmacother. 2003;1(2):75–81.PubMedCrossRef
138.
Zurück zum Zitat Samuelson TW, Singh IP, Williamson BK, Falvey H, Lee WC, Odom D, 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;229:220–9.PubMedCrossRef Samuelson TW, Singh IP, Williamson BK, Falvey H, Lee WC, Odom D, 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;229:220–9.PubMedCrossRef
139.
Zurück zum Zitat Al Habash A, Albuainain A. Long term outcome of combined phacoemulsification and excisional goniotomy with the Kahook Dual Blade in different subtypes of glaucoma. Sci Rep. 2021;11(1):10660.PubMedPubMedCentralCrossRef Al Habash A, Albuainain A. Long term outcome of combined phacoemulsification and excisional goniotomy with the Kahook Dual Blade in different subtypes of glaucoma. Sci Rep. 2021;11(1):10660.PubMedPubMedCentralCrossRef
140.
Zurück zum Zitat Al Habash A, Nagshbandi AA. Quality of life after combined cataract and minimally invasive glaucoma surgery in glaucoma patients. Clin Ophthalmol (Auckland, NZ). 2020;14:3049–56.CrossRef Al Habash A, Nagshbandi AA. Quality of life after combined cataract and minimally invasive glaucoma surgery in glaucoma patients. Clin Ophthalmol (Auckland, NZ). 2020;14:3049–56.CrossRef
141.
Zurück zum Zitat Kass MA, Gordon MO, Gao F, Heuer DK, Higginbotham EJ, Johnson CA, et al. Delaying treatment of ocular hypertension: the ocular hypertension treatment study. Arch Ophthalmol. 2010;128(3):276–87.PubMedPubMedCentralCrossRef Kass MA, Gordon MO, Gao F, Heuer DK, Higginbotham EJ, Johnson CA, et al. Delaying treatment of ocular hypertension: the ocular hypertension treatment study. Arch Ophthalmol. 2010;128(3):276–87.PubMedPubMedCentralCrossRef
142.
Zurück zum Zitat Hyman LG, Komaroff E, Heijl A, Bengtsson B, Leske MC. Treatment and vision-related quality of life in the early manifest glaucoma trial. Ophthalmology. 2005;112(9):1505–13.PubMedCrossRef Hyman LG, Komaroff E, Heijl A, Bengtsson B, Leske MC. Treatment and vision-related quality of life in the early manifest glaucoma trial. Ophthalmology. 2005;112(9):1505–13.PubMedCrossRef
143.
Zurück zum Zitat Jones L, Garway-Heath DF, Azuara-Blanco A, Crabb DP, United Kingdom Glaucoma Treatment Study I. Are Patient self-reported outcome measures sensitive enough to be used as end points in clinical trials?: evidence from the united kingdom glaucoma treatment study. Ophthalmology. 2019;126(5):682–9. Jones L, Garway-Heath DF, Azuara-Blanco A, Crabb DP, United Kingdom Glaucoma Treatment Study I. Are Patient self-reported outcome measures sensitive enough to be used as end points in clinical trials?: evidence from the united kingdom glaucoma treatment study. Ophthalmology. 2019;126(5):682–9.
144.
Zurück zum Zitat Vandenbroeck S, De Geest S, Zeyen T, Stalmans I, Dobbels F. Patient-reported outcomes (PRO’s) in glaucoma: a systematic review. Eye (Lond). 2011;25(5):555–77.PubMedCrossRef Vandenbroeck S, De Geest S, Zeyen T, Stalmans I, Dobbels F. Patient-reported outcomes (PRO’s) in glaucoma: a systematic review. Eye (Lond). 2011;25(5):555–77.PubMedCrossRef
145.
Zurück zum Zitat Walland MJ. Glaucoma treatment in Australia: changing patterns of therapy 1994–2003. Clin Exp Ophthalmol. 2004;32(6):590–6.PubMedCrossRef Walland MJ. Glaucoma treatment in Australia: changing patterns of therapy 1994–2003. Clin Exp Ophthalmol. 2004;32(6):590–6.PubMedCrossRef
146.
Zurück zum Zitat Newman AR, Andrew NH. Changes in Australian practice patterns for glaucoma management. Clin Exp Ophthalmol. 2019;47(5):571–80.PubMedCrossRef Newman AR, Andrew NH. Changes in Australian practice patterns for glaucoma management. Clin Exp Ophthalmol. 2019;47(5):571–80.PubMedCrossRef
148.
Zurück zum Zitat European Glaucoma Society. Terminology and guidelines for glaucoma. 5th ed. Savona: PubliComm; 2020. European Glaucoma Society. Terminology and guidelines for glaucoma. 5th ed. Savona: PubliComm; 2020.
149.
Zurück zum Zitat Realini T, Gazzard G, Latina M, Kass M. Low-energy selective laser trabeculoplasty repeated annually: rationale for the COAST trial. J Glaucoma. 2021;30(7):545–51.PubMedPubMedCentralCrossRef Realini T, Gazzard G, Latina M, Kass M. Low-energy selective laser trabeculoplasty repeated annually: rationale for the COAST trial. J Glaucoma. 2021;30(7):545–51.PubMedPubMedCentralCrossRef
150.
Zurück zum Zitat Ruiz-Lozano RE, Alamillo-Velazquez J, Ortiz-Morales G, Garza-Garza LA, Quiroga-Garza ME, Alvarez-Guzman C, et al. Selective laser trabeculoplasty is safe and effective in patients previously treated with prostaglandin analogs: an evidence-based review. Int Ophthalmol. 2023;43(2):677–95.PubMedCrossRef Ruiz-Lozano RE, Alamillo-Velazquez J, Ortiz-Morales G, Garza-Garza LA, Quiroga-Garza ME, Alvarez-Guzman C, et al. Selective laser trabeculoplasty is safe and effective in patients previously treated with prostaglandin analogs: an evidence-based review. Int Ophthalmol. 2023;43(2):677–95.PubMedCrossRef
151.
Zurück zum Zitat Evidence review A. Evidence reviews for selective laser trabeculoplasty in ocular hypertension or chronic open-angle glaucoma adult patients: Glaucoma: diagnosis and management. NICE Evidence Reviews Collection. London: National Institute for Health and Care Excellence; 2022. Evidence review A. Evidence reviews for selective laser trabeculoplasty in ocular hypertension or chronic open-angle glaucoma adult patients: Glaucoma: diagnosis and management. NICE Evidence Reviews Collection. London: National Institute for Health and Care Excellence; 2022.
152.
Zurück zum Zitat Seider MI, Keenan JD, Han Y. Cost of selective laser trabeculoplasty vs topical medications for glaucoma. Arch Ophthalmol. 2012;130(4):529–30.PubMedCrossRef Seider MI, Keenan JD, Han Y. Cost of selective laser trabeculoplasty vs topical medications for glaucoma. Arch Ophthalmol. 2012;130(4):529–30.PubMedCrossRef
153.
Zurück zum Zitat Shah M. Micro-invasive glaucoma surgery—an interventional glaucoma revolution. Eye Vis (Lond). 2019;6:29.PubMedCrossRef Shah M. Micro-invasive glaucoma surgery—an interventional glaucoma revolution. Eye Vis (Lond). 2019;6:29.PubMedCrossRef
154.
Zurück zum Zitat Richter GM, Coleman AL. Minimally invasive glaucoma surgery: current status and future prospects. Clin Ophthalmol. 2016;10:189–206.PubMedPubMedCentral Richter GM, Coleman AL. Minimally invasive glaucoma surgery: current status and future prospects. Clin Ophthalmol. 2016;10:189–206.PubMedPubMedCentral
155.
Zurück zum Zitat Saheb H, Ahmed II. Micro-invasive glaucoma surgery: current perspectives and future directions. Curr Opin Ophthalmol. 2012;23(2):96–104.PubMedCrossRef Saheb H, Ahmed II. Micro-invasive glaucoma surgery: current perspectives and future directions. Curr Opin Ophthalmol. 2012;23(2):96–104.PubMedCrossRef
156.
Zurück zum Zitat Radcliffe N. The case for standalone micro-invasive glaucoma surgery: rethinking the role of surgery in the glaucoma treatment paradigm. Curr Opin Ophthalmol. 2023;34(2):138–45.PubMedCrossRef Radcliffe N. The case for standalone micro-invasive glaucoma surgery: rethinking the role of surgery in the glaucoma treatment paradigm. Curr Opin Ophthalmol. 2023;34(2):138–45.PubMedCrossRef
157.
Zurück zum Zitat Pahlitzsch M, Davids AM, Winterhalter S, Zorn M, Reitemeyer E, Klamann MKJ, et al. Selective laser trabeculoplasty versus MIGS: forgotten art or first-step procedure in selected patients with open-angle glaucoma. Ophthalmol Ther. 2021;10(3):509–24.PubMedPubMedCentralCrossRef Pahlitzsch M, Davids AM, Winterhalter S, Zorn M, Reitemeyer E, Klamann MKJ, et al. Selective laser trabeculoplasty versus MIGS: forgotten art or first-step procedure in selected patients with open-angle glaucoma. Ophthalmol Ther. 2021;10(3):509–24.PubMedPubMedCentralCrossRef
158.
Zurück zum Zitat Tektas OY, Lutjen-Drecoll E. Structural changes of the trabecular meshwork in different kinds of glaucoma. Exp Eye Res. 2009;88(4):769–75.PubMedCrossRef Tektas OY, Lutjen-Drecoll E. Structural changes of the trabecular meshwork in different kinds of glaucoma. Exp Eye Res. 2009;88(4):769–75.PubMedCrossRef
159.
Zurück zum Zitat Montesano G, Ometto G, Ahmed IIK, Ramulu PY, Chang DF, Crabb DP, et al. Five-year visual field outcomes of the HORIZON trial. Am J Ophthalmol. 2023;251:143–55.PubMedCrossRef Montesano G, Ometto G, Ahmed IIK, Ramulu PY, Chang DF, Crabb DP, et al. Five-year visual field outcomes of the HORIZON trial. Am J Ophthalmol. 2023;251:143–55.PubMedCrossRef
160.
Zurück zum Zitat Wright DM, Konstantakopoulou E, Montesano G, Nathwani N, Garg A, Garway-Heath D, et al. Visual field outcomes from the multicenter, randomized controlled laser in glaucoma and ocular hypertension trial (LiGHT). Ophthalmology. 2020;127(10):1313–21.PubMedCrossRef Wright DM, Konstantakopoulou E, Montesano G, Nathwani N, Garg A, Garway-Heath D, et al. Visual field outcomes from the multicenter, randomized controlled laser in glaucoma and ocular hypertension trial (LiGHT). Ophthalmology. 2020;127(10):1313–21.PubMedCrossRef
161.
Zurück zum Zitat Fea AM, Cattel F, Gandolfi S, Buseghin G, Furneri G, Costagliola C. Cost-utility analysis of trabecular micro-bypass stents (TBS) in patients with mild-to-moderate open-angle Glaucoma in Italy. BMC Health Serv Res. 2021;21(1):824.PubMedPubMedCentralCrossRef Fea AM, Cattel F, Gandolfi S, Buseghin G, Furneri G, Costagliola C. Cost-utility analysis of trabecular micro-bypass stents (TBS) in patients with mild-to-moderate open-angle Glaucoma in Italy. BMC Health Serv Res. 2021;21(1):824.PubMedPubMedCentralCrossRef
Metadaten
Titel
Challenging the "Topical Medications-First” Approach to Glaucoma: A Treatment Paradigm in Evolution
verfasst von
Nathan M. Radcliffe
Manjool Shah
Thomas W. Samuelson
Publikationsdatum
19.10.2023
Verlag
Springer Healthcare
Erschienen in
Ophthalmology and Therapy / Ausgabe 6/2023
Print ISSN: 2193-8245
Elektronische ISSN: 2193-6528
DOI
https://doi.org/10.1007/s40123-023-00831-9

Weitere Artikel der Ausgabe 6/2023

Ophthalmology and Therapy 6/2023 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Perioperative Checkpointhemmer-Therapie verbessert NSCLC-Prognose

28.05.2024 NSCLC Nachrichten

Eine perioperative Therapie mit Nivolumab reduziert das Risiko für Rezidive und Todesfälle bei operablem NSCLC im Vergleich zu einer alleinigen neoadjuvanten Chemotherapie um über 40%. Darauf deuten die Resultate der Phase-3-Studie CheckMate 77T.

Positiver FIT: Die Ursache liegt nicht immer im Dickdarm

27.05.2024 Blut im Stuhl Nachrichten

Immunchemischer Stuhltest positiv, Koloskopie negativ – in solchen Fällen kann die Blutungsquelle auch weiter proximal sitzen. Ein Forschungsteam hat nachgesehen, wie häufig und in welchen Lokalisationen das der Fall ist.

GLP-1-Agonisten können Fortschreiten diabetischer Retinopathie begünstigen

24.05.2024 Diabetische Retinopathie Nachrichten

Möglicherweise hängt es von der Art der Diabetesmedikamente ab, wie hoch das Risiko der Betroffenen ist, dass sich sehkraftgefährdende Komplikationen verschlimmern.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.