Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Aug 8.
Published in final edited form as: J Med Chem. 2015 Sep 25;59(3):788–809. doi: 10.1021/acs.jmedchem.5b00828

Discovery of molecular therapeutics for glaucoma: Challenges, successes, and promising directions

Rebecca K Donegan 1, Raquel L Lieberman 1
PMCID: PMC5547565  NIHMSID: NIHMS882120  PMID: 26356532

Abstract

Glaucoma, a heterogeneous ocular disorder affecting ~60 million people worldwide, is characterized by painless neurodegeneration of retinal ganglion cells (RGCs), resulting in irreversible vision loss. Available therapies, which decrease the common causal risk factor of elevated intraocular pressure, delay, but cannot prevent, RGC death and blindness. Notably, it is changes in the anterior segment of the eye, particularly in the drainage of aqueous humor fluid, which are believed to bring about changes in pressure. Thus, it is primarily this region whose properties are manipulated in current and emerging therapies for glaucoma. Here, we focus on the challenges associated with developing treatments, review the available experimental methods to evaluate the therapeutic potential of new drugs, describe the development and evaluation of emerging Rho-kinase inhibitors and adenosine receptor ligands that offer the potential to improve aqueous humor outflow and protect RGCs simultaneously, and present new targets and approaches on the horizon.

Graphical Abstract

graphic file with name nihms882120u1.jpg

INTRODUCTION

Glaucoma: a brief background

Glaucoma, a group of neurodegenerative diseases of the optic nerve in the posterior eye, with varying etiologies, is a leading cause of irreversible blindness1, 2 and affects more than 60 million people worldwide.3 Glaucoma can be divided into two major subtypes, based on whether the iridocorneal angle (Figure 1) is closed (closed-angle glaucoma (CAG)), namely, in which the iris physically comes in contact with the trabecular meshwork (TM), or open (open-angle glaucoma (OAG)). Primary open-angle glaucoma (POAG), the most common form overall, refers to presentation of glaucoma symptoms without attribution to other disease, injury, or closed iridocorneal angle.4 Secondary forms of glaucoma are those attributable to other causes; for example, secondary open angle glaucoma can be a result of pseudoexfoliation (PEX) syndrome5 or prolonged use of steroids.6 Glaucoma can be further divided based on age of onset into congenital, juvenile (JOAG, for those with onset at less than 35 years of age), and the more common, adult onset.

Figure 1. Overview of the anterior segment of the eye.

Figure 1

Key anatomical structures discussed in this Perspective are labeled. The conventional pathway for aqueous humor outflow is highlighted with blue arrows.

Patients with glaucoma are typically asymptomatic until vision loss, which is caused by damage to the nerve fiber layer surrounding retinal ganglion cells (RGCs) followed by RGC death.7 A clinically described feature referred to as ‘cupping’ differentiates glaucoma from other optic neuropathies involving RGC death.8 Namely, a change is apparent in the bundled optic nerve fibers at the center of the optic disc (i.e. the ‘cup’); the extent of cupping is used to monitor disease progression in high-risk patients before any changes in visual field are obvious.1 A more readily monitored causal risk factor is elevated intraocular pressure (IOP), but it is not a necessary precondition for RGC death and vision loss in glaucoma, as in the case of a third glaucoma subtype, normal tension glaucoma (NTG).2, 9 As discussed in this Perspective, both for patients exhibiting elevated pressure as well as those with NTG, the standard of care is to slow loss of visual field by reduction of IOP.911 This approach has proven useful in reducing the loss of visual field over time, but IOP reduction alone does not halt disease progression.12 As described below, IOP-reducing methods currently used in the clinic, as well as emerging therapies, are not developed de novo to target specific molecular underpinnings of glaucoma but rather are derived from other similar disorders, such as those of the cardiovascular system.

Efforts to improve our understanding of glaucoma pathogenesis, which remains unclear due to inherent disease heterogeneity and late age onset, involve identifying genetic susceptibility. The first, best-studied, and to date most strongly glaucoma-associated gene encodes for myocilin.13 Initially named the trabecular meshwork inducible glucocorticoid response (TIGR) gene due to its link with steroid-induced secondary glaucoma,14 numerous mutations in myocilin with autosomal dominant Mendelian inheritance patterns have since been linked with POAG and JOAG, in nearly every population and country throughout the world.15, 16 Other associated genes include those for (OPTN) and WDR36, which are seen in both inherited and sporadic forms of adult onset OAG.1517 The Glu50Lys variant of OPTN is also associated with NTG.18, 19 Together, the known genetic variants of MYOC, OPTN, and WDR36 account for about 10% of glaucoma cases,15, 16 but relatives of all POAG patients are 20% more likely to develop glaucoma in their lifetime than controls, pointing to a wider genetic link.20 Genome-wide association studies (GWAS), particularly the worldwide consortium with the acronym NEIGHBOR,21 continue to report susceptibility loci for glaucoma.22, 23 For example, a gene product that has received some attention among the over 20 genes identified by GWAS is CYP1B1, associated with primary congenital glaucoma as well as POAG.17 With the exception of myocilin discussed below, however, how these proteins contribute to pathogenesis remains yet unknown.

Relevant anatomical changes with age/glaucoma

Aqueous humor (AH), the fluid found in the anterior segment, supplies nutrients to nonvascular eye tissues such as the lens and cornea. The balance between AH production and outflow is intimately tied to IOP maintenance.24 AH is produced in the non-pigmented epithelial (NPE) cells of the ciliary body, then flows to the anterior segment, and eventually drains through the (1) so-called conventional pathway via the trabecular meshwork (TM) to Schlemm’s canal (SC) and finally into the episcleral veins (Figure 1), or via (2) the uveoscleral pathway through the ciliary body and iris root, which bypasses the TM and SC.24 Active transport of chloride and sodium ions from the stroma to the AH, across both the pigmented epithelium and NPE of the ciliary body, is responsible for the majority of AH formation.25 Control of ion movement across the ciliary epithelium is regulated at least in part by cyclic AMP (cAMP) modulation. Current and emerging therapeutic targets include those in signaling cascades that ultimately affect cAMP levels (see below), but studies to date do not yet converge on whether increased levels of cAMP increase26 or decrease27 AH formation.

AH outflow facility is altered in both normal aging and glaucoma.28 In the healthy aging eye, IOP is maintained as both inflow and uveoscleral outflow tend to decrease; conventional outflow facility is unchanged.29 By comparison, glaucoma-associated IOP increases are believed to be a result of reduced outflow predominantly through the conventional pathway, not increased AH formation or decreased outflow through the uveoscleral pathway.28 Documented anatomical alterations include increased extracellular matrix (ECM) stiffness in the TM,30 increased cell stiffness of the SC, decreased SC epithelial cell pore formation, a loss of TM cells,31 and increased sheath-derived plaque formation in the TM and SC.32 Nevertheless, the extent to which these are primary culprits for pathogenesis is complicated by the fact that changes to the TM and SC due to glaucoma versus the normal aging process are difficult to distinguish.33

Model systems used to study glaucoma

A major impediment to developing new glaucoma therapeutics is the intrinsic heterogeneity of the disease. There is no established model system e.g. tissue or organ culture, animal model, or human postmortem tissue, to study the disease, develop, or systematically evaluate potential new treatments. Cell culture and organ models offer a controlled environment and are useful for initial screening of new drugs, but are derived from human postmortem tissues that vary widely34. Often the medical history of a donor eye is not well documented and the eyes may be affected by undiagnosed disease, as well as long-term drug treatments for glaucoma and/or other disorders.

The use of cultures lacks a robust clinical translation such as that seen with use of animal models.34 Yet, animal eyes, including those of birds, rodents, rabbits, dogs, cats and primates, vary greatly in their anatomical similarity to those of humans. Glaucoma in animal models may be induced by various methods including genetic mutation, treatment with steroids or photocoagulation, or in some species, occurs spontaneously.35 Induced glaucoma offers quick, but non-natural, disease progression, while spontaneous glaucoma may take years for symptoms to appear.35 Rat and mouse glaucoma models are the most commonly used animals due to cost and access, but their eyes are not as similar to humans as other animal models and their small size can make testing difficult.34, 35 Primate eyes are the most similar to those of humans, but such animals are limited in availability and expensive to study.34, 36

Standard experiments used to evaluate molecules of therapeutic potential for glaucoma

Typical drug discovery methods, namely high throughput screening of chemical libraries using biochemical or cell-based assays, cannot be used to detect reduction in IOP or changes in outflow resistance. Rather, IOP changes are typically assessed directly in animal models using rebound tonometry, a non-invasive method. Rebound tonometry uses a small probe that is rebounded off of the cornea, where the inverse of the deceleration speed of the probe correlates well with IOP.37, 38 Several measurements can be taken back-to-back or on different points of the cornea without harm. However, high variance is inherent in this measurement, and can be greater than the reductions in IOP afforded by treatment, thus complicating data interpretation when limited numbers of specimens or animals can be investigated.39 To assess outflow facility, perfusion of mounted post-mortem eye tissues is commonly employed. In this experiment, solutions containing small molecules of interest are perfused through the sample, and changes in eye tissue with increased perfusion pressures are monitored.40 Perfusion experiments are limited by availability of specimens, and variations in handling of these ex vivo samples will affect measurements.

TREATMENTS CURRENTLY USED IN THE CLINIC

Early detection is critical for preventing vision loss, as any loss of visual field due to retina degeneration is not reversible, but diagnosis is complicated by the fact that the age of onset and rate of glaucoma pathogenesis varies for different forms of the disease and varies widely among patients.41 Early signs of the disease can be missed if only IOP or variations in visual field are monitored, so ophthalmologists typically also look for changes or damage to the optic disk.1 Once diagnosed, options to treat glaucoma fall into several classes all of which target lowering IOP as a method to slow disease progression.10 IOP is modulated by decreasing AH production or by increasing outflow. Even though drugs are generally applied topically to the eye, the five classes of current therapies (Table 1A) are not selective to eye tissues and were originally developed for other diseases. In addition, there are surgical options (Table 1B). Below we briefly summarize the five classes of currently marketed drugs and surgery used; for comprehensive descriptions and other clinical considerations, we refer the reader to a number of excellent review articles.4245

Table 1.

Current glaucoma therapies used in the clinic

A. IOP Lowering Drugs
Class Example Method of IOP Reduction
Cholinomimetics (Cholinergic Agonists) Pilocarpine Increase AH outflow
Adrenergic receptor agonists (sympathomimetics) Brimonidine Reduce AH production, increase outflow
β-Adrenergic receptor antagonists (β-Blockers) Timolol Reduce AH production
Carbonic Anhydrase inhibitors Dorzolamide Reduce AH production
Prostaglandin analogues Latanoprost Increase uveoslceral outflow
B. Surgery
CAG
Type Purpose
Laser peripheral iridotomy (LPI) Widen iridocorneal angle
Iridoplasty Widen iridocorneal angle
Argon laser peripheral iridoplasty (ALPI) Contraction burns to create space between the anterior iris surface and the TM
Phacoemulsification cataract extraction (phaco/IOL) Removal of lens alleviates pressure
POAG
Type Purpose
Glaucoma filter surgery/trabeculectomy Partial removal of drainage angle allowing AH to bypass TM and alleviate pressure
Trabeculectomy with Shunt/Stent Trabeculectomy with addition of shunt or stent to alleviate pressure
Trabeculoplasty Lasers open drainage channels to alleviate pressure

Cholinomimetics

The first class of drugs used to treat glaucoma, cholinergic agonists (also known as cholinomimetics, miotics, parasympathomimetics, acetylcholine receptor agonists), have been in use as glaucoma therapeutics since the 1870s.46 Cholinergic agonists increase AH outflow through the conventional pathway by causing contraction of the ciliary body. Cholinergic agonists are divided into two classes, those that act directly by binding to muscarinic acetylcholine receptors in the ciliary body, or indirectly by inhibiting acetylcholinesterase.1, 47 Physostigmine, the first cholinergic agonist, was used during eye surgery to constrict the pupil for over a decade before its IOP reducing effects were realized.46 Pilocarpine, the most common44 and oldest46 cholinergic agonist glaucoma drug, is a nonselective muscarinic acetylcholine receptor inhibitor.44 A third molecule, carbachol, works by both direct and indirect inhibition.48

Adrenergic receptor agonists (sympathomimetics) and antagonists (β-blockers)

Adrenergic receptors are G-protein coupled receptors (GPCRs) that regulate the sympathetic nervous system. Epinephrine, the natural agonist of adrenergic receptors, was used as a glaucoma therapy in the 1950s.46 Currently, the most commonly prescribed adrenergic receptor agonists are selectively targeted to the α2 subclass (Table 1A),49 and are sometimes coupled with a β-adrenergic receptor antagonist (next paragraph), which together are particularly effective for patients who do not tolerate prostaglandin analogs (see below). The α2-adrenergic receptors are involved in a cascade that maintains vascular constriction by activating Gi proteins, which inhibit adenylyl cyclase50 and prevents protein kinase A (PKA) from phosphorylating myosin light chain (MLC) kinase, which remains active and prevents vascular relaxation.51, 52 Agonists of α2-adrenergic receptors are believed to decrease AH production by decreasing blood flow to the ciliary body,1, 5254 and increase uveoscleral outflow.55

The topical administration of β-adrenergic receptor antagonists, or β-blockers, which have been used to treat cardiovascular hypertension since the 1960s,56, 57 was a breakthrough in glaucoma treatment, since they significantly lowered IOP with fewer side effects than older glaucoma drugs. Before the introduction of prostaglandin analogs (see below), β-blockers were the most commonly prescribed treatment for glaucoma.58, 59 β-adrenergic receptors are found within the ciliary body60 and early studies showed β-blocker propranolol decreased IOP in glaucoma patients,61 but its usefulness was limited due to side effects.46 In the late 1970s, a topical timolol treatment became the first FDA approved β-blocker for the treatment of glaucoma.46 Timolol decreases the formation of AH,62, 63 perhaps via the aforementioned mechanisms involving chloride transport in ciliary epithelium and cAMP,25, 64 or by cAMP-independent mechanisms,65, 66 but does not affect outflow facility.62, 63

Carbonic Anhydrase Inhibitors

Carbonic anhydrase inhibitors (CAIs) are the best understood glaucoma therapeutic class on a molecular level. The production of bicarbonate by the Zn-dependent carbonic anhydrase enzyme is necessary for chloride transport, and thus AH formation (see above).24 Inhibition of carbonic anhydrase decreases AH formation and in turn decreases IOP. The original CAIs were sulfonamides developed as diuretics and antimicrobials.46 Current CAIs include the systemic acetazolamide and the topical dorzolamide.48 While CAIs can offer decreases in IOP similar to β-blockers (see above), this effect is seen mostly with systemic treatment, which, with prolonged use, can cause side effects such as paresthesia of the hands and feet. Topical CAIs have fewer adverse effects, but are also less potent.59

Prostaglandin Analogs

Prostaglandin analogs, the newest class of glaucoma drugs, have become the first line of treatment.67 Naturally occurring prostaglandins bind a variety of cell surface receptors and are involved in mediating smooth muscle contraction and inflammation.68 Since their discovery,69 prostaglandins and their analogs have been studied as potential therapeutics in a range of diseases. For glaucoma, the finding that prostaglandin F2α lowered IOP in monkeys bolstered the interest in further evaluating prostaglandins for therapeutic intervention.46 Prostaglandin analogs have been shown to lower IOP mainly via increasing uveoscleral outflow,70, 71 along with conventional AH outflow.72 While the specific mechanism of action of increasing outflow remains unknown, studies indicate that prostaglandin analogs affect ECM remodeling7375 and may relax the ciliary body.76 Prostaglandin analogs have short half-lives and can be administered locally to the eye at low doses, which reduces the likelihood of occurrence of side effects.67

Surgery and Implants

When drugs fail to lower IOP, more invasive measures such as laser treatment, surgery or shunt implant may be employed (Table 1B). Improvements in surgical techniques have lowered the risk of such procedures and are thus becoming more prevalent, including alongside drug treatments, for both CAG and POAG. Argon laser trabeculoplasty (ALT), or the newer, lower energy alternative, selective laser trabeculoplasty (SLT) mechanically increase outflow by damaging TM tissue, termed photocoagulation.77 How photocoagulation of the TM leads to increased AH outflow is not understood, but the effect is thought to be due to either scarring of the TM or an increase in macrophages that clear debris via phagocytosis.78, 79 SLT is generally considered a safe treatment for IOP reduction as most complications are only present for a short time after the procedure; however, additional medication may need to be used and over time repeated treatment may be required to lower IOP to clinically acceptable levels.77, 78, 80 Trabeculectomy, also called glaucoma filtration surgery, is a third alternative. Due to a high 5-year failure rate81, and complications such as cataract, this surgery is reserved until other treatments have been unsuccessful.82 The surgery creates a fistula for AH to bypass the damaged TM, enabling increased outflow.79 A shunt may be added after trabeculectomy to maintain the opening created by surgery.83, 84 Thus far, shunts appear to decrease the complications associated with trabeculectomy, but these devices are relatively new and lack long-term studies.84 Finally, specific to CAG, a hole in the iris can be created either by a laser iridotomy85 or surgical iridectomy,86 to enable AH outflow despite the closed iridocorneal angle.

NEW THERAPIES IN LATE-STAGE DEVELOPMENT

New drugs under development attempt to leverage our increasingly better understanding of the role eye tissues, particularly the TM87 and optic nerve head (ONH),88 play in glaucoma pathogenesis. As described below, Rho-associated protein kinase (ROCK) inhibitors and adenosine receptor (AR) ligands modulate the pathways involved in both ECM and cytoskeletal restructuring. In addition to lowering of IOP, these molecules appear to increase ONH blood flow or protect against RGC loss.89 The neuroprotective benefits of these molecules are exciting because they are orthogonal to IOP reduction, which, from a clinical perspective, already has inexpensive and successful options (see prior section). The lack of clinical tools with sufficient sensitivity to unequivocally demonstrate neuroprotection is an impediment, however. Currently available retina/optic nerve imaging tools and visual field tests cannot detect the subtle changes that occur at a cellular level early in the disease process, when neuroprotection would be most impactful.90 Delivery of neuroprotective agents to the posterior eye is also an important area of consideration (see also Outlook).

ROCK inhibitors

ROCKs are serine/threonine kinases that are downstream effectors of Rho GTPases. ROCKs phosphorylate substrates involved in regulation of F-actin stress fiber formation, focal adhesion,91, 92 as well as cell-cell adhesion and migration properties.93, 94 Downstream effects on actin cytoskeleton organization and smooth muscle contractility are relevant to many cellular processes, and dysregulation of the ROCK-Rho signaling system is associated with diseases of tissues throughout the body.95, 96 Known ROCK substrates are numerous and include MLC kinase, Lin11, Isl1 and Mec3 Kinase (LIM Kinase), and adducin, among others.93 ROCKs are composed of an N-terminal kinase domain (Figure 2a), a coiled-coil domain with a Rho-binding site, and a pleckstrin homology (PH) domain.96, 97 Upon binding of GTP-activated Rho GTPase, the ROCK autoinhibitory complex between the N-terminal kinase domain and the C-terminal Rho-binding and PH domains is disrupted to enhance ROCK phosphorylation activity.93 There are two ~160 kDa ROCK isozymes, ROCK1 and ROCK2, which share 92% sequence similarity in their kinase domain.97, 98 ROCK1, predominantly expressed in non-neuronal tissues such as the skeletal muscles, heart and lung, and ROCK2, primarily expressed in the brain, phosphorylate the same substrates, suggesting they have at least partially overlapping functions.99 ROCK1 is required for stress fiber and focal adhesion formation, whereas ROCK2 is involved in phagocytosis and formation of the fibronectin fibrillar matrix.100, 101

Figure 2. ROCK inhibition.

Figure 2

(a) Cartoon representation of ROCK-1 bi-lobed kinase domain in complex with R-1 (PDB code 2F2U). Canonical domain features highlighted: Small N-terminal lobe; larger C-terminal lobe; P-loop, phosphate binding loop containing glycine-rich motif; C-loop, catalytic loop; A-loop, activation loop. (b) Zoom into active site and residues within hydrogen bonding distance of fasudil presented in ball-and-stick. (c) Summary of proposed signaling cascade for ROCK inhibition leading to reduced IOP.

The ROCK-Rho signaling system is pertinent to AH outflow and glaucoma.102, 103 Both ROCK isoforms are expressed in tissues throughout the eye including in the TM, ciliary body, corneal epithelium and the retina.104 The selective role of ROCK1 and ROCK2 in glaucoma remains unknown, however. While ROCK1 is highly expressed in the ciliary epithelium, ROCK2 is expressed in the retina,104 and both ROCK1 and ROCK2 knockout mice have lower IOP than wild type mice.105

Development of ROCK inhibitors to treat glaucoma

Modulation of ROCK is an active therapeutic direction to treat cancers,106 cardiovascular disorders,95 metabolic disease,107 neurodegenerative disorders,96 as well as glaucoma (Table 2). As such, considerable effort has been placed in developing selective ROCK inhibitors, and then testing them in potential therapeutic contexts; review articles detailing the extended history of the discovery, improvement, and validation of the ROCK kinases have been written.96, 108111 Briefly, ROCK inhibitors predate the discovery of the ROCK enzyme and were derived from naphthalene-based calmodulin (CaM) antagonists, namely, isoquinoline, which serendipitously inhibits protein kinases at high concentrations.112 The isoquinoline derivative R-1 (HA-1077/fasudil), which has been used clinically in Japan as a vasodilator to prevent cerebral vasospasm,113 and the related R-2 (K-115), have been tested in clinical trials for glaucoma (see below). Like other kinase inhibitor targets, these molecules competitively bind in the ROCK adenosine triphosphate (ATP) binding pocket and thus selectivity of ROCK over other kinases is difficult. For example, R-1 has been shown to inhibit protein kinase C-related protein kinase 2 (PRK2),114 cyclic GMP-dependent protein kinase G (PKG) and protein kinase C (PKC).111 Structures of ROCK1 and ROCK2 in complex with R-1 (Figure 2b) and other inhibitors have revealed nuances in pocket shape and physicochemical properties compared to other kinases,97 leading to compounds with more potent inhibition and specificity than R-1,111, 115 including some that have been tested in glaucoma models116 and are in clinical trials (Table 2).117 Many other newer-generation isoquinoline derivatives with nanomolar IC50 values, better selectivity for ROCK, and promising pharmacokinetics properties118123 have yet to be tested for treating glaucoma, however. Pyridine is a second well-studied ROCK inhibitor scaffold and has likewise been subjected to extensive optimization for potency and selectivity.124126 R-3 (Y-27632), the first selective inhibitor for ROCKs127 and derivatives such as R-4 (Y-39983),128 R-5,126 R-6, and R-7129 have been examined for effects on IOP. Newer scaffolds identified via high throughput screening, such as pyrazole, indazole, aminofurazans, 2,4-diaminopyrimidines, benzodioxanes, urea derivatives and benzothiophenes, have expanded the chemical space for ROCK inhibition.108110, 130, 131 Inhibitors based on these new scaffolds appear promising in other contexts, including overcoming issues of bioavailability,124, 132139 which motivates their consideration for glaucoma. Thus far, treatment with R-8 (SR-3677), a derivative of the pyrazole scaffold,109, 140, 141 improves outflow facility in perfused porcine eyes;141 R-9, a potent and selective ROCK2 inhibitor derived from additional structure-activity-relationship (SAR) studies of the urea scaffold130, lowers IOP in a rat model; R-10, an SAR-based derivative of the benzothiphene scaffold, yielded a comparable reduction in IOP to R-4.131

Table 2.

ROCK inhibitors

Compound (Other Names or Identifiers) Structure Potency (nM) a,b Clinical Trials for IOP reduction
ROCK1 ROCK2
R-1 (Fasudil, HA- 1077) graphic file with name nihms882120t1.jpg 260
320
R-2 (K-115, Ripasudil) graphic file with name nihms882120t2.jpg 51
19
Phase II (Approved for treating glaucoma and ocular hypertension in Japan)
R-3c (Y-27632) graphic file with name nihms882120t3.jpg 200
140
R-4 c (Y-39983, RKI-983, SNJ-1656) graphic file with name nihms882120t4.jpg N/A
3.6
Discontinued after Phase II
R-5 graphic file with name nihms882120t5.jpg 300
78
R-6 graphic file with name nihms882120t6.jpg 346
75
R-7 graphic file with name nihms882120t7.jpg 105
24
R-8c
SR-3677
graphic file with name nihms882120t8.jpg 56
3
R-9 graphic file with name nihms882120t9.jpg N/A
<1
R-10 graphic file with name nihms882120t10.jpg 20
N/A
R-11 Scaffold for AMA-0076 graphic file with name nihms882120t11.jpg N/A
2.5 (reported for compound where X=F
R= CH2-oxolan-2-yl)
Phase II - completed
R-12 (AR-12141 Precursor to AR-12286) graphic file with name nihms882120t12.jpg 6
1.5
AR-12286 in Phase II
R-13 (AR-12432 Precursor to AR-13324) graphic file with name nihms882120t13.jpg 1.2
1.1
AR-13324 in Phase III
a

IC50 values unless otherwise noted.

b

References R-1115, R-2272, R-3109, R-4,119 R-5126, R-6,129 R-7,129 R-8,109, 141 R-9,130 R-10,131 R-11,156 R-12, 157 R-13157.

c

Tested in perfusion studies. N/A: not available.

Proposed IOP-lowering mechanisms (Figure 2c)

ROCK inhibitors have been tested in perfusion studies using porcine, bovine, and rabbit eyes, and in vivo in both ocular normotensive and hypertensive rabbit eyes.94 These studies have repeatedly demonstrated that ROCK inhibitors decrease IOP by increasing AH drainage through the TM via cytoskeletal restructuring.94, 142, 143 For example, treatment of perfused bovine eyes with R-3 leads to changes in cellular morphology of TM and SC cells, decreases the number of focal adhesions, increases permeability of SC cells,144 and widens extracellular spaces in the TM of perfused eyes.142 Such changes have been attributed to a decrease in MLC phosphorylation upon ROCK inhibition.142, 145 Consistent with this model, MLC kinase inhibitors have also been shown to affect the actin cytoskeleton of cultured human TM cells and reduce IOP of rabbit eyes,146 similar to the effects seen with ROCK inhibitors. In addition to inhibitors of MLC kinase, those targeted to LIM kinase have also been shown to reduce IOP.143, 147 ROCK activation of LIM kinase leads to inhibition of actin depolymerization by cofilin.142 Conversely, LIM kinase inhibition, either directly or through upstream inhibition of ROCK, appears to induce TM relaxation via depolymerization of actin filaments.143 Thus, ROCK inhibitors have not only shown promise as glaucoma therapeutics, but studies of their downstream effects have opened up new potential IOP lowering targets, namely, MLC kinase and LIM kinase.

Other potential therapeutic benefits

Besides lowering IOP, ROCK inhibitors are active in the posterior eye. R-1 increases ONH blood flow in rabbits by restoring induced blood flow impairment and related optic nerve damage.148 Similar results were obtained for the R-4 in restoring ONH blood flow, also in rabbits.149 R-1, R-3 and R-4 have been further shown to protect RGCs from apoptosis. R-1 protects the retina of rats from both ischemia/reperfusion induced damage150 and N-methyl-D-aspartate (NMDA) induced excitotoxicity.151 In rats treated with R-4, the number of RGCs with regenerating axons was increased.149 R-3 protected RGCs from ischemia induced damage in rat eyes ex vivo152 and increased RGC regeneration after optic nerve crush both in rats and RGC culture.153 Finally, R-3 prevented postoperative scarring in a glaucoma surgery rabbit model by inhibiting collagen deposition and fibroproliferation, increasing positive surgery outcomes over controls.154

Clinical Trials

With the consistent success of lowering IOP upon treatment in animal models and their neuroprotective effects, several ROCK inhibitors have entered clinical trials (clinicaltrials.gov).94, 109, 155 R-2 (from Kowa Pharmceuticals), is currently in Phase II trials as is the pyridine derivative R-11 (AMA-0076, Amakem).156 R-4 (RKI-983 or SNJ-1656 from Senju and Novartis, respectively) was discontinued during Phase II. Aerie Pharmaceuticals has put forth several isoquinoline derivative ROCK inhibitors R-12 (AR-12286), R-13 (AR-13324), and R-13 combined with the commonly prescribed IOP-reducing prostaglandin analog latanaprost (PG-324). At the time of writing this Perspective, Aerie Pharmaceuticals is recruiting subjects for Phase III trials for R-13.157

Adenosine Receptor Ligands

Adenosine receptors (ARs), GPCRs that regulate adenylyl cyclase and thus affect the production of cAMP, are expressed in tissues throughout the eye including the ciliary body,158 the TM,159 SC,160 and the retina158 and have been implicated in maintenance of IOP.161 At the cellular level, AR classes A1 and A3 deactivate adenylyl cyclase via Gi proteins, and A2A and A2B activate adenylyl cyclase through Gs proteins.162 A1AR and A3AR activate phospholipase C (PLC), which is part of the mitogen-activated protein kinase (MAPK) and the extracellular signal related kinase (ERK1/2) pathway. A1AR activation of ERK1/2 has been shown to lead to secretion of matrix metalloproteinases (MMPs) and accelerate ECM turnover.163 A2AAR and A2BAR-mediated stimulation of cAMP production also modulates ERK1/2; however, this activates connective tissue growth factor (CTGF), increasing ECM deposition.164 Thus, depending on the AR class, agonists or antagonists should elicit the desired effect of lowered IOP (Figure 3a).165 For example, to promote IOP maintenance, enhancement of ECM turnover via an A1AR agonist or a decrease in ECM deposition with an A2AAR antagonist should lower IOP.

Figure 3. AR ligands.

Figure 3

(a) Summary of proposed signaling cascade for AR agonist and antagonist leading to reduced IOP. Dashed arrows imply the possibility of additional intervening steps in the signaling cascade. (b) Left: Cartoon representation of A2AAR with bound AR-2 colored in rainbow from N-terminus (blue) to C-terminus (red). Perpendicular helix 8 (red) may be involved in protein-protein interactions. Right: Zoom into ligand binding pocket and residues within hydrogen bonding distance of AR-2 presented in ball-and-stick. (c) Summary of SAR efforts using adenosine and xanthine scaffolds.

Development of AR ligands to treat glaucoma (Table 3)

Table 3.

AR agonists, antagonists

Compound Function Structure Affinity/Potencya,b (nM) Clinical Trials
AR-1 (Adenosine) Non-selective agonist graphic file with name nihms882120t14.jpg A1 310
A2A 730
A2B 23500
A3 290 (EC50 values)
AR-2 (NECA) Non-selective agonist graphic file with name nihms882120t15.jpg A1 14
A2A 20
A2b 140
A3 25
AR-3 (CHA) A1 agonist graphic file with name nihms882120t16.jpg 1.3–2.3
AR-4 (R-PIA) A1 agonist graphic file with name nihms882120t17.jpg A1 2
A3 16
AR-5 (INO-8875) A1 agonist graphic file with name nihms882120t18.jpg 0.97 Phase I/II
AR-6 (CPA) A1 agonist graphic file with name nihms882120t19.jpg 5.9
AR-7 (CF-101, IB-MECA) A3 agonist graphic file with name nihms882120t20.jpg A3 1.8
A1 51
Phase II
AR-8 (CGS-21680) A2A agonist graphic file with name nihms882120t21.jpg A2A 27
A3 67
AR-9 (ATL-313) A2A agonist graphic file with name nihms882120t22.jpg 0.7 nM (Kd) Phase I
AR-10 (2-O-Ado) A2A agonist graphic file with name nihms882120t23.jpg 31.8
AR-11 (2-CN-Ado) A2A agonist graphic file with name nihms882120t24.jpg 49.3
AR-12 (OPA-6566) A2A agonist N/A N/A Phase II
AR-13 (ZM241385) A2A antagonist graphic file with name nihms882120t25.jpg 1.6
AR-14 (MRS-1097) A3 antagonist graphic file with name nihms882120t26.jpg 108
AR-15 (MRS-1191) A3 antagonist graphic file with name nihms882120t27.jpg 31.4
AR-16 (MRS-1523) A3 antagonist graphic file with name nihms882120t28.jpg 19
AR-17 (LJ-1251) A3 antagonist graphic file with name nihms882120t29.jpg 4.16
AR-18 (MRS-1292) A3 antagonist graphic file with name nihms882120t30.jpg 42 (IC50)
AR-19 (LJ-979) A3 antagonist graphic file with name nihms882120t31.jpg 15.5
AR-20 (MRS-3826) A3 antagonist graphic file with name nihms882120t32.jpg 910
AR-21 (MRS-3827) A3 antagonist graphic file with name nihms882120t33.jpg N/A
AR-22 (MRS-3771) A3 antagonist graphic file with name nihms882120t34.jpg 29
AR-23 (OT-7999) A3 antagonist graphic file with name nihms882120t35.jpg 0.61
a

Ki values unless otherwise noted.

b

Ki/IC50/EC50/Kd value references: AR-1,273 AR-2274, AR-3275, AR-4155, AR-5155, AR-6276, AR-7155, AR-8274, 275 AR-9277, AR-10176, AR-11176, AR-13173, AR-14278, AR-15278, AR-16189, AR-17180, 211, 279, AR-18189, AR-19180, AR-20180, AR-22180, AR-23274 N/A: not available.

AR-1 (adenosine) was linked to heart rate modulation and arterial blood pressure more than 80 years ago166 and soon after was used clinically to treat arrhythmia.167 Ligands for particular AR classes have been developed for decades,168 for a variety of diseases beyond cardiovascular, such as rheumatoid arthritis and Parkinson’s disease,162, 169, 170 by taking advantage of their varying natural affinities for adenosine.169 Because ARs are expressed throughout the body, including in the brain, heart, lung and blood vessels,171 drug development for any particular disorder is fraught with significant side effects. Extensive SAR studies have been completed, guided by the available A2AAR structures (Figure 3b), including those in complex with AR-1 (PDB ID 2YDO),172 as well as synthetic agonists such as 5′-N-ethylcarboxamidoadenosine (AR-2, NECA, Figure 3b) (PDB ID 2YDV)172 and antagonists173.

Most AR agonists shown to decrease IOP are derivatives of AR-1 (Table 3, Figure 3c). In general, compounds with N6-cycloalkyl substitutions favor A1AR (Figure 3c),155, 161, 168 and several including N6-cyclohexyladenosine (AR-3, CHA), (R)-N6-(2-phenyl-1-methylethyl) adenosine (AR-4, R-PIA), AR-5 (INO8875), and N6-cyclopentyladenosine (AR-6, CPA) have been tested for their effect on IOP. A3ARs are better targeted by N6-benzene or N6-substituted benzene ligands (Figure 3c), but to date, only AR-7 (CF-101/IB-MECA) has shown promise for lowering IOP.155, 161, 169, 174, 175 Agonists selective for A2AAR and A2BAR are those typically modified at the C2 position, and agonists selective for A2AARs are further substituted at the 5′ C within the ribose portion of the scaffold (Figure 3c).169, 174, 176 A2AAR agonists demonstrated to lower IOP include AR-8 (CGS-21680), AR-9 (ATL-313), AR-10 (2-O-Ado) and AR-11 (2-CN-Ado). The fewest number of described agonists are those selective for A2BAR, which is less tolerant of substitutions at positions that confer selectivity to other ARs; to the best of our knowledge, available A2BAR agonists have not been studied in the context of glaucoma.165

AR antagonists shown to reduce IOP include one targeted to A2AAR and several to A3AR (Table 3). To date, A1AR antagonists have shown little promise as IOP lowering drugs as they increase IOP in both rabbits and mice, and effects of A2BAR antagonists in the context of IOP and glaucoma have not yet been reported.165 A1, A2A and A2BAR antagonists are largely derivatives of xanthine (Figure 3c), including the natural antagonist caffeine. A3ARs have low affinity for caffeine and other xanthine derivatives, leading to a range of examined scaffolds, which vary in their core structure and include purine and pyridine derivatives, quinazoline derivatives, tricyclic xanthines, and nuceloside derivatives.177180 A2AAR antagonists include xanthine derivatives modified at C8 with styryl groups and nitrogen polyheterocyclic compounds,179 such as AR-13 (ZM241385), which reduces IOP in mice.165. A3AR antagonists tested for their effect on IOP include pyridine derivatives AR-14AR-16, nucleoside derivatives AR-17AR-22, and the tricyclic xanthine derivative AR-23.

Proposed IOP-lowering mechanisms (Figure 3a)

The molecular role(s) of the AR classes in IOP regulation is not well established, as there appears to be both issues of species specificity, as in the case of the A2AR agonist AR-8,161, 181 as well as selectivity issues, as in the case of A1AR agonists AR-3 and AR-4 respectively,168 and is thus an important area for further investigation. Available evidence from studies conducted in bovine and monkey eyes indicates that A1AR agonists lower IOP by increasing outflow182, 183 through the conventional pathway.182 At the molecular level, decreased outflow resistance from activation of A1AR with agonists has been shown to result from enhanced signaling leading to elevated levels of relevant matrix metalloproteases 2 and 9 (MMP-2, MMP-9), 163,182, 184 and thus ECM remodeling.185, 186 Consistent with this finding, outflow increase observed with A1AR activation can be reversed by MMP inhibition.182 A1AR,187 A2AAR, and A3AR agonists have all also been shown to reduce TM cell volume,159 which in turn, independently increases outflow facility.188 A3AR antagonists AR-14, AR-15, and AR-16 prevent chloride release and reduce AH production in NPE cells in the ciliary body.189, 190 Decreased IOP has been documented in studies of mice after treatment with A3AR antagonists as well as after A3AR knockout,190, 191 but not all studies of A3AR converge.165 For example, in human clinical trials as a treatment for dry eye syndrome, the A3AR agonist AR-7, i.e. not an antagonist, was found to lower IOP.192 It is suspected that chronic administration of AR-7 in this study led to lower IOP via the documented “effect inversion” pharmacological mechanism seen for other AR ligands in other contexts,193 or due to inadvertent cross-activation of A1ARs.169

Other potential therapeutic benefits

RGC death in glaucoma is associated with ATP-binding P2X7 purinergic receptors at the cell surface due to an influx of Ca2+194197 and, more controversially, due to prolonged activation of glutamate-binding NMDA and non-NMDA receptors.198200 Activation of A1ARs and A3ARs by adenosine can inhibit the effects of both glutamate- and ATP- induced Ca2+ influx to prevent RGC death.201203 Treatment with adenosine has further been shown to increase ONH blood flow in healthy human eyes,204 and RGC loss has also been attributed to reduced ONH blood flow, as progression of the disease correlates with reduced ONH blood flow in humans205, 206 and in a rhesus monkey model of glaucoma.207 A1 and A2A subtypes were both specifically shown to be involved in ONH blood flow increase in rabbit eyes208 but involvement of A3AR may also be relevant, as in one study, treatment of gerbils with A3AR agonist AR-7 improved post-ischemic neuronal blood flow.209 Finally, there may be a role for A2AAR in neuroprotection by controlling inflammation, analogous to reports related to Alzheimer’s and Parkinson’s disease.210

Clinical trials

As of the writing of this Perspective, selected compounds are in preclinical (AR-17)211, Phase I (AR-9)169, 174, and Phase II (AR-5 155, AR-12, and AR-7) trials (Table 3).155, 165

SELECTED PROMISING NEW DIRECTIONS AND EMERGING TARGETS

Numerous targets are actively being pursued for IOP reduction with potential neuroprotective benefits.212 These include other targets to (a) increase ONH blood flow213, (b) increase TM outflow via ECM remodeling or altering TM cell contractility, (c) increase uveoscleral outflow, and/or (d) decrease AH production. Here we highlight just a few examples of such targets, others are listed in Table 4.

Table 4.

Other therapeutic strategies under consideration.

Drug/target/pathway Proposed Therapeutic Mode of Action
Angiopoietin-like molecules Increase TM outflow through reorganization of the ECM.280
Cannabinoids Decrease AH production and increases TM outflow, also may be neuroprotective.213
Cochlin Regulation may modulate TM outflow.281
CTGF Outflow increase by modification of TM actomyosin cytoskeleton.282, 283
Endothelin-1 Increase TM outflow by relaxation of TM, also shown to be neuroprotective of RGCs.284
Ghrelin Increase TM outflow by relaxation of the TM.285
Latrunculins Increase TM outflow by reducing cell contractility and cell-cell and cell-matrix adhesions. Drugs have reached human trials.286
MMP-2, MMP-9 Increase TM turnover, possible neuron regeneration in retina (see text).
Melatonin Increase TM outflow through adrenoceptor and cholinoceptor modulation.287
Myocilin Decrease of TM cell stress and death (see text).
NGF Protect RGCs from neurodegeneration (see text).
Nitric oxide (NO) Increase TM outflow by relaxation of the TM and ciliary muscle.288 A NO-donating prostaglandin drug has entered clinical trials.289
Transforming Growth Factor-β (TGF-β) Increase TM outflow through ECM remodeling or modification of the actin cytoskeleton.290

Nerve Growth Factor (NGF)

NGF is an emerging treatment for glaucoma214 based on its ability to prevent neural apoptosis, and its clinical evaluation in a number of non-ocular diseases including Alzheimer’s, Parkinson’s, diabetes, and HIV-related neuropathies.215 NGF binds to the cell surface receptor tropomyosin kinase receptor A (TrKA) (Figure 4a), activating a signaling cascade which includes ERK1/2 and, through subsequent upregulation of B-cell lymphoma 2 (Bcl-2), prevents apoptosis.216, 217 Treatment with exogenous NGF has been shown to protect RGCs from death in rats after optic nerve sectioning218 and ischemic injury,219 to reduce retinal damage due to increased IOP in rabbits,220 and to protect RGCs in a rat glaucoma model.221 Interestingly, in a small human trial, three patients with advanced glaucoma treated with NGF-containing eye drops showed improvements in visual field and optic nerve function that lasted for several weeks after treatment, with minimal side effects.221 Results from Phase I trials indicate NGF is safe with limited side effects with ocular administration,222 and, as of the writing of this Perspective, Phase II trials are ongoing for NGF as a dry eye treatment.

Figure 4. Emerging new potential targets.

Figure 4

(a) NGF in complex with TrkA extracellular domain (PDB code 2IFG).268 (b) Full-length pro-MMP-2 (PDB code 1CK7), with domains highlighted.269 (c) myoc-OLF 5-bladed propeller. Top: cartoon representation. Bottom: surface representation with bound glycerol molecules on surface presented as magenta sticks.231 (d) Grp94 overall domain structure (PDB code 2O1U)270 and zoom into N-terminal ATP binding pocket with radamide inhibitor bound (PDB code 2GFD).271

Matrix Metalloproteases

Zinc-dependent matrix metalloproteases are involved in a wide range of functions including bone remodeling, blastocyst implantation, nerve growth, and their namesake, ECM remodeling.223 ECM remodeling, in particular, plays a role in maintaining IOP as MMPs 1, 2, 3, 9 and 14 are all upregulated under high IOP conditions,224 inhibition of MMPs increase outflow resistance,186 and, as discussed earlier, increased activity of both MMP-2 (Figure 4b) and MMP-9 are involved in decreased outflow resistance in the TM.182, 183, 185, 186 Substrates of MMP-1, MMP-2 and MMP-9 include collagens, fibronectin and elastin; whereas MMP-1 is upregulated, MMP-2 and MMP-9 have been found to be reduced in POAG eyes.185 In principle, increasing MMP activity in the TM could be useful as a glaucoma therapy to restore normal outflow through the TM by increasing turnover of the ECM, particularly in steroid induced glaucoma where MMPs are downregulated with prolonged steroid treatment.225 In a gene therapy-based study, induced MMP-1 expression successfully compensated for MMP-1 downregulation caused by steroid administration, and both prevented and reversed steroid-induced ocular hypertension in sheep.226 MMP-1 was secreted to the ECM, degraded collagen type 1 in the TM, and thereby increased outflow.225 There is also some evidence supporting a role for MMPs in neurogenesis,227 neurodegeneration,227, 228 death after neural injury,229 and repair,227 germane to reversing retina degeneration, but molecular details are still forthcoming. Thus, further studies are needed to determine the potential of modulating MMPs for glaucoma in both capacities of lowering IOP and neuroprotection.

Myocilin

Myocilin was the first gene linked with inherited forms of glaucoma13 and is also upregulated in steroid induced glaucoma.14 Missense mutations in myocilin account for ~5% of all POAG and ~10% of all JOAG cases4, 230, and are clustered within its olfactomedin (myoc-OLF) domain.230, 231 Myocilin is expressed in anterior segment tissues including the TM,232, 233. Recent studies have implicated myocilin in cell proliferation and survival through the ERK1/2 pathway, which involves the aforementioned MMPs,234 as well as in programmed RGC cell death.235 While interacting partner(s) of myocilin and precise biological function remains elusive, it is an attractive de novo therapeutic target for glaucoma patients with inherited mutations.

The prevailing mechanism proposed for myocilin pathogenesis caused by mutations within myoc-OLF is that of a gain of function, namely, instead of being secreted to the TM, mutant myocilins aggregate within the ER of TM cells, causing TM cell death. TM cells are highly phagocytotic and clear debris in surrounding extracellular tissue for proper AH filtration.236 Thus, TM cell death is thought to bring about dysregulation of outflow through the TM, leading to elevated IOP, and then glaucoma.4 Disease-causing myoc-OLF variants are less stable than wild type,237 and grow degradation-resistant amyloid-like fibrils in vitro, which have also been detected in cells.238, 239 Notably, the absence of myocilin, either due to the presence of a premature stop codon in humans, or due to its knock-out in mice, does not result in glaucoma,240, 241 suggesting that viable therapeutic strategies would prevent or alleviate aggregation and its toxic effects.

One avenue is to inhibit myocilin misfolding as a means to prevent aggregation and escape detection by ER-associated degradation (ERAD). The result would be the secretion of mutant myocilin, which could restore partial function or take advantage of TM cell phagocytosis for facile degradation. Early studies demonstrated that mutant myocilin secretion could be rescued by culturing TM cells at a lower temperature,242244 presumably because aggregation rates are slowed at temperatures below the unfolding temperature of mutant myocilin. Thus far, attempts to rescue secretion using small molecules have included both chemical and pharmacological chaperone approaches. For the former, chemical chaperones trimethylamine-N-oxide (TMAO)245 and 4-phenylbutyrate (4PBA)246, which is approved to treat urea cycle disorders and is under consideration for other ER-stress inducing misfolding diseases like cystic fibrosis247, improved secretion of mutant myocilin in cell culture. Related, a series of osmolytes were shown to increase thermal stability of mutants to better than wild-type levels in vitro,237, 248 with simple compounds like sucrose exhibiting the most potent stability enhancement. The high concentrations of osmolytes required to enhance the stability and secretion of mutant myocilin limits their usefulness as therapeutics, however. With no known function for myocilin, there is no biochemical assay for the discovery of tailored molecules, so-called pharmacological chaperones, and secondary assays such as cell secretion and inhibition of aggregation are low-throughput, making their use as a primary screening method laborious. One high-throughput, target-independent stability assay was developed recently for myoc-OLF in which the protein is first slightly destabilized with denaturant, and potential ligand scaffolds are then identified by monitoring subsequent re-stabilization using a straightforward fluorescence read-out. From this assay, two compounds were found that restored wild type myoc-OLF thermal stability, inhibited amyloid formation in vitro and increased secretion in a cell model.249 The recent crystal structure of myoc-OLF (Figure 4c) should accelerate the discovery of new selective ligands with similar properties through in silico and rational drug design methods. To this end, several small molecules were found serendipitously to bind to the myoc-OLF surface in the crystal, suggesting the surface is druggable (Figure 4c).231

An alternative method to prevent the intracellular build up of mutant myocilin is to manipulate ERAD-specific molecular chaperones, an approach also under consideration for a number of other diseases.250 For glaucoma, treatment of a Y437H-mutant myocilin mouse model with the aforementioned chemical chaperone 4PBA reduces IOP.251 The targets of 4PBA are most likely chaperones, such as Hsc70,252 Hsp70,253 and glucose regulated protein 78 (Grp78),254 but not myocilin248. The lack of a clear molecular target for 4PBA, however, is a disadvantage for SAR studies to generate more potent and selective molecules. A more directed approach may be to target Grp94 (Figure 4d), which has been shown to recognize mutant myocilin in a tight interaction that abrogates retrotranslocation for degradation by the proteasome.255 Although the reasons for the unproductive interaction are not yet clear, Grp94 increases the rate of myoc-OLF aggregation and co-precipitates with myoc-OLF in vitro.256 In cell culture, if levels of Grp94 are reduced by treatment with RNAi, or activity is inhibited using molecules directed to the N-terminal ATP binding site (Figure 4d), mutant myocilin clearance is promoted by autophagy.255 Incubation of Grp94 with a more selective inhibitor than radamide (see Figure 4d) reestablishes baseline aggregation rates and prevents co-aggregation in vitro.256 From a medicinal chemistry perspective, this proteostasis approach is attractive for several reasons: (1) degradation of myocilin should not cause glaucoma (2) co-crystal structures of Grp94 with inhibitors are tractable, enabling a pathway for development of new molecules and SAR, (3) straightforward secondary assays to assess aggregation in vitro and in cells are accessible, and (4) myocilin-glaucoma mouse models are available for testing optimized compounds. Nevertheless, given that Grp94 levels are investigated in cancer257, 258 and are correlated with apoptosis,259, 260 this approach will require considerable work to prevent such activity and ensure selectivity over other similar chaperones.

OUTLOOK

Progress critical to the development of new glaucoma drugs will continue to require highly interdisciplinary teams composed of individuals spanning fields of expertise from physics to medicine, to biology, biochemistry, and engineering. New targets will emerge as our understanding of normal and pathological aspects of TM biomechanics and molecular details of the mechanisms regulating AH improves. Susceptibility genes found through GWAS and family studies worldwide will also continue to identify new potential targets and research directions. One area of growth for the field is to lower the barrier to testing potential glaucoma drugs with a high likelihood of translation to humans, for example, a three dimensional model of the TM in which changes correlate with IOP reduction;87 until a broadly accepted and robust system is developed, testing molecules for IOP reduction will remain extremely low throughput, relying on perfusion studies or animal models. Besides small molecules, gene therapy with viral vectors and small interfering RNAs (siRNAs),261, 262 analogous to other diseases such as Alzheimer’s and Parkinson’s,263 are also in clinical trials. Facile and accurate methods to evaluate neuroprotection efficacy need to be developed. Finally, patient compliance is critical for therapeutic outcomes in the clinic. This issue is being addressed through advanced delivery methods such as implants, contact lenses, microneedles, and gels,264267 which are being tested with current therapies. In the long term, such systems will offer localized, sustained, delivery of drugs, and better bioavailability in particular tissues, to improve the effectiveness of all classes of glaucoma therapies.

Acknowledgments

Glaucoma-related research in the Lieberman lab has been funded by the Glaucoma Research Foundation, American Health Assistance Foundation (now BrightFocus Foundation), the Pew Scholars Program in Biomedical Sciences, and the National Eye Institute (R01EY021205). R. K. D. was sponsored in part by a Georgia Tech Molecular Biophysics Senior Award.

ABBREVIATIONS

4PBA

4-phenylbutyrate

AH

aqueous humor

ALT

alternative laser trabeculoplasty

AR

adenosine receptor

Bcl-2

B-cell lymphoma 2

CAG

closed-angle glaucoma

CAI

carbonic anhydrase inhibitors

CaM

calmodulin

CTGF

connective tissue growth factor

ECM

extracellular matrix

ERAD

ER-associated degradation

ERK1/2

extracellular signal related kinase

GWAS

genome-wide association studies

IOP

intraocular pressure

JOAG

juvenile open-angle glaucoma

LIM Kinase

Lin11, Isl1 and Mec3 kinase

MLC

myosin light chain

myoc-OLF

myocilin olfactomedin domain

NGF

nerve growth factor

NPE

non-pigmented epithelial

NTG

normal tension glaucoma

OAG

open-angle glaucoma

ONH

optic nerve head

OPTN

optineurin

PEX

pseudoexfoliation

PH

pleckstrin homology

PKG

cyclic GMP-dependent protein kinase G

POAG

primary open-angle glaucoma

PRK2

protein kinase 2

RGC

retinal ganglion cell

ROCK

Rho-associated protein kinase

SAR

structure-activity-relationship

SC

Schlemm’s canal

siRNAs

small interfering RNAs

SLT

selective laser trabeculoplasty

TGFβ

Transforming growth factor-β

TM

trabecular meshwork

TMAO

trimethylamine-N-oxide

TrKA

tropomyosin kinase receptor A

Biographies

Rebecca K. Donegan earned her B.Sc. in chemistry from Middle Tennessee State University and defended her Ph.D. dissertation in April 2015. Her doctoral work under the supervision of Raquel Lieberman included solving the crystal structure and characterizing the calcium binding of the olfactomedin domain of myocilin.

Raquel L. Lieberman received her B. Sc. In Chemistry from Massachusetts Institute of Technology in 1998 and her Ph. D. in Chemistry from Northwestern University in 2005 where she worked with Prof. Amy C. Rosenzweig on biophysical and structural studies of an intramembrane metalloenzyme. After conducting postdoctoral work with Prof. Michael S. Wolfe (Center for Neurological Diseases, Harvard Medical School) and Prof. Gregory A. Petsko (Brandeis) where she expanded to molecular aspects of protein misfolding, she joined the faculty in the School of Chemistry and Biochemistry at Georgia Institute of Technology and is currently an Associate Professor. Research projects in Dr. Lieberman’s lab focus on molecular aspects of proteins implicated in misfolding disorders.

References

  • 1.Alward WL. Medical management of glaucoma. New Engl J Med. 1998;339:1298–1307. doi: 10.1056/NEJM199810293391808. [DOI] [PubMed] [Google Scholar]
  • 2.Casson RJ, Chidlow G, Wood JPM, Crowston JG, Goldberg I. Definition of glaucoma: clinical and experimental concepts. Clin Experiment Ophthalmol. 2012;40:341–349. doi: 10.1111/j.1442-9071.2012.02773.x. [DOI] [PubMed] [Google Scholar]
  • 3.Quigley HA, Broman AT. The number of people with glaucoma worldwide in 2010 and 2020. Br J Ophthalmol. 2006;90:262–267. doi: 10.1136/bjo.2005.081224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Kwon YH, Fingert JH, Kuehn MH, Alward WL. Primary open-angle glaucoma. New Engl J Med. 2009;360:1113–1124. doi: 10.1056/NEJMra0804630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Schlötzer-Schrehardt U. Genetics and genomics of pseudoexfoliation syndrome/glaucoma. Middle East Afr J Ophthalmol. 2011;18:30–36. doi: 10.4103/0974-9233.75882. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Kersey JP, Broadway DC. Corticosteroid-induced glaucoma: a review of the literature. Eye. 2005;20:407–416. doi: 10.1038/sj.eye.6701895. [DOI] [PubMed] [Google Scholar]
  • 7.Vrabec JP, Levin LA. The neurobiology of cell death in glaucoma. Eye. 2007;21:S11–S14. doi: 10.1038/sj.eye.6702880. [DOI] [PubMed] [Google Scholar]
  • 8.Foster PJ, Buhrmann R, Quigley HA, Johnson GJ. The definition and classification of glaucoma in prevalence surveys. Br J Ophthalmol. 2002;86:238–242. doi: 10.1136/bjo.86.2.238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Collaborative Normal-Tension Glaucoma Study Group. The effectiveness of intraocular pressure reduction in the treatment of normal-tension glaucoma. Am J Ophthalmol. 1998;126:498–505. doi: 10.1016/s0002-9394(98)00272-4. [DOI] [PubMed] [Google Scholar]
  • 10.Schwartz K, Budenz D. Current management of glaucoma. Curr Opin Ophthalmol. 2004;15:119–126. doi: 10.1097/00055735-200404000-00011. [DOI] [PubMed] [Google Scholar]
  • 11.Collaborative Normal-Tension Glaucoma Study Group. Comparison of glaucomatous progression between untreated patients with normal-tension glaucoma and patients with therapeutically reduced intraocular pressures. Am J Ophthalmol. 1998;126:487–497. doi: 10.1016/s0002-9394(98)00223-2. [DOI] [PubMed] [Google Scholar]
  • 12.Agis Investigators. The Advanced Glaucoma Intervention Study (AGIS): 7. The relationship between control of intraocular pressure and visual field deterioration. Am J Ophthalmol. 2000;130:429–440. doi: 10.1016/s0002-9394(00)00538-9. [DOI] [PubMed] [Google Scholar]
  • 13.Stone EM, Fingert JH, Alward WL, Nguyen TD, Polansky JR, Sunden SL, Nishimura D, Clark AF, Nystuen A, Nichols BE, Mackey DA, Ritch R, Kalenak JW, Craven ER, Sheffield VC. Identification of a gene that causes primary open angle glaucoma. Science. 1997;275:668–670. doi: 10.1126/science.275.5300.668. [DOI] [PubMed] [Google Scholar]
  • 14.Polansky J, Fauss D, Chen P, Chen H, Lütjen-Drecoll E, Johnson D, Kurtz R, Ma ZD, Bloom E, Nguyen T. Cellular pharmacology and molecular biology of the trabecular meshwork inducible glucocorticoid response gene product. Ophthalmologica. 1997;211:126–139. doi: 10.1159/000310780. [DOI] [PubMed] [Google Scholar]
  • 15.Fan BJ, Wang DY, Lam DSC, Pang CP. Gene mapping for primary open angle glaucoma. Clin Biochem. 2006;39:249–258. doi: 10.1016/j.clinbiochem.2005.11.001. [DOI] [PubMed] [Google Scholar]
  • 16.Allingham R, Liu Y, Rhee DJ. The genetics of primary open-angle glaucoma: a review. Exp Eye Res. 2009;88:837–844. doi: 10.1016/j.exer.2008.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Vasiliou V, Gonzalez FJ. Role of CYP1B1 in glaucoma. Annu Rev Pharmacol Toxicol. 2008;48:333–358. doi: 10.1146/annurev.pharmtox.48.061807.154729. [DOI] [PubMed] [Google Scholar]
  • 18.Aung T, Rezaie T, Okada K, Viswanathan AC, Child AH, Brice G, Bhattacharya SS, Lehmann OJ, Sarfarazi M, Hitchings RA. Clinical features and course of patients with glaucoma with the E50K mutation in the optineurin gene. Invest Ophthalmol Vis Sci. 2005;46:2816–2822. doi: 10.1167/iovs.04-1133. [DOI] [PubMed] [Google Scholar]
  • 19.Rezaie T, Child A, Hitchings R, Brice G, Miller L, Coca-Prados M, Héon E, Krupin T, Ritch R, Kreutzer D. Adult-onset primary open-angle glaucoma caused by mutations in optineurin. Science. 2002;295:1077–1079. doi: 10.1126/science.1066901. [DOI] [PubMed] [Google Scholar]
  • 20.Wolfs RC, Klaver CC, Ramrattan RS, van Duijn CM, Hofman A, de Jong PT. Genetic risk of primary open-angle glaucoma: population-based familial aggregation study. Arch Ophthalmol. 1998;116:1640–1645. doi: 10.1001/archopht.116.12.1640. [DOI] [PubMed] [Google Scholar]
  • 21.Wiggs JL, Hauser MA, Abdrabou W, Allingham RR, Budenz DL, DelBono E, Friedman DS, Kang JH, Gaasterland D, Gaasterland T. The NEIGHBOR consortium primary open-angle glaucoma genome-wide association study: rationale, study design, and clinical variables. J Glaucoma. 2013;22:517–525. doi: 10.1097/IJG.0b013e31824d4fd8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Thorleifsson G, Walters GB, Hewitt AW, Masson G, Helgason A, DeWan A, Sigurdsson A, Jonasdottir A, Gudjonsson SA, Magnusson KP. Common variants near CAV1 and CAV2 are associated with primary open-angle glaucoma. Nat Genet. 2010;42:906–909. doi: 10.1038/ng.661. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Wiggs JL, Kang JH, Yaspan BL, Mirel DB, Laurie C, Crenshaw A, Brodeur W, Gogarten S, Olson LM, Abdrabou W. Common variants near CAV1 and CAV2 are associated with primary open-angle glaucoma in caucasians from the United States. Hum Mol Genet. 2011;20:4707–4713. doi: 10.1093/hmg/ddr382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Goel M, Picciani RG, Lee RK, Bhattacharya SK. Aqueous humor dynamics: a review. Open Ophthalmol J. 2010;4:52–59. doi: 10.2174/1874364101004010052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Do C, Civan M. Basis of chloride transport in ciliary epithelium. J Membr Biol. 2004;200:1–13. doi: 10.1007/s00232-004-0688-5. [DOI] [PubMed] [Google Scholar]
  • 26.Chen S, Inoue R, Inomata H, Ito Y. Role of cyclic AMPp-induced Cl conductance in aqueous humour formation by the dog ciliary epithelium. Br J Pharmacol. 1994;112:1137–1145. doi: 10.1111/j.1476-5381.1994.tb13202.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Do CW, Kong CW, To CH. cAMP inhibits transepithelial chloride secretion across bovine ciliary body/epithelium. Invest Ophthalmol Vis Sci. 2004;45:3638–3643. doi: 10.1167/iovs.03-1343. [DOI] [PubMed] [Google Scholar]
  • 28.Gabelt BAT, Kaufman PL. Changes in aqueous humor dynamics with age and glaucoma. Prog Retin Eye Res. 2005;24:612–637. doi: 10.1016/j.preteyeres.2004.10.003. [DOI] [PubMed] [Google Scholar]
  • 29.Toris CB, Yablonski ME, Wang YL, Camras CB. Aqueous humor dynamics in the aging human eye. Am J Ophthalmol. 1999;127:407–412. doi: 10.1016/s0002-9394(98)00436-x. [DOI] [PubMed] [Google Scholar]
  • 30.Last JA, Pan T, Ding Y, Reilly CM, Keller K, Acott TS, Fautsch MP, Murphy CJ, Russell P. Elastic modulus determination of normal and glaucomatous human trabecular meshwork. Invest Ophthalmol Vis Sci. 2011;52:2147–2152. doi: 10.1167/iovs.10-6342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Stamer WD, Braakman ST, Zhou EH, Ethier CR, Fredberg JJ, Overby DR, Johnson M. Biomechanics of Schlemm’s canal endothelium and intraocular pressure reduction. Prog Retin Eye Res. 2015;44:86–98. doi: 10.1016/j.preteyeres.2014.08.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Tektas OY, Lutjen-Drecoll E. Structural changes of the trabecular meshwork in different kinds of glaucoma. Exp Eye Res. 2009;88:769–775. doi: 10.1016/j.exer.2008.11.025. [DOI] [PubMed] [Google Scholar]
  • 33.Grossniklaus HE, Nickerson JM, Edelhauser HF, Bergman LA, Berglin L. Anatomic alterations in aging and age-related diseases of the eye. Invest Ophthalmol Vis Sci. 2013;54:ORSF23–ORSF27. doi: 10.1167/iovs.13-12711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Weinreb RN, Lindsey JD. The importance of models in glaucoma research. J Glaucoma. 2005;14:302–304. doi: 10.1097/01.ijg.0000169395.47921.02. [DOI] [PubMed] [Google Scholar]
  • 35.McNally S, O’Brien CJ. Drug discovery in glaucoma and the role of animal models. Drug Discov Today Dis Models. 2014;10:e207–e214. [Google Scholar]
  • 36.Bouhenni AR, Dunmire J, Sewell A, Edward DP. Animal models of glaucoma. Biomed Res Int. 2012;2012 doi: 10.1155/2012/692609. Article ID 692609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Kontiola A. A new electromechanical method for measuring intraocular pressure. Doc Ophthalmol. 1997;93:265–276. doi: 10.1007/BF02569066. [DOI] [PubMed] [Google Scholar]
  • 38.Martinez-de-la-Casa JM, Garcia-Feijoo J, Castillo A, Garcia-Sanchez J. Reproducibility and clinical evaluation of rebound tonometry. Invest Ophthalmol Vis Sci. 2005;46:4578–4580. doi: 10.1167/iovs.05-0586. [DOI] [PubMed] [Google Scholar]
  • 39.Cervino A. Rebound tonometry: new opportunities and limitations of non-invasive determination of intraocular pressure. Br J Ophthalmol. 2006;90:1444–1446. doi: 10.1136/bjo.2006.102970. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Hann CR, Vercnocke AJ, Bentley MD, Jorgensen SM, Fautsch MP. Anatomic changes in Schlemm’s canal and collector channels in normal and primary open-angle glaucoma eyes using low and high perfusion pressures. Invest Ophthalmol Vis Sci. 2014;55:5834–5841. doi: 10.1167/iovs.14-14128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Weinreb RN, Khaw PT. Primary open-angle glaucoma. Lancet. 2004;363:1711–1720. doi: 10.1016/S0140-6736(04)16257-0. [DOI] [PubMed] [Google Scholar]
  • 42.Bucolo C, Salomone S, Drago F, Reibaldi M, Longo A, Uva MG. Pharmacological management of ocular hypertension: current approaches and future prospective. Curr Opin Pharm. 2013;13:50–55. doi: 10.1016/j.coph.2012.09.012. [DOI] [PubMed] [Google Scholar]
  • 43.Beidoe G, Mousa SA. Current primary open-angle glaucoma treatments and future directions. Clin Ophthalmol. 2012;6:1699–1707. doi: 10.2147/OPTH.S32933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Bagnis A, Papadia M, Scotto R, Traverso CE. Current and emerging medical therapies in the treatment of glaucoma. Expert Opin Emerg Drugs. 2011;16:293–307. doi: 10.1517/14728214.2011.563733. [DOI] [PubMed] [Google Scholar]
  • 45.Weinreb RN, Aung T, Medeiros FA. The pathophysiology and treatment of glaucoma: a review. JAMA. 2014;311:1901–1911. doi: 10.1001/jama.2014.3192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Realini T. A history of glaucoma pharmacology. Optom Vis Sci. 2011;88:36–38. doi: 10.1097/OPX.0b013e3182058ead. [DOI] [PubMed] [Google Scholar]
  • 47.Wiederholt M, Schäfer R, Wagner U, Lepple-Wienhues A. Contractile response of the isolated trabecular meshwork and ciliary muscle to cholinergic and adrenergic agents. Ger J Ophthalmol. 1996;5:146–153. [PubMed] [Google Scholar]
  • 48.Hoyng PJ, van Beek L. Pharmacological therapy for glaucoma. Drugs. 2000;59:411–434. doi: 10.2165/00003495-200059030-00003. [DOI] [PubMed] [Google Scholar]
  • 49.Arthur S, Cantor LB. Update on the role of alpha-agonists in glaucoma management. Exp Eye Res. 2011;93:271–283. doi: 10.1016/j.exer.2011.04.002. [DOI] [PubMed] [Google Scholar]
  • 50.Remaury A, Larrouy D, Daviaud D, Rouot B, Paris H. Coupling of the alpha 2-adrenergic receptor to the inhibitory G-protein Gi and adenylate cyclase in HT29 cells. Biochem J. 1993;292:283–288. doi: 10.1042/bj2920283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Lim JJ, Liu YH, Khin ESW, Bian JS. Vasoconstrictive effect of hydrogen sulfide involves downregulation of cAMP in vascular smooth muscle cells. Am J Physiol Cell Physiol. 2008;295:C1261–C1270. doi: 10.1152/ajpcell.00195.2008. [DOI] [PubMed] [Google Scholar]
  • 52.Bausher LP, Horio B. Regulation of cyclic AMP production in adult human ciliary processes. Exp Eye Res. 1995;60:43–48. doi: 10.1016/s0014-4835(05)80082-x. [DOI] [PubMed] [Google Scholar]
  • 53.Sears ML, Neufeld AH. Editorial: Adrenergic modulation of the outflow of aqueous humor. Invest Ophthalmol Vis Sci. 1975;14:83–86. [PubMed] [Google Scholar]
  • 54.Reitsamer HA, Posey M, Kiel JW. Effects of a topical α2 adrenergic agonist on ciliary blood flow and aqueous production in rabbits. Exp Eye Res. 2006;82:405–415. doi: 10.1016/j.exer.2005.07.015. [DOI] [PubMed] [Google Scholar]
  • 55.Toris CB, Camras CB, Yablonski ME. Acute versus chronic effects of brimonidine on aqueous humor dynamics in ocular hypertensive patients. Am J Ophthalmol. 1999;128:8–14. doi: 10.1016/s0002-9394(99)00076-8. [DOI] [PubMed] [Google Scholar]
  • 56.Baker JG, Hill SJ, Summers RJ. Evolution of β-blockers: from anti-anginal drugs to ligand-directed signalling. Trends Pharmacol Sci. 2011;32:227–234. doi: 10.1016/j.tips.2011.02.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Prichard B, Gillam P. Use of propranolol (Inderal) in treatment of hypertension. Br Med J. 1964;2:725–727. doi: 10.1136/bmj.2.5411.725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Katz I, Hubbard W, Getson A, Gould A. Intraocular pressure decrease in normal volunteers following timolol ophthalmic solution. Invest Ophthalmol Vis Sci. 1976;15:489–492. [PubMed] [Google Scholar]
  • 59.Marquis RE, Whitson JT. Management of glaucoma: focus on pharmacological therapy. Drugs Aging. 2005;22:1–21. doi: 10.2165/00002512-200522010-00001. [DOI] [PubMed] [Google Scholar]
  • 60.Bromberg BB, Gregory DS, Sears ML. Beta-adrenergic receptors in ciliary processes of the rabbit. Invest Ophthalmol Vis Sci. 1980;19:203–207. [PubMed] [Google Scholar]
  • 61.Phillips C, Howitt G, Rowlands D. Propranolol as ocular hypotensive agent. Br J Ophthalmol. 1967;51:222–226. doi: 10.1136/bjo.51.4.222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Coakes RL, Brubaker RF. The mechanism of timolol in lowering intraocular pressure. In the normal eye. Arch Ophthalmol. 1978;96:2045–2048. doi: 10.1001/archopht.1978.03910060433007. [DOI] [PubMed] [Google Scholar]
  • 63.Zimmerman TJ, Harbin R, Pett M, Kaufman HE. Timolol and facility of outflow. Invest Ophthalmol Vis Sci. 1977;16:623–624. [PubMed] [Google Scholar]
  • 64.Gilbert SBWEM. Beta-adrenergic receptors, from their discovery and characterization through their manipulation to beneficial clinical application. Cardiology. 2012;122:104–112. doi: 10.1159/000339271. [DOI] [PubMed] [Google Scholar]
  • 65.McLaughlin CW, Peart D, Purves RD, Carré DA, Peterson-Yantorno K, Mitchell CH, Macknight AD, Civan MM. Timolol may inhibit aqueous humor secretion by cAMP-independent action on ciliary epithelial cells. Am J Physiol Cell Physiol. 2001;281:C865–C875. doi: 10.1152/ajpcell.2001.281.3.C865. [DOI] [PubMed] [Google Scholar]
  • 66.Neufeld AH. Experimental studies on the mechanism of action of timolol. Surv Ophthalmol. 1979;23:363–370. doi: 10.1016/0039-6257(79)90229-7. [DOI] [PubMed] [Google Scholar]
  • 67.Cracknell KPB, Grierson I. Prostaglandin analogues in the anterior eye: Their pressure lowering action and side effects. Exp Eye Res. 2009;88:786–791. doi: 10.1016/j.exer.2008.08.022. [DOI] [PubMed] [Google Scholar]
  • 68.Ricciotti E, FitzGerald GA. Prostaglandins and inflammation. Atertio Thromb Vasc Biol. 2011;31:986–1000. doi: 10.1161/ATVBAHA.110.207449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Von Euler U. On the specific vaso-dilating and plain muscle stimulating substances from accessory genital glands in man and certain animals (prostaglandin and vesiglandin) J Physiol. 1936;88:213–234. doi: 10.1113/jphysiol.1936.sp003433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Nilsson SF, Drecoll E, Lütjen-Drecoll E, Toris CB, Krauss AHP, Kharlamb A, Nieves A, Guerra T, Woodward DF. The prostanoid EP2 receptor agonist butaprost increases uveoscleral outflow in the cynomolgus monkey. Invest Ophthalmol Vis Sci. 2006;47:4042–4049. doi: 10.1167/iovs.05-1627. [DOI] [PubMed] [Google Scholar]
  • 71.Weinreb RN, Toris CB, Gabelt BAT, Lindsey JD, Kaufman PL. Effects of prostaglandins on the aqueous humor outflow pathways. Surv Ophthalmol. 2002;47(Supplement 1):S53–S64. doi: 10.1016/s0039-6257(02)00306-5. [DOI] [PubMed] [Google Scholar]
  • 72.Toris CB, Yablonski ME, Wang YL, Hayashi M. Prostaglandin A2 increases uveoscleral outflow and trabecular outflow facility in the cat. Exp Eye Res. 1995;61:649–657. doi: 10.1016/s0014-4835(05)80015-6. [DOI] [PubMed] [Google Scholar]
  • 73.Hinz B, Rösch S, Ramer R, Tamm ER, Brune K. Latanoprost induces matrix metalloproteinase-1 expression in human nonpigmented ciliary epithelial cells through a cyclooxygenase-2-dependent mechanism. FASEB J. 2005;19:1929–1931. doi: 10.1096/fj.04-3626fje. [DOI] [PubMed] [Google Scholar]
  • 74.Lindsey JD, Kashiwagi K, Kashiwagi F, Weinreb RN. Prostaglandins alter extracellular matrix adjacent to human ciliary muscle cells in vitro. Invest Ophthalmol Vis Sci. 1997;38:2214–2223. [PubMed] [Google Scholar]
  • 75.Ocklind A. Effect of latanoprost on the extracellular matrix of the ciliary muscle. A study on cultured cells and tissue sections. Exp Eye Res. 1998;67:179–191. doi: 10.1006/exer.1998.0508. [DOI] [PubMed] [Google Scholar]
  • 76.Zhao X, Pearson KE, Stephan DA, Russell P. Effects of prostaglandin analogues on human ciliary muscle and trabecular meshwork cells. Invest Ophthalmol Vis Sci. 2003;44:1945–1952. doi: 10.1167/iovs.02-0920. [DOI] [PubMed] [Google Scholar]
  • 77.Melamed S, Simon GJB, Levkovitch-Verbin H. Selective laser trabeculoplasty as primary treatment for open-angle glaucoma: a prospective, nonrandomized pilot study. Arch Ophthalmol. 2003;121:957–960. doi: 10.1001/archopht.121.7.957. [DOI] [PubMed] [Google Scholar]
  • 78.Damji KF, Shah KC, Rock WJ, Bains HS, Hodge WG. Selective laser trabeculoplasty vargon laser trabeculoplasty: a prospective randomised clinical trial. Br J Ophthalmol. 1999;83:718–722. doi: 10.1136/bjo.83.6.718. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Johnson DH, Johnson M. How does nonpenetrating glaucoma surgery work? Aqueous outflow resistance and glaucoma surgery. J Glaucoma. 2001;10:55–67. doi: 10.1097/00061198-200102000-00011. [DOI] [PubMed] [Google Scholar]
  • 80.Ayala M, Chen E. Comparison of selective laser trabeculoplasty (SLT) in primary open angle glaucoma and pseudoexfoliation glaucoma. Clin Ophthalmol. 2011;5:1469–1673. doi: 10.2147/OPTH.S25636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Gedde SJ, Schiffman JC, Feuer WJ, Herndon LW, Brandt JD, Budenz DL. Treatment outcomes in the tube versus trabeculectomy (TVT) study after five years of follow-up. Am J Ophthalmol. 2012;153:789–803. e782. doi: 10.1016/j.ajo.2011.10.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Edmunds B, Thompson J, Salmon J, Wormald R. The national survey of trabeculectomy. III. Early and late complications. Eye. 2002;16:297–303. doi: 10.1038/sj.eye.6700148. [DOI] [PubMed] [Google Scholar]
  • 83.Spiegel D, Kobuch K. Trabecular meshwork bypass tube shunt: initial case series. Br J Ophthalmol. 2002;86:1228–1231. doi: 10.1136/bjo.86.11.1228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Francis BA, Singh K, Lin SC, Hodapp E, Jampel HD, Samples JR, Smith SD. Novel glaucoma procedures: a report by the American Academy of Ophthalmology. Ophthalmology. 2011;118:1466–1480. doi: 10.1016/j.ophtha.2011.03.028. [DOI] [PubMed] [Google Scholar]
  • 85.Nolan WP, Foster PJ, Devereux JG, Uranchimeg D, Johnson GJ, Baasanhu J. YAG laser iridotomy treatment for primary angle closure in east Asian eyes. Br J Ophthalmol. 2000;84:1255–1259. doi: 10.1136/bjo.84.11.1255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Playfair TJ, Watson PG. Management of acute primary angle-closure glaucoma: a long-term follow-up of the results of peripheral iridectomy used as an initial procedure. Br J Ophthalmol. 1979;63:17–22. doi: 10.1136/bjo.63.1.17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Dautriche CN, Xie Y, Sharfstein ST. Walking through trabecular meshwork biology: Toward engineering design of outflow physiology. Biotechnol Adv. 2014;32:971–983. doi: 10.1016/j.biotechadv.2014.04.012. [DOI] [PubMed] [Google Scholar]
  • 88.Yamamoto T, Kitazawa Y. Vascular pathogenesis of normal-tension glaucoma: a possible pathogenetic factor, other than intraocular pressure, of glaucomatous optic neuropathy. Prog Retin Eye Res. 1998;17:127–143. doi: 10.1016/s1350-9462(97)00009-8. [DOI] [PubMed] [Google Scholar]
  • 89.Sena DF, Lindsley K. Neuroprotection for treatment of glaucoma in adults. Cochrane Database Syst Rev. 2013;2:CD006539. doi: 10.1002/14651858.CD006539.pub3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Chua B, Goldberg I. Neuroprotective agents in glaucoma therapy: recent developments and future directions. Expert Rev Ophthalmol. 2010;5:627–636. [Google Scholar]
  • 91.Leung T, Chen XQ, Manser E, Lim L. The p160 RhoA-binding kinase ROK alpha is a member of a kinase family and is involved in the reorganization of the cytoskeleton. Mol Cell Biol. 1996;16:5313–5327. doi: 10.1128/mcb.16.10.5313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Amano M, Ito M, Kimura K, Fukata Y, Chihara K, Nakano T, Matsuura Y, Kaibuchi K. Phosphorylation and activation of myosin by Rho-associated kinase (Rho-kinase) J Biol Chem. 1996;271:20246–20249. doi: 10.1074/jbc.271.34.20246. [DOI] [PubMed] [Google Scholar]
  • 93.Riento K, Ridley AJ. Rocks: multifunctional kinases in cell behaviour. Nat Rev Mol Cell Biol. 2003;4:446–456. doi: 10.1038/nrm1128. [DOI] [PubMed] [Google Scholar]
  • 94.Wang SK, Chang RT. An emerging treatment option for glaucoma: Rho kinase inhibitors. Clin Ophthalmol. 2014;8:883–890. doi: 10.2147/OPTH.S41000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Satoh K, Fukumoto Y, Shimokawa H. Rho-kinase: important new therapeutic target in cardiovascular diseases. Am J Physiol Heart Circ Physiol. 2011;301:H287–H296. doi: 10.1152/ajpheart.00327.2011. [DOI] [PubMed] [Google Scholar]
  • 96.Mueller BK, Mack H, Teusch N. Rho kinase, a promising drug target for neurological disorders. Nat Rev Drug Discov. 2005;4:387–398. doi: 10.1038/nrd1719. [DOI] [PubMed] [Google Scholar]
  • 97.Jacobs M, Hayakawa K, Swenson L, Bellon S, Fleming M, Taslimi P, Doran J. The structure of dimeric ROCK I reveals the mechanism for ligand selectivity. J Biol Chem. 2006;281:260–268. doi: 10.1074/jbc.M508847200. [DOI] [PubMed] [Google Scholar]
  • 98.Nakagawa O, Fujisawa K, Ishizaki T, Saito Y, Nakao K, Narumiya S. ROCK-I and ROCK-II, two isoforms of Rho-associated coiled-coil forming protein serine/threonine kinase in mice. FEBS Lett. 1996;392:189–193. doi: 10.1016/0014-5793(96)00811-3. [DOI] [PubMed] [Google Scholar]
  • 99.Hahmann C, Schroeter T. Rho-kinase inhibitors as therapeutics: from pan inhibition to isoform selectivity. Cell Mol Life Sci. 2010;67:171–177. doi: 10.1007/s00018-009-0189-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Yoneda A, Multhaupt HAB, Couchman JR. The Rho kinases I and II regulate different aspects of myosin II activity. J Cell Biol. 2005;170:443–453. doi: 10.1083/jcb.200412043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Yoneda A, Ushakov D, Multhaupt HA, Couchman JR. Fibronectin matrix assembly requires distinct contributions from Rho kinases I and-II. Mol Biol Cell. 2007;18:66–75. doi: 10.1091/mbc.E06-08-0684. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Kjøller L, Hall A. Signaling to Rho GTPases. Exp Cell Res. 1999;253:166–179. doi: 10.1006/excr.1999.4674. [DOI] [PubMed] [Google Scholar]
  • 103.Fukata Y, Kaibuchi K, Amano M. Rho–Rho-kinase pathway in smooth muscle contraction and cytoskeletal reorganization of non-muscle cells. Trends Pharmacol Sci. 2001;22:32–39. doi: 10.1016/s0165-6147(00)01596-0. [DOI] [PubMed] [Google Scholar]
  • 104.Fukiage C, Mizutani K, Kawamoto Y, Azuma M, Shearer TR. Involvement of phosphorylation of myosin phosphatase by ROCK in trabecular meshwork and ciliary muscle contraction. Biochem Biophys Res Commun. 2001;288:296–300. doi: 10.1006/bbrc.2001.5751. [DOI] [PubMed] [Google Scholar]
  • 105.Whitlock NA, Harrison B, Mixon T, Yu XQ, Wilson A, Gerhardt B, Eberhart DE, Abuin A, Rice DS. Decreased intraocular pressure in mice following either pharmacological or genetic inhibition of ROCK. J Ocul Pharmacol Ther. 2009;25:187–194. doi: 10.1089/jop.2008.0142. [DOI] [PubMed] [Google Scholar]
  • 106.Aznar S, Fernández-Valerón P, Espina C, Lacal JC. Rho GTPases: potential candidates for anticancer therapy. Cancer Lett. 2004;206:181–191. doi: 10.1016/j.canlet.2003.08.035. [DOI] [PubMed] [Google Scholar]
  • 107.Noda K, Nakajima S, Godo S, Saito H, Ikeda S, Shimizu T, Enkhjargal B, Fukumoto Y, Tsukita S, Yamada T, Katagiri H, Shimokawa H. Rho-kinase inhibition ameliorates metabolic disorders through activation of AMPK pathway in mice. PLoS ONE. 2014;9:e110446. doi: 10.1371/journal.pone.0110446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Guan R, Xu X, Chen M, Hu H, Ge H, Wen S, Zhou S, Pi R. Advances in the studies of roles of Rho/Rho-kinase in diseases and the development of its inhibitors. Eur J Med Chem. 2013;70:613–622. doi: 10.1016/j.ejmech.2013.10.048. [DOI] [PubMed] [Google Scholar]
  • 109.Pan P, Shen M, Yu H, Li Y, Li D, Hou T. Advances in the development of Rho-associated protein kinase (ROCK) inhibitors. Drug Discov Today. 2013;18:1323–1333. doi: 10.1016/j.drudis.2013.09.010. [DOI] [PubMed] [Google Scholar]
  • 110.LoGrasso PV, Feng Y. Rho kinase (ROCK) inhibitors and their application to inflammatory disorders. Curr Top Med Chem. 2009;9:704–723. doi: 10.2174/156802609789044452. [DOI] [PubMed] [Google Scholar]
  • 111.Tamura M, Nakao H, Yoshizaki H, Shiratsuchi M, Shigyo H, Yamada H, Ozawa T, Totsuka J, Hidaka H. Development of specific Rho-kinase inhibitors and their clinical application. Biochim Biophys Acta. 2005;1754:245–252. doi: 10.1016/j.bbapap.2005.06.015. [DOI] [PubMed] [Google Scholar]
  • 112.Hidaka H, Inagaki M, Kawamoto S, Sasaki Y. Isoquinolinesulfonamides, novel and potent inhibitors of cyclic nucleotide-dependent protein kinase and protein kinase C. Biochemistry. 1984;23:5036–5041. doi: 10.1021/bi00316a032. [DOI] [PubMed] [Google Scholar]
  • 113.Ono-Saito N, Niki I, Hidaka H. H-Series protein kinase inhibitors and potential clinical applications. Pharmacol Ther. 1999;82:123–131. doi: 10.1016/s0163-7258(98)00070-9. [DOI] [PubMed] [Google Scholar]
  • 114.Davies S, Reddy H, Caivano M, Cohen P. Specificity and mechanism of action of some commonly used protein kinase inhibitors. Biochem J. 2000;351:95–105. doi: 10.1042/0264-6021:3510095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Shibuya M, Hirai S, Seto M, Satoh S-i, Ohtomo E Group FISS. Effects of fasudil in acute ischemic stroke: results of a prospective placebo-controlled double-blind trial. J Neurol Sci. 2005;238:31–39. doi: 10.1016/j.jns.2005.06.003. [DOI] [PubMed] [Google Scholar]
  • 116.Sumi K, Inoue Y, Nishio M, Naito Y, Hosoya T, Suzuki M, Hidaka H. IOP-lowering effect of isoquinoline-5-sulfonamide compounds in ocular normotensive monkeys. Bioorg Med Chem Lett. 2014;24:831–834. doi: 10.1016/j.bmcl.2013.12.085. [DOI] [PubMed] [Google Scholar]
  • 117.Yamamoto K, Maruyama K, Himori N, Omodaka K, Yokoyama Y, Shiga Y, Morin R, Nakazawa T. The novel Rho kinase (ROCK) inhibitor K-115: A new candidate drug for neuroprotective treatment in glaucoma. Invest Ophthalmol Vis Sci. 2014;55:7126–7136. doi: 10.1167/iovs.13-13842. [DOI] [PubMed] [Google Scholar]
  • 118.Lavogina D, Kalind K, Bredihhina J, Hurt M, Vaasa A, Kasari M, Enkvist E, Raidaru G, Uri A. Conjugates of 5-isoquinolinesulfonylamides and oligo-D-arginine possess high affinity and selectivity towards Rho kinase (ROCK) Bioorg Med Chem Lett. 2012;22:3425–3430. doi: 10.1016/j.bmcl.2012.03.101. [DOI] [PubMed] [Google Scholar]
  • 119.Ray P, Wright J, Adam J, Bennett J, Boucharens S, Black D, Cook A, Brown AR, Epemolu O, Fletcher D, Haunso A, Huggett M, Jones P, Laats S, Lyons A, Mestres J, de Man J, Morphy R, Rankovic Z, Sherborne B, Sherry L, van Straten N, Westwood P, Zaman GZR. Fragment-based discovery of 6-substituted isoquinolin-1-amine based ROCK-I inhibitors. Bioorg Med Chem Lett. 2011;21:97–101. doi: 10.1016/j.bmcl.2010.11.060. [DOI] [PubMed] [Google Scholar]
  • 120.Iwakubo M, Takami A, Okada Y, Kawata T, Tagami Y, Sato M, Sugiyama T, Fukushima K, Taya S, Amano M, Kaibuchi K, Iijima H. Design and synthesis of Rho kinase inhibitors (III) Bioorg Med Chem. 2007;15:1022–1033. doi: 10.1016/j.bmc.2006.10.028. [DOI] [PubMed] [Google Scholar]
  • 121.Wu F, Büttner FH, Chen R, Hickey E, Jakes S, Kaplita P, Kashem MA, Kerr S, Kugler S, Paw Z. Substituted 2H-isoquinolin-1-one as potent Rho-Kinase inhibitors. Part 1: Hit-to-lead account. Bioorg Med Chem Lett. 2010;20:3235–3239. doi: 10.1016/j.bmcl.2010.04.070. [DOI] [PubMed] [Google Scholar]
  • 122.Ginn JD, Bosanac T, Chen R, Cywin C, Hickey E, Kashem M, Kerr S, Kugler S, Li X, Prokopowicz A. Substituted 2H-isoquinolin-1-ones as potent Rho-kinase inhibitors: part 2, optimization for blood pressure reduction in spontaneously hypertensive rats. Bioorg Med Chem Lett. 2010;20:5153–5156. doi: 10.1016/j.bmcl.2010.07.014. [DOI] [PubMed] [Google Scholar]
  • 123.Bosanac T, Hickey ER, Ginn J, Kashem M, Kerr S, Kugler S, Li X, Olague A, Schlyer S, Young ER. Substituted 2H-isoquinolin-1-ones as potent Rho-kinase inhibitors: part 3, aryl substituted pyrrolidines. Bioorg Med Chem Lett. 2010;20:3746–3749. doi: 10.1016/j.bmcl.2010.04.069. [DOI] [PubMed] [Google Scholar]
  • 124.Li R, Martin MP, Liu Y, Wang B, Patel RA, Zhu JY, Sun N, Pireddu R, Lawrence NJ, Li J, Haura EB, Sung SS, Guida WC, Schonbrunn E, Sebti SM. Fragment-based and structure-guided discovery and optimization of Rho kinase inhibitors. J Med Chem. 2012;55:2474–2478. doi: 10.1021/jm201289r. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Ho KK, Beasley JR, Belanger L, Black D, Chan JH, Dunn D, Hu B, Klon A, Kultgen SG, Ohlmeyer M. Triazine and pyrimidine based ROCK inhibitors with efficacy in spontaneous hypertensive rat model. Bioorg Med Chem Lett. 2009;19:6027–6031. doi: 10.1016/j.bmcl.2009.09.046. [DOI] [PubMed] [Google Scholar]
  • 126.Henderson AJ, Hadden M, Guo C, Douglas N, Decornez H, Hellberg MR, Rusinko A, McLaughlin M, Sharif N, Drace C. 2, 3-Diaminopyrazines as Rho kinase inhibitors. Bioorg Med Chem Lett. 2010;20:1137–1140. doi: 10.1016/j.bmcl.2009.12.012. [DOI] [PubMed] [Google Scholar]
  • 127.Uehata M, Ishizaki T, Satoh H, Ono T, Kawahara T, Morishita T, Tamakawa H, Yamagami K, Inui J, Maekawa M, Narumiya S. Calcium sensitization of smooth muscle mediated by a Rho-associated protein kinase in hypertension. Nature. 1997;389:990–994. doi: 10.1038/40187. [DOI] [PubMed] [Google Scholar]
  • 128.Tokushige H, Inatani M, Nemoto S, Sakaki H, Katayama K, Uehata M, Tanihara H. Effects of topical administration of Y-39983, a selective Rho-associated protein kinase inhibitor, on ocular tissues in rabbits and monkeys. Invest Ophthalmol Vis Sci. 2007;48:3216–3222. doi: 10.1167/iovs.05-1617. [DOI] [PubMed] [Google Scholar]
  • 129.Chen HH, Namil A, Severns B, Ward J, Kelly C, Drace C, McLaughlin MA, Yacoub S, Li B, Patil R. In vivo optimization of 2, 3-diaminopyrazine Rho kinase inhibitors for the treatment of glaucoma. Bioorg Med Chem Lett. 2014;24:1875–1879. doi: 10.1016/j.bmcl.2014.03.017. [DOI] [PubMed] [Google Scholar]
  • 130.Yin Y, Cameron MD, Lin L, Khan S, Schröter T, Grant W, Pocas J, Chen YT, Schürer S, Pachori A. Discovery of potent and selective urea-based ROCK inhibitors and their effects on intraocular pressure in rats. ACS Med Chem Lett. 2010;1:175–179. doi: 10.1021/ml1000382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Davis RL, Kahraman M, Prins TJ, Beaver Y, Cook TG, Cramp J, Cayanan CS, Gardiner EMM, McLaughlin MA, Clark AF, Hellberg MR, Shiau AK, Noble SA, Borchardt AJ. Benzothiophene containing Rho kinase inhibitors: efficacy in an animal model of glaucoma. Bioorg Med Chem Lett. 2010;20:3361–3366. doi: 10.1016/j.bmcl.2010.04.020. [DOI] [PubMed] [Google Scholar]
  • 132.Feng Y, Cameron MD, Frackowiak B, Griffin E, Lin L, Ruiz C, Schröter T, LoGrasso P. Structure–activity relationships, and drug metabolism and pharmacokinetic properties for indazole piperazine and indazole piperidine inhibitors of ROCK-II. Bioorg Med Chem Lett. 2007;17:2355–2360. doi: 10.1016/j.bmcl.2006.12.043. [DOI] [PubMed] [Google Scholar]
  • 133.Goodman KB, Cui H, Dowdell SE, Gaitanopoulos DE, Ivy RL, Sehon CA, Stavenger RA, Wang GZ, Viet AQ, Xu W. Development of dihydropyridone indazole amides as selective Rho-kinase inhibitors. J Med Chem. 2007;50:6–9. doi: 10.1021/jm0609014. [DOI] [PubMed] [Google Scholar]
  • 134.Sehon CA, Wang GZ, Viet AQ, Goodman KB, Dowdell SE, Elkins PA, Semus SF, Evans C, Jolivette LJ, Kirkpatrick RB. Potent, selective and orally bioavailable dihydropyrimidine inhibitors of Rho kinase (ROCK1) as potential therapeutic agents for cardiovascular diseases. J Med Chem. 2008;51:6631–6634. doi: 10.1021/jm8005096. [DOI] [PubMed] [Google Scholar]
  • 135.Shaw D, Hollingworth G, Soldermann N, Sprague E, Schuler W, Vangrevelinghe E, Duggan N, Thomas M, Kosaka T, Waters N, van Eis MJ. Novel ROCK inhibitors for the treatment of pulmonary arterial hypertension. Bioorg Med Chem Lett. 2014;24:4812–4817. doi: 10.1016/j.bmcl.2014.09.002. [DOI] [PubMed] [Google Scholar]
  • 136.Stavenger RA, Cui H, Dowdell SE, Franz RG, Gaitanopoulos DE, Goodman KB, Hilfiker MA, Ivy RL, Leber JD, Marino JP. Discovery of aminofurazan-azabenzimidazoles as inhibitors of Rho-kinase with high kinase selectivity and antihypertensive activity. J Med Chem. 2007;50:2–5. doi: 10.1021/jm060873p. [DOI] [PubMed] [Google Scholar]
  • 137.Pireddu R, Forinash K, Sun NN, Martin MP, Sung SS, Alexander B, Zhu JY, Guida WC, Schönbrunn E, Sebti SM. Pyridylthiazole-based ureas as inhibitors of Rho-associated protein kinases. Cancer Res. 2012;72:3904–3904. doi: 10.1039/C2MD00320A. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Patel RA, Forinash KD, Pireddu R, Sun Y, Sun N, Martin MP, Schönbrunn E, Lawrence NJ, Sebti SM. RKI-1447 is a potent inhibitor of the Rho-associated ROCK kinases with anti-invasive and antitumor activities in breast cancer. Cancer Res. 2012;72:5025–5034. doi: 10.1158/0008-5472.CAN-12-0954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Schirok H, Kast R, Figueroa-Pérez S, Bennabi S, Gnoth MJ, Feurer A, Heckroth H, Thutewohl M, Paulsen H, Knorr A. Design and synthesis of potent and selective azaindole-based Rho kinase (ROCK) inhibitors. ChemMedChem. 2008;3:1893–1904. doi: 10.1002/cmdc.200800211. [DOI] [PubMed] [Google Scholar]
  • 140.Chen YT, Bannister TD, Weiser A, Griffin E, Lin L, Ruiz C, Cameron MD, Schürer S, Duckett D, Schröter T, LoGrasso P, Feng Y. Chroman-3-amides as potent Rho kinase inhibitors. Bioorg Med Chem Lett. 2008;18:6406–6409. doi: 10.1016/j.bmcl.2008.10.080. [DOI] [PubMed] [Google Scholar]
  • 141.Feng Y, Yin Y, Weiser A, Griffin E, Cameron MD, Lin L, Ruiz C, Schürer SC, Inoue T, Rao PV. Discovery of substituted 4-(pyrazol-4-yl)-phenylbenzodioxane-2-carboxamides as potent and highly selective Rho kinase (ROCK-II) inhibitors. J Med Chem. 2008;51:6642–6645. doi: 10.1021/jm800986w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Rao PV, Deng PF, Kumar J, Epstein DL. Modulation of aqueous humor outflow facility by the Rho kinase–specific inhibitor Y-27632. Invest Ophthalmol Vis Sci. 2001;42:1029–1037. [PubMed] [Google Scholar]
  • 143.Harrison BA, Almstead ZY, Burgoon H, Gardyan M, Goodwin NC, Healy J, Liu Y, Mabon R, Marinelli B, Samala L. Discovery and development of LX7101, a dual LIM-kinase and ROCK inhibitor for the treatment of glaucoma. ACS Med Chem Lett. 2014;6:84–88. doi: 10.1021/ml500367g. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Lu Z, Overby DR, Scott PA, Freddo TF, Gong H. The mechanism of increasing outflow facility by Rho-kinase inhibition with Y-27632 in bovine eyes. Exp Eye Res. 2008;86:271–281. doi: 10.1016/j.exer.2007.10.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Rao PV, Deng P, Sasaki Y, Epstein DL. Regulation of myosin light chain phosphorylation in the trabecular meshwork: role in aqueous humour outflow facility. Exp Eye Res. 2005;80:197–206. doi: 10.1016/j.exer.2004.08.029. [DOI] [PubMed] [Google Scholar]
  • 146.Honjo M, Inatani M, Kido N, Sawamura T, Yue BYJT, Honda Y, Tanihara H. A myosin light chain kinase inhibitor, ML-9, lowers the intraocular pressure in rabbit eyes. Exp Eye Res. 2002;75:135–142. doi: 10.1006/exer.2002.2009. [DOI] [PubMed] [Google Scholar]
  • 147.Harrison BA, Whitlock NA, Voronkov MV, Almstead ZY, Gu K-j, Mabon R, Gardyan M, Hamman BD, Allen J, Gopinathan S. Novel class of LIM-kinase 2 inhibitors for the treatment of ocular hypertension and associated glaucoma. J Med Chem. 2009;52:6515–6518. doi: 10.1021/jm901226j. [DOI] [PubMed] [Google Scholar]
  • 148.Sugiyama T, Shibata M, Kajiura S, Okuno T, Tonari M, Oku H, Ikeda T. Effects of fasudil, a Rho-associated protein kinase inhibitor, on optic nerve head blood flow in rabbits. Invest Ophthalmol Vis Sci. 2011;52:64–69. doi: 10.1167/iovs.10-5265. [DOI] [PubMed] [Google Scholar]
  • 149.Tokushige H, Waki M, Takayama Y, Tanihara H. Effects of Y-39983, a selective Rho-associated protein kinase inhibitor, on blood flow in optic nerve head in rabbits and axonal regeneration of retinal ganglion cells in rats. Curr Eye Res. 2011;36:964–970. doi: 10.3109/02713683.2011.599106. [DOI] [PubMed] [Google Scholar]
  • 150.Song H, Gao D. Fasudil, a Rho-associated protein kinase inhibitor, attenuates retinal ischemia and reperfusion injury in rats. Int J Mol Med. 2011;28:193–198. doi: 10.3892/ijmm.2011.659. [DOI] [PubMed] [Google Scholar]
  • 151.Kitaoka Y, Kitaoka Y, Kumai T, Lam TT, Kuribayashi K, Isenoumi K, Munemasa Y, Motoki M, Kobayashi S, Ueno S. Involvement of RhoA and possible neuroprotective effect of fasudil, a Rho kinase inhibitor, in NMDA-induced neurotoxicity in the rat retina. Brain Res. 2004;1018:111–118. doi: 10.1016/j.brainres.2004.05.070. [DOI] [PubMed] [Google Scholar]
  • 152.Hirata A, Inatani M, Inomata Y, Yonemura N, Kawaji T, Honjo M, Tanihara H. Y-27632, a Rho-associated protein kinase inhibitor, attenuates neuronal cell death after transient retinal ischemia. Graefes Arch Clin Exp Ophthalmol. 2008;246:51–59. doi: 10.1007/s00417-007-0666-6. [DOI] [PubMed] [Google Scholar]
  • 153.Lingor P, Tönges L, Pieper N, Bermel C, Barski E, Planchamp V, Bähr M. ROCK inhibition and CNTF interact on intrinsic signalling pathways and differentially regulate survival and regeneration in retinal ganglion cells. Brain. 2008;131:250–263. doi: 10.1093/brain/awm284. [DOI] [PubMed] [Google Scholar]
  • 154.Honjo M, Tanihara H, Kameda T, Kawaji T, Yoshimura N, Araie M. Potential role of Rho-associated protein kinase inhibitor Y-27632 in glaucoma filtration surgery. Invest Ophthalmol Vis Sci. 2007;48:5549–5557. doi: 10.1167/iovs.07-0878. [DOI] [PubMed] [Google Scholar]
  • 155.Chen J, Runyan SA, Robinson MR. Novel ocular antihypertensive compounds in clinical trials. Clin Ophthalmol. 2011;5:667–677. doi: 10.2147/OPTH.S15971. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Boland S, Defert O, Alen J, Bourin A, Castermans K, Kindt N, Boumans N, Panitti L, Van de Velde S, Stalmans I. 3-[2-(Aminomethyl)-5-[(pyridin-4-yl) carbamoyl] phenyl] benzoates as soft ROCK inhibitors. Bioorg Med Chem Lett. 2013;23:6442–6446. doi: 10.1016/j.bmcl.2013.09.040. [DOI] [PubMed] [Google Scholar]
  • 157.deLong MA, Yingling J, Lin CW, Sherman B, Sturdivant J, Heintzelman G, Lathem C, van Haarlem T, Kopczynski C. Discovery and SAR of a class of ocularly-active compounds displaying a dual mechanism of activity for the treatment of glaucoma. ARVO Meeting Abstracts. Invest Ophthalmol Vis Sci. 2012;53:3867. [Google Scholar]
  • 158.Kvanta A, Seregard S, Sejersen S, Kull B, Fredholm BB. Localization of Adenosine Receptor Messenger RNAs in the Rat Eye. Exp Eye Res. 1997;65:595–602. doi: 10.1006/exer.1996.0352. [DOI] [PubMed] [Google Scholar]
  • 159.Fleischhauer J, Mitchell C, Stamer W, Karl M, Peterson-Yantorno K, Civan M. Common actions of adenosine receptor agonists in modulating human trabecular meshwork cell transport. J Membr Biol. 2003;193:121–136. doi: 10.1007/s00232-002-2013-5. [DOI] [PubMed] [Google Scholar]
  • 160.Karl MO, Fleischhauer JC, Stamer WD, Peterson-Yantorno K, Mitchell CH, Stone RA, Civan MM. Differential P1-purinergic modulation of human Schlemm’s canal inner-wall cells. Am J Physiol Cell Physiol. 2005;288:C784–C794. doi: 10.1152/ajpcell.00333.2004. [DOI] [PubMed] [Google Scholar]
  • 161.Avila MY, Stone RA, Civan MM. A1-, A2A- and A3-subtype adenosine receptors modulate intraocular pressure in the mouse. Br J Pharmacol. 2001;134:241–245. doi: 10.1038/sj.bjp.0704267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Fredholm BB. Adenosine receptors as drug targets. Exp Cell Res. 2010;316:1284–1288. doi: 10.1016/j.yexcr.2010.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Husain S, Shearer TW, Crosson CE. Mechanisms linking adenosine A1 receptors and extracellular signal-regulated kinase 1/2 activation in human trabecular meshwork cells. J Pharmacol Exp Ther. 2007;320:258–265. doi: 10.1124/jpet.106.110981. [DOI] [PubMed] [Google Scholar]
  • 164.Agarwal R, Agarwal P. Newer targets for modulation of intraocular pressure: focus on adenosine receptor signaling pathways. Expert Opin Ther Targets. 2014;18:527–539. doi: 10.1517/14728222.2014.888416. [DOI] [PubMed] [Google Scholar]
  • 165.Zhong Y, Yang Z, Huang WC, Luo X. Adenosine, adenosine receptors and glaucoma: An updated overview. Biochim Biophys Acta. 2013;1830:2882–2890. doi: 10.1016/j.bbagen.2013.01.005. [DOI] [PubMed] [Google Scholar]
  • 166.Drury A, Szent-Györgyi Av. The physiological activity of adenine compounds with especial reference to their action upon the mammalian heart. J Physiol. 1929;68:213–237. doi: 10.1113/jphysiol.1929.sp002608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Chen JF, Eltzschig HK, Fredholm BB. Adenosine receptors as drug targets - what are the challenges? Nat Rev Drug Discov. 2013;12:265–286. doi: 10.1038/nrd3955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Jacobson KA, Van Galen PJ, Williams M. Adenosine receptors: pharmacology, structure-activity relationships, and therapeutic potential. J Med Chem. 1992;35:407–422. doi: 10.1021/jm00081a001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Müller CE, Jacobson KA. Recent developments in adenosine receptor ligands and their potential as novel drugs. Biochim Biophys Acta. 2011;1808:1290–1308. doi: 10.1016/j.bbamem.2010.12.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Haskó G, Linden J, Cronstein B, Pacher P. Adenosine receptors: therapeutic aspects for inflammatory and immune diseases. Nat Rev Drug Discov. 2008;7:759–770. doi: 10.1038/nrd2638. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Fredholm BB, Arslan G, Halldner L, Kull B, Schulte G, Wasserman W. Structure and function of adenosine receptors and their genes. Naunyn-Schmiedebergs Arch Pharmacol. 2000;362:364–374. doi: 10.1007/s002100000313. [DOI] [PubMed] [Google Scholar]
  • 172.Lebon G, Warne T, Edwards PC, Bennett K, Langmead CJ, Leslie AGW, Tate CG. Agonist-bound adenosine A2A receptor structures reveal common features of GPCR activation. Nature. 2011;474:521–525. doi: 10.1038/nature10136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Doré Andrew S, Robertson N, Errey James C, Ng I, Hollenstein K, Tehan B, Hurrell E, Bennett K, Congreve M, Magnani F, Tate Christopher G, Weir M, Marshall Fiona H. Structure of the adenosine A2A receptor in complex with ZM241385 and the xanthines XAC and caffeine. Structure. 2011;19:1283–1293. doi: 10.1016/j.str.2011.06.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Yan L, Burbiel JC, Maaß A, Müller CE. Adenosine receptor agonists: from basic medicinal chemistry to clinical development. Expert Opin Emerg Drugs. 2003;8:537–576. doi: 10.1517/14728214.8.2.537. [DOI] [PubMed] [Google Scholar]
  • 175.Zhang K, Zhang L, Weinreb RN. Ophthalmic drug discovery: novel targets and mechanisms for retinal diseases and glaucoma. Nat Rev Drug Discov. 2012;11:541–559. doi: 10.1038/nrd3745. [DOI] [PubMed] [Google Scholar]
  • 176.Konno T, Murakami A, Uchibori T, Nagai A, Kogi K, Nakahata N. Involvement of adenosine A2a receptor in intraocular pressure decrease induced by 2-(1-octyn-1-yl) adenosine or 2-(6-cyano-1-hexyn-1-yl) adenosine. J Pharmacol Sci. 2005;97:501–509. doi: 10.1254/jphs.fp0040730. [DOI] [PubMed] [Google Scholar]
  • 177.Daly J. Caffeine analogs: biomedical impact. Cell Mol Life Sci. 2007;64:2153–2169. doi: 10.1007/s00018-007-7051-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Okamura T, Kurogi Y, Hashimoto K, Sato S, Nishikawa H, Kiryu K, Nagao Y. Structure–activity relationships of adenosine A3 receptor ligands: new potential therapy for the treatment of glaucoma. Bioorg Med Chem Lett. 2004;14:3775–3779. doi: 10.1016/j.bmcl.2004.04.099. [DOI] [PubMed] [Google Scholar]
  • 179.Li AH, Moro S, Melman N, Ji X-d, Jacobson KA. Structure-activity relationships and molecular modeling of 3, 5-diacyl-2, 4-dialkylpyridine derivatives as selective A3 adenosine receptor antagonists. J Med Chem. 1998;41:3186–3201. doi: 10.1021/jm980093j. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Wang Z, Do CW, Avila MY, Peterson-Yantorno K, Stone RA, Gao ZG, Joshi B, Besada P, Jeong LS, Jacobson KA, Civan MM. Nucleoside-derived antagonists to A3 adenosine receptors lower mouse intraocular pressure and act across species. Exp Eye Res. 2010;90:146–154. doi: 10.1016/j.exer.2009.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Crosson CE. Adenosine receptor activation modulates intraocular pressure in rabbits. J Pharmacol Exp Ther. 1995;273:320–326. [PubMed] [Google Scholar]
  • 182.Crosson CE, Sloan CF, Yates PW. Modulation of conventional outflow facility by the adenosine A1 agonist N6-cyclohexyladenosine. Invest Ophthalmol Vis Sci. 2005;46:3795–3799. doi: 10.1167/iovs.05-0421. [DOI] [PubMed] [Google Scholar]
  • 183.Tian B, Gabelt BAT, Crosson CE, Kaufman PL. Effects of adenosine agonists on intraocular pressure and aqueous humor dynamics in cynomolgus monkeys. Exp Eye Res. 1997;64:979–989. doi: 10.1006/exer.1997.0296. [DOI] [PubMed] [Google Scholar]
  • 184.Shearer TW, Crosson CE. Adenosine A1 receptor modulation of MMP-2 secretion by trabecular meshwork cells. Invest Ophthalmol Vis Sci. 2002;43:3016–3020. [PubMed] [Google Scholar]
  • 185.Keller KE, Aga M, Bradley JM, Kelley MJ, Acott TS. Extracellular matrix turnover and outflow resistance. Exp Eye Res. 2009;88:676–682. doi: 10.1016/j.exer.2008.11.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Bradley J, Vranka J, Colvis CM, Conger DM, Alexander JP, Fisk AS, Samples JR, Acott TS. Effect of matrix metalloproteinases activity on outflow in perfused human organ culture. Invest Ophthalmol Vis Sci. 1998;39:2649–2658. [PubMed] [Google Scholar]
  • 187.Karl MO, Peterson-Yantorno K, Civan MM. Cell-specific differential modulation of human trabecular meshwork cells by selective adenosine receptor agonists. Exp Eye Res. 2007;84:126–134. doi: 10.1016/j.exer.2006.09.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Al-Aswad LA, Gong H, Lee D, O’Donnell ME, Brandt JD, Ryan WJ, Schroeder A, Erickson KA. Effects of Na-K-2Cl cotransport regulators on outflow facility in calf and human eyes in vitro. Invest Ophthalmol Vis Sci. 1999;40:1695–1701. [PubMed] [Google Scholar]
  • 189.Mitchell CH, Peterson-Yantorno K, Carré DA, McGlinn AM, Coca-Prados M, Stone RA, Civan MM. A3 adenosine receptors regulate Cl channels of nonpigmented ciliary epithelial cells. Am J Physiol Cell Physiol. 1999;276:C659–C666. doi: 10.1152/ajpcell.1999.276.3.C659. [DOI] [PubMed] [Google Scholar]
  • 190.Yang H, Avila MY, Peterson-Yantorno K, Coca-Prados M, Stone RA, Jacobson KA, Civan MM. The cross-species A3 adenosine-receptor antagonist MRS 1292 inhibits adenosine-triggered human nonpigmented ciliary epithelial cell fluid release and reduces mouse intraocular pressure. Curr Eye Res. 2005;30:747–754. doi: 10.1080/02713680590953147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Avila MY, Stone RA, Civan MM. Knockout of A3 adenosine receptors reduces mouse intraocular pressure. Invest Ophthalmol Vis Sci. 2002;43:3021–3026. [PubMed] [Google Scholar]
  • 192.Avni I, Garzozi HJ, Barequet IS, Segev F, Varssano D, Sartani G, Chetrit N, Bakshi E, Zadok D, Tomkins O. Treatment of dry eye syndrome with orally administered CF101: data from a phase 2 clinical trial. Ophthalmology. 2010;117:1287–1293. doi: 10.1016/j.ophtha.2009.11.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Jacobson KA, von Lubitz DKJE, Daly JW, Fredholm BB. Adenosine receptor ligands: differences with acute versus chronic treatment. Trends Pharmacol Sci. 1996;17:108–113. doi: 10.1016/0165-6147(96)10002-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Mitchell CH, Lu W, Hu H, Zhang X, Reigada D, Zhang M. The P2X7 receptor in retinal ganglion cells: A neuronal model of pressure-induced damage and protection by a shifting purinergic balance. Purinergic Signal. 2008;4:313–321. doi: 10.1007/s11302-008-9125-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Zhang X, Zhang M, Laties AM, Mitchell CH. Stimulation of P2X7 receptors elevates Ca2+ and kills retinal ganglion cells. Invest Ophthalmol Vis Sci. 2005;46:2183–2191. doi: 10.1167/iovs.05-0052. [DOI] [PubMed] [Google Scholar]
  • 196.Kakurai K, Sugiyama T, Kurimoto T, Oku H, Ikeda T. Involvement of P2X7 receptors in retinal ganglion cell death after optic nerve crush injury in rats. Neurosci Lett. 2013;534:237–241. doi: 10.1016/j.neulet.2012.11.060. [DOI] [PubMed] [Google Scholar]
  • 197.Niyadurupola N, Sidaway P, Ma N, Rhodes JD, Broadway DC, Sanderson J. P2X7 receptor activation mediates retinal ganglion cell death in a human retina model of ischemic neurodegeneration. Invest Ophthalmol Vis Sci. 2013;54:2163–2170. doi: 10.1167/iovs.12-10968. [DOI] [PubMed] [Google Scholar]
  • 198.Ullian E, Barkis W, Chen S, Diamond J, Barres B. Invulnerability of retinal ganglion cells to NMDA excitotoxicity. Mol Cell Neurosci. 2004;26:544–557. doi: 10.1016/j.mcn.2004.05.002. [DOI] [PubMed] [Google Scholar]
  • 199.Sucher NJ, Lipton SA, Dreyer EB. Molecular basis of glutamate toxicity in retinal ganglion cells. Vision Res. 1997;37:3483–3493. doi: 10.1016/S0042-6989(97)00047-3. [DOI] [PubMed] [Google Scholar]
  • 200.Vorwerk CK, Gorla MS, Dreyer EB. An experimental basis for implicating excitotoxicity in glaucomatous optic neuropathy. Surv Ophthalmol. 1999;43:S142–S150. doi: 10.1016/s0039-6257(99)00017-x. [DOI] [PubMed] [Google Scholar]
  • 201.Hartwick AT, Lalonde MR, Barnes S, Baldridge WH. Adenosine A1-receptor modulation of glutamate-induced calcium influx in rat retinal ganglion cells. Invest Ophthalmol Vis Sci. 2004;45:3740–3748. doi: 10.1167/iovs.04-0214. [DOI] [PubMed] [Google Scholar]
  • 202.Zhang X, Zhang M, Laties AM, Mitchell CH. Balance of purines may determine life or death of retinal ganglion cells as A3 adenosine receptors prevent loss following P2X7 receptor stimulation. J Neurochem. 2006;98:566–575. doi: 10.1111/j.1471-4159.2006.03900.x. [DOI] [PubMed] [Google Scholar]
  • 203.Zhang M, Hu H, Zhang X, Lu W, Lim J, Eysteinsson T, Jacobson KA, Laties AM, Mitchell CH. The A3 adenosine receptor attenuates the calcium rise triggered by NMDA receptors in retinal ganglion cells. Neurochem Int. 2010;56:35–41. doi: 10.1016/j.neuint.2009.08.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Polska E, Ehrlich P, Luksch A, Fuchsjäger-Mayrl G, Schmetterer L. Effects of adenosine on intraocular pressure, optic nerve head blood flow, and choroidal blood flow in healthy humans. Invest Ophthalmol Vis Sci. 2003;44:3110–3114. doi: 10.1167/iovs.02-1133. [DOI] [PubMed] [Google Scholar]
  • 205.Sugiyama T, Shibata M, Kojima S, Ikeda T. Optic nerve head blood flow in glaucoma. In: Kubena T, editor. The Mystery of Glaucoma. InTech; 2011. pp. 207–218. [Google Scholar]
  • 206.Flammer J, Orgül S. Optic nerve blood-flow abnormalities in glaucoma. Prog Retin Eye Res. 1998;17:267–289. doi: 10.1016/s1350-9462(97)00006-2. [DOI] [PubMed] [Google Scholar]
  • 207.Wang L, Cull GA, Piper C, Burgoyne CF, Fortune B. Anterior and posterior optic nerve head blood flow in nonhuman primate experimental glaucoma model measured by laser speckle imaging technique and microsphere method. Invest Ophthalmol Vis Sci. 2012;53:8303–8309. doi: 10.1167/iovs.12-10911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Hirao M, Oku H, Goto W, Sugiyama T, Kobayashi T, Ikeda T. Effects of adenosine on optic nerve head circulation in rabbits. Exp Eye Res. 2004;79:729–735. doi: 10.1016/j.exer.2004.08.008. [DOI] [PubMed] [Google Scholar]
  • 209.Von Lubitz DK, Lin RCS, Popik P, Carter MF, Jacobson KA. Adenosine A3 receptor stimulation and cerebral ischemia. Eur J Pharmacol. 1994;263:59–67. doi: 10.1016/0014-2999(94)90523-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Santiago AR, Baptista FI, Santos PF, Cristóvão G, Ambrósio AF, Cunha RA, Gomes CA. Role of microglia adenosine A 2A receptors in retinal and brain neurodegenerative diseases. Mediators Inflamm. 2014;2014 doi: 10.1155/2014/465694. Article ID 465694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Pal S, Choi WJ, Choe SA, Heller CL, Gao ZG, Chinn M, Jacobson KA, Hou X, Lee SK, Kim HO. Structure–activity relationships of truncated adenosine derivatives as highly potent and selective human A 3 adenosine receptor antagonists. Bioorg Med Chem. 2009;17:3733–3738. doi: 10.1016/j.bmc.2009.03.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Rocha-Sousa A, Rodrigues-Araújo J, Gouveia P, Barbosa-Breda J, Azevedo-Pinto S, Pereira-Silva P, Leite-Moreira A. New therapeutic targets for intraocular pressure lowering. Int Sch Res Notices. 2013;2013 doi: 10.1155/2013/261386. Article ID 261386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Wentz SM, Kim NJ, Wang J, Amireskandari A, Siesky B, Harris A. Novel therapies for open-angle glaucoma. F1000Prime Rep. 2014;6:102. doi: 10.12703/P6-102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Wang H, Wang R, Thrimawithana T, Little PJ, Xu J, Feng Z-P, Zheng W. The nerve growth factor signaling and its potential as therapeutic target for glaucoma. Biomed Res Int. 2014;2014 doi: 10.1155/2014/759473. Article ID 759473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Aloe L, Rocco ML, Bianchi P, Manni L. Nerve growth factor: from the early discoveries to the potential clinical use. J Transl Med. 2012;10:239–239. doi: 10.1186/1479-5876-10-239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Kaplan DR, Miller FD. Signal transduction by the neutrophin receptors. Curr Opin Cell Biol. 1997;9:213–221. doi: 10.1016/s0955-0674(97)80065-8. [DOI] [PubMed] [Google Scholar]
  • 217.Roberti G, Mantelli F, Macchi I, Massaro-Giordano M, Centofanti M. Nerve growth factor modulation of retinal ganglion cell physiology. J Cell Physiol. 2014;229:1130–1133. doi: 10.1002/jcp.24573. [DOI] [PubMed] [Google Scholar]
  • 218.Carmignoto G, Maffei L, Candeo P, Canella R, Comelli C. Effect of NGF on the survival of rat retinal ganglion cells following optic nerve section. J Neurosci. 1989;9:1263–1272. doi: 10.1523/JNEUROSCI.09-04-01263.1989. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Sivilia S, Giuliani A, Fernández M, Turba ME, Forni M, Massella A, De Sordi N, Giardino L, Calzà L. Intravitreal NGF administration counteracts retina degeneration after permanent carotid artery occlusion in rat. BMC Neurosci. 2009;10:52. doi: 10.1186/1471-2202-10-52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Lambiase A, Centofanti M, Micera A, Manni GL, Mattei E, De Gregorio A, de Feo G, Bucci MG, Aloe L. Nerve growth factor (NGF) reduces and NGF antibody exacerbates retinal damage induced in rabbit by experimental ocular hypertension. Graefes Arch Clin Exp Ophthalmol. 1997;235:780–785. doi: 10.1007/BF02332863. [DOI] [PubMed] [Google Scholar]
  • 221.Lambiase A, Aloe L, Centofanti M, Parisi V, Mantelli F, Colafrancesco V, Manni GL, Bucci MG, Bonini S, Levi-Montalcini R. Experimental and clinical evidence of neuroprotection by nerve growth factor eye drops: implications for glaucoma. Proc Natl Acad Sci U S A. 2009;106:13469–13474. doi: 10.1073/pnas.0906678106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Ferrari MP, Mantelli F, Sacchetti M, Antonangeli MI, Cattani F, D’Anniballe G, Sinigaglia F, Ruffini PA, Lambiase A. Safety and pharmacokinetics of escalating doses of human recombinant nerve growth factor eye drops in a double-masked, randomized clinical trial. BioDrugs. 2014;28:275–283. doi: 10.1007/s40259-013-0079-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Nagase H, Woessner JF. Matrix metalloproteinases. J Biol Chem. 1999;274:21491–21494. doi: 10.1074/jbc.274.31.21491. [DOI] [PubMed] [Google Scholar]
  • 224.De Groef L, Van Hove I, Dekeyster E, Stalmans I, Moons L. MMPs in the trabecular meshwork: promising targets for future glaucoma therapies? Invest Ophthalmol Vis Sci. 2013;54:7756–7763. doi: 10.1167/iovs.13-13088. [DOI] [PubMed] [Google Scholar]
  • 225.Spiga MG, Borrás T. Development of a gene therapy virus with a glucocorticoid-inducible MMP1 for the treatment of steroid glaucoma. Invest Ophthalmol Vis Sci. 2010;51:3029–3041. doi: 10.1167/iovs.09-4918. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Gerometta R, Spiga MG, Borrás T, Candia OA. Treatment of sheep steroid–induced ocular hypertension with a glucocorticoid-inducible MMP1 gene therapy virus. Invest Ophthalmol Vis Sci. 2010;51:3042–3048. doi: 10.1167/iovs.09-4920. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Rosenberg GA. Matrix metalloproteinases and their multiple roles in neurodegenerative diseases. Lancet Neurol. 2009;8:205–216. doi: 10.1016/S1474-4422(09)70016-X. [DOI] [PubMed] [Google Scholar]
  • 228.Yong VW. Metalloproteinases: mediators of pathology and regeneration in the CNS. Nat Rev Neurosci. 2005;6:931–944. doi: 10.1038/nrn1807. [DOI] [PubMed] [Google Scholar]
  • 229.Chintala SK, Zhang X, Austin JS, Fini ME. Deficiency in matrix metalloproteinase gelatinase B (MMP-9) protects against retinal ganglion cell death after optic nerve ligation. J Biol Chem. 2002;277:47461–47468. doi: 10.1074/jbc.M204824200. [DOI] [PubMed] [Google Scholar]
  • 230.Fingert JH, Heon E, Liebmann JM, Yamamoto T, Craig JE, Rait J, Kawase K, Hoh ST, Buys YM, Dickinson J, Hockey RR, Williams-Lyn D, Trope G, Kitazawa Y, Ritch R, Mackey DA, Alward WL, Sheffield VC, Stone EM. Analysis of myocilin mutations in 1703 glaucoma patients from five different populations. Hum Mol Genet. 1999;8:899–905. doi: 10.1093/hmg/8.5.899. [DOI] [PubMed] [Google Scholar]
  • 231.Donegan RK, Hill SE, Freeman DM, Nguyen E, Orwig SD, Turnage KC, Lieberman RL. Structural basis for misfolding in myocilin-associated glaucoma. Hum Mol Genet. 2015;24:2111–2124. doi: 10.1093/hmg/ddu730. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Lutjen-Drecoll E, May CA, Polansky JR, Johnson DH, Bloemendal H, Nguyen TD. Localization of the stress proteins alpha B-crystallin and trabecular meshwork inducible glucocorticoid response protein in normal and glaucomatous trabecular meshwork. Invest Ophthalmol Vis Sci. 1998;39:517–525. [PubMed] [Google Scholar]
  • 233.Takahashi H, Noda S, Mashima Y, Kubota R, Ohtake Y, Tanino T, Kudoh J, Minoshima S, Oguchi Y, Shimizu N. The myocilin (MYOC) gene expression in the human trabecular meshwork. Curr Eye Res. 2000;20:81–84. [PubMed] [Google Scholar]
  • 234.Joe MK, Kwon HS, Cojocaru R, Tomarev SI. Myocilin regulates cell proliferation and survival. J Biol Chem. 2014;289:10155–10167. doi: 10.1074/jbc.M113.547091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235.Koch MA, Rosenhammer B, Koschade SE, Braunger BM, Volz C, Jägle H, Tamm ER. Myocilin modulates programmed cell death during retinal development. Exp Eye Res. 2014;125:41–52. doi: 10.1016/j.exer.2014.04.016. [DOI] [PubMed] [Google Scholar]
  • 236.Buller C, Johnson DH, Tschumper RC. Human trabecular meshwork phagocytosis. Observations in an organ culture system. Invest Ophthalmol Vis Sci. 1990;31:2156–2163. [PubMed] [Google Scholar]
  • 237.Burns JN, Turnage KC, Walker CA, Lieberman RL. The stability of myocilin olfactomedin domain variants provides new insight into glaucoma as a protein misfolding disorder. Biochemistry. 2011;50:5824–5833. doi: 10.1021/bi200231x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Orwig SD, Perry CW, Kim LY, Turnage KC, Zhang R, Vollrath D, Schmidt-Krey I, Lieberman RL. Amyloid fibril formation by the glaucoma-associated olfactomedin domain of myocilin. J Mol Biol. 2012;421:242–255. doi: 10.1016/j.jmb.2011.12.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239.Hill SE, Donegan RK, Lieberman RL. The glaucoma-associated olfactomedin domain of myocilin forms polymorphic fibrils that are constrained by partial unfolding and peptide sequence. J Mol Biol. 2014;426:921–935. doi: 10.1016/j.jmb.2013.12.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240.Lam DS, Leung YF, Chua JK, Baum L, Fan DS, Choy KW, Pang CP. Truncations in the TIGR gene in individuals with and without primary open-angle glaucoma. Invest Ophthalmol Vis Sci. 2000;41:1386–1391. [PubMed] [Google Scholar]
  • 241.Kim BS, Savinova OV, Reedy MV, Martin J, Lun Y, Gan L, Smith RS, Tomarev SI, John SW, Johnson RL. Targeted disruption of the myocilin gene (myoc) suggests that human glaucoma-causing mutations are gain of function. Mol Cell Biol. 2001;21:7707–7713. doi: 10.1128/MCB.21.22.7707-7713.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242.Vollrath D, Liu Y. Temperature sensitive secretion of mutant myocilins. Exp Eye Res. 2006;82:1030–1036. doi: 10.1016/j.exer.2005.10.007. [DOI] [PubMed] [Google Scholar]
  • 243.Gobeil S, Letartre L, Raymond V. Functional analysis of the glaucoma-causing TIGR/myocilin protein: integrity of amino-terminal coiled-coil regions and olfactomedin homology domain is essential for extracellular adhesion and secretion. Exp Eye Res. 2006;82:1017–1029. doi: 10.1016/j.exer.2005.11.002. [DOI] [PubMed] [Google Scholar]
  • 244.Liu Y, Vollrath D. Reversal of mutant myocilin non-secretion and cell killing: implications for glaucoma. Hum Mol Genet. 2004;13:1193–1204. doi: 10.1093/hmg/ddh128. [DOI] [PubMed] [Google Scholar]
  • 245.Jia LY, Gong B, Pang CP, Huang Y, Lam DSC, Wang N, Yam GHF. Correction of the disease phenotype of myocilin-causing glaucoma by a natural osmolyte. Invest Ophthalmol Vis Sci. 2009;50:3743–3749. doi: 10.1167/iovs.08-3151. [DOI] [PubMed] [Google Scholar]
  • 246.Yam GHF, Gaplovska-Kysela K, Zuber C, Roth J. Sodium 4-phenylbutyrate acts as a chemical chaperone on misfolded myocilin to rescue cells from endoplasmic reticulum stress and apoptosis. Invest Ophthalmol Vis Sci. 2007;48:1683–1690. doi: 10.1167/iovs.06-0943. [DOI] [PubMed] [Google Scholar]
  • 247.Balch WE, Morimoto RI, Dillin A, Kelly JW. Adapting proteostasis for disease intervention. science. 2008;319:916–919. doi: 10.1126/science.1141448. [DOI] [PubMed] [Google Scholar]
  • 248.Burns JN, Orwig SD, Harris JL, Watkins JD, Vollrath D, Lieberman RL. Rescue of glaucoma-causing mutant myocilin thermal stability by chemical chaperones. ACS Chem Biol. 2010;5:477–487. doi: 10.1021/cb900282e. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249.Orwig SD, Chi PV, Du Y, Hill SE, Cavitt MA, Suntharalingam A, Turnage KC, Dickey CA, France S, Fu H, Lieberman RL. Ligands for glaucoma-associated myocilin discovered by a generic binding assay. ACS Chem Biol. 2013;9:517–525. doi: 10.1021/cb4007776. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 250.Guerriero CJ, Brodsky JL. The delicate balance between secreted protein folding and endoplasmic reticulum-associated degradation in human physiology. Physiol Rev. 2012;92:537–576. doi: 10.1152/physrev.00027.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251.Zode GS, Bugge KE, Mohan K, Grozdanic SD, Peters JC, Koehn DR, Anderson MG, Kardon RH, Stone EM, Sheffield VC. Topical ocular sodium 4-phenylbutyrate rescues glaucoma in a myocilin mouse model of primary open-angle glaucoma. Invest Ophthalmol Vis Sci. 2012;53:1557–1565. doi: 10.1167/iovs.11-8837. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252.Rubenstein RC, Zeitlin PL. Sodium 4-phenylbutyrate downregulates Hsc70: implications for intracellular trafficking of ΔF508-CFTR. Am J Physiol Cell Physiol. 2000;278:C259–C267. doi: 10.1152/ajpcell.2000.278.2.C259. [DOI] [PubMed] [Google Scholar]
  • 253.Gong B, Zhang LY, Lam DSC, Pang CP, Yam GHF. Sodium 4-phenylbutyrate ameliorates the effects of cataract-causing mutant gammaD-crystallin in cultured cells. Mol Vis. 2010;16:997–1003. [PMC free article] [PubMed] [Google Scholar]
  • 254.Basseri S, Lhoták Š, Sharma AM, Austin RC. The chemical chaperone 4-phenylbutyrate inhibits adipogenesis by modulating the unfolded protein response. J Lipid Res. 2009;50:2486–2501. doi: 10.1194/jlr.M900216-JLR200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255.Suntharalingam A, Abisambra JF, O’Leary JC, Koren J, Zhang B, Joe MK, Blair LJ, Hill SE, Jinwal UK, Cockman M. Glucose-regulated protein 94 triage of mutant myocilin through endoplasmic reticulum-associated degradation subverts a more efficient autophagic clearance mechanism. J Biol Chem. 2012;287:40661–40669. doi: 10.1074/jbc.M112.384800. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 256.Stothert AR, Suntharalingam A, Huard DJ, Fontaine SN, Crowley VM, Mishra S, Blagg BS, Lieberman RL, Dickey CA. Exploiting the interaction between Grp94 and aggregated myocilin to treat glaucoma. Hum Mol Genet. 2014;23:6470–6480. doi: 10.1093/hmg/ddu367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257.Zhang X, Zhang L, Wang S, Wu D, Yang W. Decreased functional expression of Grp78 and Grp94 inhibits proliferation and attenuates apoptosis in a human gastric cancer cell line in vitro. Oncol Lett. 2015;9:1181–1186. doi: 10.3892/ol.2014.2831. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 258.Wang Q, He Z, Zhang J, Wang Y, Wang T, Tong S, Wang L, Wang S, Chen Y. Overexpression of endoplasmic reticulum molecular chaperone GRP94 and GRP78 in human lung cancer tissues and its significance. Cancer Detect Prev. 2005;29:544–551. doi: 10.1016/j.cdp.2005.09.010. [DOI] [PubMed] [Google Scholar]
  • 259.Reddy RK, Lu J, Lee AS. The endoplasmic reticulum chaperone glycoprotein GRP94 with Ca2+-binding and antiapoptotic properties is a novel proteolytic target of calpain during etoposide-induced apoptosis. J Biol Chem. 1999;274:28476–28483. doi: 10.1074/jbc.274.40.28476. [DOI] [PubMed] [Google Scholar]
  • 260.Bando Y, Katayama T, Aleshin A, Manabe T, Tohyama M. GRP94 reduces cell death in SH-SY5Y cells perturbated calcium homeostasis. Apoptosis. 2004;9:501–508. doi: 10.1023/B:APPT.0000031446.95532.ad. [DOI] [PubMed] [Google Scholar]
  • 261.Borrás T. Advances in glaucoma treatment and management: gene therapy. Invest Ophthalmol Vis Sci. 2012;53:2506–2510. doi: 10.1167/iovs.12-9483o. [DOI] [PubMed] [Google Scholar]
  • 262.Martínez T, González MV, Roehl I, Wright N, Pañeda C, Jiménez AI. In vitro and in vivo efficacy of SYL040012, a novel siRNA compound for treatment of glaucoma. Mol Ther. 2014;22:81–91. doi: 10.1038/mt.2013.216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 263.Simonato M, Bennett J, Boulis NM, Castro MG, Fink DJ, Goins WF, Gray SJ, Lowenstein PR, Vandenberghe LH, Wilson TJ. Progress in gene therapy for neurological disorders. Nat Rev Neurol. 2013;9:277–291. doi: 10.1038/nrneurol.2013.56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 264.Patel A, Cholkar K, Agrahari V, Mitra AK. Ocular drug delivery systems: An overview. World J Pharmacol. 2013;2:47–64. doi: 10.5497/wjp.v2.i2.47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 265.Guzman-Aranguez A, Colligris B, Pintor J. Contact lenses: promising devices for ocular drug delivery. J Ocul Pharmacol Ther. 2013;29:189–199. doi: 10.1089/jop.2012.0212. [DOI] [PubMed] [Google Scholar]
  • 266.Molokhia SA, Thomas SC, Garff KJ, Mandell KJ, Wirostko BM. Anterior eye segment drug delivery systems: current treatments and future challenges. J Ocul Pharmacol Ther. 2013;29:92–105. doi: 10.1089/jop.2012.0241. [DOI] [PubMed] [Google Scholar]
  • 267.Achouri D, Alhanout K, Piccerelle P, Andrieu V. Recent advances in ocular drug delivery. Drug Dev Ind Pharm. 2013;39:1599–1617. doi: 10.3109/03639045.2012.736515. [DOI] [PubMed] [Google Scholar]
  • 268.Wehrman T, He X, Raab B, Dukipatti A, Blau H, Garcia KC. Structural and mechanistic insights into nerve growth factor interactions with the TrkA and p75 receptors. Neuron. 2007;53:25–38. doi: 10.1016/j.neuron.2006.09.034. [DOI] [PubMed] [Google Scholar]
  • 269.Morgunova E, Tuuttila A, Bergmann U, Isupov M, Lindqvist Y, Schneider G, Tryggvason K. Structure of human pro-matrix metalloproteinase-2: activation mechanism revealed. Science. 1999;284:1667–1670. doi: 10.1126/science.284.5420.1667. [DOI] [PubMed] [Google Scholar]
  • 270.Dollins DE, Warren JJ, Immormino RM, Gewirth DT. Structures of GRP94-nucleotide complexes reveal mechanistic differences between the hsp90 chaperones. Mol Cell. 28:41–56. doi: 10.1016/j.molcel.2007.08.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271.Immormino RM, Metzger LE, IV, Reardon PN, Dollins DE, Blagg BSJ, Gewirth DT. Different poses for ligand and chaperone in inhibitor-bound Hsp90 and GRP94: implications for paralog-specific drug design. J Mol Biol. 2009;388:1033–1042. doi: 10.1016/j.jmb.2009.03.071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 272.Garnock-Jones KP. Ripasudil: first global approval. Drugs. 2014;74:2211–2215. doi: 10.1007/s40265-014-0333-2. [DOI] [PubMed] [Google Scholar]
  • 273.Fredholm BB, Irenius E, Kull B, Schulte G. Comparison of the potency of adenosine as an agonist at human adenosine receptors expressed in Chinese hamster ovary cells1. Biochem Pharmacol. 2001;61:443–448. doi: 10.1016/s0006-2952(00)00570-0. [DOI] [PubMed] [Google Scholar]
  • 274.Jacobson KA, Gao ZG. Adenosine receptors as therapeutic targets. Nat Rev Drug Discov. 2006;5:247–264. doi: 10.1038/nrd1983. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 275.Cunha RA, Constantino M, Sebastião A, Johansson B, Fredholm BB. Evidence for high-affinity binding sites for the adenosine A2A receptor agonist [3H] CGS 21680 in the rat hippocampus and cerebral cortex that are different from striatal A2A receptors. Naunyn-Schmiedebergs Arch Pharmacol. 1996;353:261–271. doi: 10.1007/BF00168627. [DOI] [PubMed] [Google Scholar]
  • 276.Mathôt R, Wenden E, Soudijn W, IJzerman A, Danhof M. Deoxyribose analogues of N6-cyclopentyladenosine (CPA): partial agonists at the adenosine A1 receptor in vivo. Br J Pharmacol. 1995;116:1957–1964. doi: 10.1111/j.1476-5381.1995.tb16398.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 277.Day YJ, Li Y, Rieger JM, Ramos SI, Okusa MD, Linden J. A2A adenosine receptors on bone marrow-derived cells protect liver from ischemia-reperfusion injury. J Immunol. 2005;174:5040–5046. doi: 10.4049/jimmunol.174.8.5040. [DOI] [PubMed] [Google Scholar]
  • 278.Jacobson KA, Park KS, Jiang JL, Kim YC, Olah ME, Stiles GL, Ji XD. Pharmacological characterization of novel A(3) adenosine receptor-selective antagonists. Neuropharmacology. 1997;36:1157–1165. doi: 10.1016/s0028-3908(97)00104-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 279.Jeong LS, Choe SA, Gunaga P, Kim HO, Lee HW, Lee SK, Tosh DK, Patel A, Palaniappan KK, Gao ZG, Jacobson KA, Moon HR. Discovery of a new nucleoside template for human A3 adenosine receptor ligands: d-4′-thioadenosine derivatives without 4′-hydroxymethyl group as highly potent and selective antagonists. J Med Chem. 2007;50:3159–3162. doi: 10.1021/jm070259t. [DOI] [PubMed] [Google Scholar]
  • 280.Comes N, Buie LK, Borrás T. Evidence for a role of angiopoietin-like 7 (ANGPTL7) in extracellular matrix formation of the human trabecular meshwork: implications for glaucoma. Genes Cells. 2011;16:243–259. doi: 10.1111/j.1365-2443.2010.01483.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 281.Goel M, Sienkiewicz AE, Picciani R, Wang J, Lee RK, Bhattacharya SK. Cochlin, intraocular pressure regulation and mechanosensing. PLoS ONE. 2012;7:e34309. doi: 10.1371/journal.pone.0034309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 282.Iyer P, Maddala R, Pattabiraman PP, Rao PV. Connective tissue growth factor–mediated upregulation of neuromedin: expression in trabecular meshwork cells and its role in homeostasis of aqueous humor outflow. Invest Ophthalmol Vis Sci. 2012;53:4952–4962. doi: 10.1167/iovs.12-9681. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 283.Junglas B, Kuespert S, Seleem AA, Struller T, Ullmann S, Bösl M, Bosserhoff A, Köstler J, Wagner R, Tamm ER, Fuchshofer R. Connective tissue growth factor causes glaucoma by modifying the actin cytoskeleton of the trabecular meshwork. Am J Pathol. 2012;180:2386–2403. doi: 10.1016/j.ajpath.2012.02.030. [DOI] [PubMed] [Google Scholar]
  • 284.Rosenthal R, Fromm M. Endothelin antagonism as an active principle for glaucoma therapy. Br J Pharmacol. 2011;162:806–816. doi: 10.1111/j.1476-5381.2010.01103.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 285.Rocha-Sousa A, Pereira-Silva P, Tavares-Silva M, Azevedo-Pinto S, Rodrigues-Araújo J, Pinho S, Avelino A, Falcão-Reis F, Leite-Moreira A. Identification of the ghrelin-GHSR 1 system and its influence in the modulation of induced ocular hypertension in rabbit and rat eyes. Peptides. 2014;57:59–66. doi: 10.1016/j.peptides.2014.04.013. [DOI] [PubMed] [Google Scholar]
  • 286.Rasmussen CA, Kaufman PL, Ritch R, Haque R, Brazzell RK, Vittitow JL. Latrunculin B reduces intraocular pressure in human ocular hypertension and primary open-angle glaucoma. Transl Vis Sci Technol. 2014;3:1. doi: 10.1167/tvst.3.5.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 287.Pintor J, Peláez T, Hoyle CH, Peral A. Ocular hypotensive effects of melatonin receptor agonists in the rabbit: further evidence for an MT3 receptor. Br J Pharmacol. 2003;138:831–836. doi: 10.1038/sj.bjp.0705118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 288.Wiederholt M, Sturm A, Lepple-Wienhues A. Relaxation of trabecular meshwork and ciliary muscle by release of nitric oxide. Invest Ophthalmol Vis Sci. 1994;35:2515–2520. [PubMed] [Google Scholar]
  • 289.Kaufman PL, Rasmussen CA. Advances in glaucoma treatment and management: outflow drugs. Invest Ophthalmol Vis Sci. 2012;53:2495–2500. doi: 10.1167/iovs.12-9483m. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 290.Fuchshofer R, Tamm ER. The role of TGF-β in the pathogenesis of primary open-angle glaucoma. Cell Tissue Res. 2012;347:279–290. doi: 10.1007/s00441-011-1274-7. [DOI] [PubMed] [Google Scholar]

RESOURCES