Abstract
Background:
Glaucoma is a progressive optic neuropathy characterized by retinal ganglion cell death and altera-tions of visual field. Elevated intraocular pressure (IOP) is considered the main risk factor of glaucoma, even though other factors cannot be ruled out, such as epigenetic mechanisms.
Objective:
An overview of the ultimate promising experimental drugs to manage glaucoma has been provided.
Results:
In particular, we have focused on purinergic ligands, KATP channel activators, gases (nitric oxide, carbon monoxide and hydrogen sulfide), non-glucocorticoid steroidal compounds, neurotrophic factors, PI3K/Akt activators, citicoline, histone deacetylase inhibitors, cannabinoids, dopamine and serotonin receptors ligands, small interference RNA, and Rho kinase in-hibitors.
Conclusions:
The review has been also endowed of a brief chapter on last reports about potential neuroprotective benefits of anti-glaucoma drugs already present in the market.
Keywords: Glaucoma, anti-glaucoma drugs, intraocular pressure, retinal ganglion cells, purinergic ligands, Rho kinase inhibitors, dopamine ligands, nitric oxide, histone deacetylase inhibitors
1. Introduction
Glaucoma is an optic neuropathy characterized by retinal ganglion cell (RGC) death and irreversible peripheral and central visual field loss. The WHO epidemiologic analysis identified glaucoma as the second worldwide leading cause of irreversible blindness, accounting for 12% of blindness cases [http://www.who.int/blindness/causes/en/]. The causes of glaucoma have not been univocally identified, yet. Intraocular pressure (IOP), above 21 mmHg, is the most recognized risk factor of glaucoma; the higher is the IOP, the greater the likelihood of glaucoma. Primary open angle glaucoma (POAG) is a commonly bilateral disease of adult onset and it is characterized by high IOP, optic nerve damage and visual field loss. POAG is the most prevalent type of glaucoma. Primary angle-closure glaucoma (PACG) is less common, the terms “angle closure” refers to occlusion of the trabecular meshwork by the peripheral iris obstructing aqueous outflow. Elevated IOP can cause a “mechanical damage” of the optic nerve head (ONH) and then RGCs death. Beside POAG and PACG, there are patients with normal-tension glaucoma (NTG) that is usually regarded as a variant of POAG. NTG is characterized by IOP equal to or less than 21 mmHg, optic nerve damage, an open anterior chamber angle and visual field loss. Any etiological factors distinct from those in POAG have not been conclusively assessed, even though various mechanisms have been suggested, including anomalies in vascular function and in the structure of optic nerve [1, 2]. It is noteworthy that some NTG patients have been found to have marked nocturnal IOP spikes [1, 3]. Furthermore, recent findings supported the mechanical damage of glaucoma, because OHN compression was found to be related to decreased cerebral spinal fluid pressure, along with increased stiffness of sclera and lamina cribosa connective tissue, particularly in NTG patients [4, 5].
Because POAG patients could benefit IOP lowering medical interventions, glaucoma is considered by the World Health Organization (WHO), an avoidable cause of blindness, and the disease is listed within the Priority Eye Diseases of WHO.
2. Neuroprotection
Besides existence of IOP lowering medical interventions, including pharmacological treatments and ocular surgery procedures (e.g. trabeculectomy), the awareness about glaucoma should not decrease, because symptoms occur when most of the RGCs die.
Nowadays, the unmet medical need in glaucoma is related mainly to: i. ambiguous etiology of glaucoma; ii. late diagnosis; iii. disease progression (RGC death) despite IOP control.
Thus, research of new drugs should not only be focused on IOP lowering agents, but also on neuroprotective molecules (Fig. 1).
Fig. (1).
PubMed entries on neuroprotection strategy and glaucoma treatment. Entries have been retrieved from PubMed using the following formula in the advanced search PubMed engine: “search terms”; YYYY:YYYY [edat]; (e.g. “glaucoma AND IOP”; 2007:2012 [edat]).
While basic research is currently looking for neuroprotective agents, clinical development of neuroprotective drugs for glaucoma has decreased due to several reasons such as: i. uncertain mechanism of pathogenesis disease; ii. uncertain therapeutic targets; iii. unpredictable glaucoma animal models; and iv. limited reliable clinical functional end-points [6]. Progression of visual field loss is, generally, used as a functional endpoint in clinical trials, aiming at evaluation of neuroprotective effects of drugs in glaucoma patients. However, because progression of glaucoma is slow and visual field progression has shown high intra- and inter-patient variability, clinical trials for assessment of efficacy of neuroprotective drugs would need longer time (at least 4-5 years), that means high costs and business risks. With these perspectives, pharmaceutical industries would not easily invest in neuroprotection programs [6]. Another critical point of the choice of visual field loss progression as functional clinical endpoint is that visual field loss occurs when about 50% of RGCs are dead [6]. Thus, basic research is currently focused on validation of new methods for early diagnosis of glaucoma, aimed at the evaluation of RCGs death or dysfunction, that can be evaluated by means of in-vivo retinal imaging and electrophysiological studies [7-9]. Recently, pattern electroretinogram (PERG) amplitude was found to be an innovative functional endpoint for evaluation of RGCs functionality in patients with suspected glaucoma [10]; because PERG amplitude, an index of RGCs dysfunction, was found to be decreased before RGCs death and related structural modification at ONH [11].
A typical example of lack of valuable functional endopoints in clinical trial is represented by the memantine study. After phase III, memantine, a NMDA receptor antagonist, was not further developed as glaucoma treatment, simply because no significant difference was found between the placebo and the drug [12]. The failure of memantine clinical trial had a bad impact on further development of this drug, and in general on investments on neuroprotective agents by pharmaceutical companies [13]. However, this failure in 2008 has not stopped the research and development of new neuroprotection strategies for glaucoma; thus, we have reported in this review the latest updates on basic preclinical and clinical research results on neuroprotective agents for glaucoma.
The recent review by Levin and co-author (2017) confirmed that the main anatomical targets of putative neuroprotective drugs are RGCs, and sites that promote RGCs survival such as Müller cells, astrocytes and retinal microvascular cells [14]. Moreover, other anatomical targets, according to the mechanical hypothesis of glaucoma, are the sclera [15-18] and sites of production and drainage of aqueous humor (AH) [1]: ciliary bodies (site of production of AH-inflow) and trabecular meshwork (site of drainage of AH-outflow).
Several biochemical pathways and pharmacological targets have been explored in order to obtain neuroprotection [19]. Glutamate receptors are recognized as the main pharmacological targets of neuroprotective drugs, because uncontrolled activation of glutamate receptors leads to neurodegeration (exicitoxicity cascade); thus memantine, a NMDA receptor antagonist, was first developed for Alzheimer’s disease and then for glaucoma [19]. There is a general consensus that development of memantine was mainly doomed due to the trial design, rather than the lack of drug efficacy; therefore, the strategy of excitotoxicity cascade inhibition should not be totally abandoned by neuroscientists and ophthalmologists.
3. Purinergic ligands
There is an increasing interest about purinergic receptors as pharmacological targets to manage glaucoma [20]. The adenosine A1 receptor (A1R) agonist INO-8875 (i.e. trabodenoson developed by Inotek pharma) [1] was able to decrease IOP by increasing AH outflow [21], and was recently evaluated in a phase III multi-center, randomized, double-masked, active-(0.5% timolol) and placebo-controlled study [NCT02565173]. However, this study had only one clinical endpoint, i.e. the IOP assessment. Visual function measurement in a future clinical study with trabodenoson, can be of value, because it was found that A1R activation promoted RGCs survival in an in vitro model of inflammatory damage [22] and promoted protective effects in a variety of in vitro and in vivo models of CNS neurodegeneration [23-25]. The adenosine A2R agonist, OPA-6566, reached the phase I/II in a clinical trial [NCT01410188] [1]; however, we were not able to retrieve any information about further development of this drug. Within the adenosine receptors, the A3R might exert modulation of the exicitotoxicity cascade, as suggested by recent reports on A3R agonists [26, 27]. Recently, it was found that A3R activation prevented RGCs death in both in vitro and in vivo models of optic neuropathy [28]. 2-Cl-IB-MECA, an A3R agonist, decreased the number of apoptotic cells treated with kainic acid and NMDA, furthermore, this compound promoted RGCs survival in retinal ischemia-reperfusion injury and in partial optic nerve transection models [28]. However, the role of A3R in glaucoma is still controversial, for instance PBF-677, an A3R antagonist, has been developed for glaucoma treatment (NCT02639975). Furthermore, it was found that either adenosine A3 receptor genetic knockdown [29] or its pharmacological inhibition led to decrease of IOP [30]. On the contrary, CF101, an A3R agonist, originally developed as an oral anti-inflammatory agent for dry eye, was able to decrease IOP in glaucomatous patients [31]. Actually, CF101 is being developed for glaucoma (NCT01033422). The controversies about involvement of A3R in IOP regulation could be related to the “inversion effect”, which was found to be a typical characteristic of adenosine receptors, as it was theorized by Jacobson and co-authors (1996) [32]. Indeed, the “inversion effect” can explain why the pharmacological effect of an adenosine ligand, administered in acute, can be totally inverted when it is administered chronically. Since glaucoma therapy is chronic, the “inversion effect” should be taken into account in a trial that involves A3R or other adenosine receptor ligands.
The neuroprotective action of A3R agonists may include the reduction of intracellular [Ca2+] release, due to the activation of purinergic P2X7 receptor [33-35]. Therefore, P2X7 antagonists can be potentially useful as neuroprotective agents in glaucoma [19]. P2X7 antagonists increased RGCs survival after NMDA challenge (intravitreal injection) [36]. On the contrary, P2X7 activation promoted the mechanosensitive release of IL-3, leading to RGCs survival through activation of the IL-3Rβ receptor. Therefore, P2X7 activation can have a role in retinal protection in case of non-ischemic IOP elevation [37, 38], however, several experimental models of glaucoma highlighted that P2X7 activation could have detrimental effects on RGCs survival [39-43].
4. KATP channel activators
KATP channels are inward-rectifying potassium channels that modulate membrane excitability on the basis of the metabolic state of the cell, because KATP channels are inhibited by micromolar concentrations of intracellular ATP. KATP channels subunits Kir6.1, Kir6.2, and SUR2B are expressed in the trabecular meshwork [44]. Activators of KATP channels (diazoxide and nicorandil) decreased IOP in rats and mice [44, 45] by increasing AH outflow, through trabecular meshwork (TM) or uveo-scleral pathway. Additionally, KR-31378 (KATP channel opener) was found to protect RGCs in a rat model of chronic retinal ischemia [46]. KR-31378 is a synonymous of DNB-001 developed by Danube Pharmaceuticals Inc. This compound was tested in a phase I/II trial assessing safety and efficacy of DNB-001 by oral route in naïve patients with ocular hypertension [NCT00683501]. However, no further development steps of DNB-001 have been reported, so far. KATP openers are generally hydrophobic compounds and this characteristic is a common barrier for development of KATP openers delivered in aqueous eye drops formulations, which are commonly preferred for chronic treatment of glaucoma [47]. However, pharmaceutical technology research is now focusing on ophthalmic formulation of hydrophobic drugs encapsulated into solid lipid nanoparticles or nanostructured lipid carriers, these nanoparticle formulations have shown high loading capacity, high ocular drug availability, high stability and good ocular safety [48, 49].
5. Gaseous messengers
Nitric oxide (NO), carbon monoxide (CO) and hydrogen sulphide (H2S) are gaseous molecules that have long been considered as highly toxic gases and environment pollutants. Recently, it has been highlighted that a cross-talk with these three gases exist in the eye [50, 51]. Particularly, NO, CO and H2S regulated IOP and ocular vascular tone.
Preclinical studies reported an increased retinal immunostaining of nitric oxide synthase (NOS), in a rat model of ocular hypertension [52], and, non-selective inhibition of NOS isoforms by systemic administration of L-NAME protected RGCs in a retinal ischemia-reperfusion injury model [53]. In particular, a study on healthy human volunteers revealed that NOS could have a role in basal autoregulation of optic nerve head blood flow (ONHBF), but not when intraocular pressure is experimentally increased [54]. Thus, a study on ONHBF and the effects of NO in POAG patients, would help in understanding the role of NO and NOS in ocular hypertension and glaucoma. Nitric oxide can exert both neuroprotective and neurotoxic effects, generally depending on NO concentration. NO donor compounds were found to be neuroprotective in in-vitro and in-vivo models of glaucoma [55]; e.g. nipradilol (β-blocker with a NO releasing moiety) [56, 57]. However, a comparative clinical study on NTG patients did not highlight differences between the
For references please see the text. NB. Latanoprost, timolol and brimonidine are currently approved as IOP lowering drugs; we included these compounds in the present table because are under evaluation for potential retinal protection activity.
nipradilol and timolol treatment in terms of visual field loss progression [58]. The promising preclinical results about NO-donor compounds, which are capable to decrease IOP by increasing AH outflow, led to development of latanoprostene bunod (LBN) [55, 59]. LNB is a prostaglandin analogue with a NO releasing moiety; LNB is also known as PF-03187207, BOL-303259-X, NCX-116, and Vesneo™. LBN was found to be more effective in decreasing IOP than latanoprost [1, 60], however, its potential neuroprotective effect needs to be further investigated [61]. We have retrieved only a clinical trial (NCT00441883), which evaluated the effect of LBN on visual field (secondary outcome) after 28 days of treatment, however, this time slot was quite short and any potential neuroprotective effect of LNB treatment would not be highlighted. Indeed, further studies about the role of NO and specific NOS isoforms in ocular vascular regulation and neuroprotection of RGCs, are needed.
The effect of NO on modulation of IOP can be also linked to µ3 opioid receptor [62], because it was found that the reduction of IOP after ocular instillation of morphine was reversed by pretreatment with L-NAME [63]. A series of findings might suggest that opioid receptors activation protects the retina from damage associated to different experimental models of glaucoma [64-67]. Three opioid receptor subtypes (δ, κ, μ) were found to be expressed in rat retina [67]. Activation of δ-opioid receptor protects RGCs in an experimental model of elevated IOP inducing a downregulation of inducible NOS (iNOS) [65]. It is noteworthy that δ-opioid receptor activation can counteract neuroinflammation [68], which is detrimental to RGCs function and progression of glaucoma [69, 70].
6. Non-glucocorticoid steroidal com-pounds
The neuroprotective potential of estrogens was recently extensively reviewed by Engler-Chiurazzi and co-authors (2016) [71]. The estrogen 17β-estradiol (E2) showed protective effects on RGCs in different experimental models of glaucoma [72-74], including the ischemia-reperfusion injury model [75]. Additionally, 17β-estradiol eye drops promoted RCGs survival and preserved visual function in an experimental model of glaucoma without decreasing IOP [76]. Furthermore, a study reported that estrogen levels could affect the risk of glaucoma in women [77]. We have then looked up in literature for molecules with similar properties of estrogens, but bearing poor estrogenic effects and non-glucocorticoid activity. Such lead compound would be 17α-Ethynyl-androst-5ene-3β, 7β, 17β-triol (HE3286) [78], which is a synthetic derivative of dehydroepiandrosterone, that was initially developed as an anti-inflammatory drug to treat diabetes and autoimmune diseases. Dehydroepiandrosterone exerted neuroprotective effects in several preclinical studies [79-83], and its synthetic derivative HE3286 protected optic nerve head and RGCs from damage, in an experimental model of optic neuritis [84]. Furthermore, HE3286 enhanced RGCs survival and expression of brain derived neurotrophic factor (BDNF) in retina and optic nerve head in an experimental model of ocular hypertension; however, HE3286 did not decrease the IOP in glaucomatous rats [85]. Indeed, development of HE3286, or related derivatives, eye drops could be a novelty in the therapeutic glaucoma panorama.
7. Neurotrophic factors
Glaucoma progression could be related to neurotrophins deprivation [86]; interestingly, low serum levels of BDNF and nerve growth factor (NGF) were associated to early-moderate stages of glaucoma [87]. NGF serum levels in glaucomatous patients correlated with visual field modification in comparison to control group, but this correlation was not found between BDNF levels and visual field deviation in glaucomatous patients. The potential therapeutic value of BDNF and NGF to treat glaucoma is noteworthy, and the main point that breaks the development of these factors as eye drops is related to the drug delivery challenges [88]. Local administration such as intravitreal injection of neurotrophic factors cannot be pursued because glaucoma treatment is lifelong. Ocular topical treatment of neurotrophin is challenging due to protein stability and degradation sensitivity, however, besides these problems, human recombinant NGF (hrNGF) ophthalmic formulation (180 μg/ml) is currently under clinical development in a phase Ib trial for treatment of glaucoma (NCT02855450).
Additionally, the ciliary neurotrophic factor (CNTF), delivered through an intravitreal implant “NT-501”, has been recently investigated for treatment of glaucoma in the NCT01408472 clinical trial. CNTF exerted, in several models of RGCs death and optic nerve ischemia, protective effects most likely by means of activation of JAK/STAT, MAPK/ERK, and PI3K/Akt pathways [89]. Furthermore, after optic nerve crush procedure in animals, CNTF treatment led to regeneration of optic nerve axons, and restored the visual function in the animals with damaged optic nerve [90].
8. PI3K/Akt activators
Akt is a downstream component of the phosphoinositide 3-kinase (PI-3K) signaling that exerts prosurvival and antiapoptotic effects, and glycogen synthase kinase-3β (GSK-3β) is a downstream substrate of Akt. Increased levels of pAKT led to phosphorilation of GSK3β, which was in turn inactivated, leading to RGC survival after ischemic-reperfusion insult [91]. It has been demonstrated that co-administration of forskolin, an adenylate cyclase activator, along with homotaurine and L-carnosine significantly increased pGSK-3β protecting RGCs from ischemic insult [92, 93]. Direct inhibition of GSK3β has been explored as neuroprotection strategy, and tideglusib, a GSK3β inhibitor, was able to protect RCGs from damage induced by intravitreal injection of NMDA [94]. Up-stream activation of the PI3K/Akt pathway can occur at transmembrane receptors such as apelin receptor (APJ) [95], a GPCR, which shares high sequence homology with the angiotensin II receptor. The endogenous ligand of APJ is the 36 aminoacid peptide called apelin, that protected RGCs from damage induced by NMDA when injected intravitreally in rats [96]. Actually, there are ongoing studies on stable apelin analogues [97], as well as small molecules that activate APJ [98, 99].
9. Citicoline
Citicoline or cytidine 5'-diphosphocholine might protect RGCs by mimicking neurotrophic factors [100] and by inhibiting excitotoxicity in retinal tissue [101]. Our literature search has highlighted one randomized, prospective and masked study with open angle glaucoma patients, which were treated four months with citicoline eye drops.
Topical treatment (4 months) with citicoline in open angle glaucoma patients induced an enhancement of the retinal function (increase of PERG amplitude) with a consequent improvement of visual cortex activity (shortening and increase of visual evoked potentials (VEP) implicit time and amplitude, respectively) [102]. Previously, Parisi et al. (2008) investigated citicoline treatment (oral and intramuscular administration) on retinal function and neural conduction in the visual pathways of glaucoma patients with moderate visual defects [103]. In this study, authors showed that one year-treatment with citicoline improved retinal function (evaluated by pattern electroretinogram recordings) and neural conduction along visual pathways (evaluated by visual evoked potential recordings) in glaucoma patients. Besides the small number of published papers, citicoline has been used as neuroprotective agent for long time and its use for treatment of glaucoma started in the early 2000s, on the basis of ameliorated visual functions reported in patients systemically treated with citicoline [104]. Furthermore, we have retrieved three studies by search “citicoline AND optic neuropathy” on clinicaltrial.gov: NCT01338389; NCT03046693; NCT02984813.
10. Histone deacetylase inhibitors
The treatment with histone deacetylase (HDAC) inhibitors, in order to increase RGCs survival, was investigated for the first time in 2010 and 2012 by Pelzel H.R. and co-authors [105, 106]. In an experimental (optic nerve crush) and a genetic model of glaucoma (aged DBA/2JR3/R3 mice), HDAC3 accumulated in the nucleus and increased the deacetylation of H4 during the early phase of RGCs death. In this study, the inhibition of HDAC resulted in increased RGCs survival. Valproate, a drug currently used to manage seizures, exhibited protective effects on RGCs because is a HDAC inhibitor [107]. This mechanism could be related to increased expression of the BDNF receptor TrkB [107]; however, valproate showed toxic effects on RGCs at high doses [108]. Furthermore, broad-spectrum HDAC inhibitors are characterized by several adverse reactions, that could involve the gastrointestinal tract, immune and cardiovascular systems [109]. Entinostat (MS-275), a selective HDAC1 and HDAC3, showed fewer mild side effects than valproate [110]. Furthermore, entinostat decreased the rate of RGCs loss in retina of animals after optic nerve crush procedure [111]. In this perspective, more efforts should be carried out in order to develop selective HDAC1,3 inhibitors that might have larger therapeutic index in comparison to non-selective HDAC inhibitors [112].
11. Dopaminergic and serotonergic ligands
Dopaminergic receptors (DR) belong to a class of aminergic GPCRs; D2R-like and specifically D3R agonists were found to be effective in decreasing IOP [1, 113-115]. Pattern electroretinogram (PERG) response of Parkinson’s Disease (PD) patients on dopaminergic therapy was better in comparison to PERG response of untreated PD patients, therefore, the hypothesis of dopamine deficiency behind visual dysfunction has been built on the basis of this finding [116]. Furthermore, PERG latencies were found to be delayed in patients on haloperidol therapy [117]. Thus, the dopaminergic system might have a regulatory role in RGCs physiology [118]. Currently, there are no reports on potential neuroprotective effects of dopaminergic agonists in experimental models of glaucoma, however, recent reports showed that dopaminergic agonists exhibited neuroprotection in models of neurodegenerative diseases, such as Alzheimer’s disease and Parkinson’s disease [119-121]. Glaucoma might have common pathological features with neurodegenerative diseases of CNS [69, 122], then, dopaminergic agonists capable to decrease IOP might also exert retinal neuroprotection. Our search on clinicaltrials.gov (“dopaminergic agonist AND eye”) has highlighted the lack of trials involving dopaminergic agonists for treatment of glaucoma. However, there are clinical trials for investigation of dopaminergic agonist effects on visual function (primary outcomes: retinal fiber layer thickness, multifocal electroretinography, visual acuity, visual field): NCT01663935; NCT01620164; NCT00812760. Interestingly, we found that carbidopa/levodopa combination is currently being under evaluation for improvement of visual function in patients with diabetic retinopathy [NCT02706977] and retinitis pigmentosa [NCT02837640].
The class of serotonergic receptors is a large family, that includes one ionotropic receptor (5-HT3) and six metabotropic GPCRs (5-HT1, 5-HT2, 5-HT4, 5-HT5, 5-HT6 and 5-HT7 receptors). The expression of serotonergic receptors in ocular tissues was investigated, and serotonin receptors were expressed in ocular tissues localized either in the anterior or posterior chambers of the eye, thus the serotonergic system can exert a series of physiological function in the eye [123]. The 5-HT1, 5-HT2, and with less extend 5-HT7 subfamily, have been previously identified as interesting pharmacological targets for modulation of IOP [1, 124, 125]. Two serotonergic ligands reached the phase II of clinical development; however, the development of these two drugs, AL-37807 (Alcon) and BVT28949 (Swedish Orphan Biovitrum), was discontinued. Besides that, serotonergic ligands have a high potential as IOP lowering drugs and protection agents. In fact, it was demonstrated that 5HT1A ligands protect RGCs from death in a model of retinal ischemia and after NMDA insult [126], and neurons in others paradigms of neurodegenerative diseases [127, 128].
12. Cannabinoids
Cannabinoids can control not only IOP, but they could be interesting neuroprotective compounds. In 1996, Yoles and coauthors reported that dexanabinol (HU-211) exerted neuroprotective action in a model of optic nerve injury [129]; however, despite its structural similarity to cannabinoids, dexanabinol it was characterized as NMDA antagonist [130] and NFκB inhibitor [131]. HU-210, the enantiomer of HU-211, exhibited neuroprotective activity in a genetic model of retinitis pigmentosa (P23H rats) [132]. Furthermore, chronic treatment with (–)-Δ9-tetrahydrocannabinol (Δ9-THC), administered through intraperitoneal injection, decreased IOP and reduced death of RGCs in experimental model of ocular hypertension [133]. Recently, the ocular pharmacokinetic profile of Δ9-THC soluble prodrug was evaluated in vivo [134]. However, 1 h after topical administration to rabbit’s eye, this Δ9-THC prodrug was only detected in the anterior segment of the eye, no traces of the drug were determined in the back of the eye [134]. Indeed, Δ9-THC prodrug eye drops would be innovative IOP-lowering agent, but due to the poor bioavailability to the back of the eye could not able to protect RGCs.
Additionally, it was found that the activation of CB1 and TRPV1 receptors by WIN 55212-2 and methanandamide exhibits retinal protection [135].
The role of TRPV1 receptor in glaucoma is still controversial [136-139], however the increasing interest on TRPV1 receptor as pharmacological target to manage glaucoma deserves further investigated.
We have looked up in clinicaltrial.gov database, and retrieved two studies: the first [NCT02080676] has the objective to evaluate IOP and optic nerve structure in marijuana smokers, the aim of the second trial [NCT03078309] is to assess visual function effects of cannabis administration in retinitis pigmentosa patients, in comparison to healthy subjects. These two trials are recent, in particular the NCT02080676 is currently recruiting adult patients with no ocular pathology that are being treated in neurology or pain clinics with medical cannabis. The NCT03078309 is active but is not yet open for patient recruitment. The NCT03078309 will recruit patients with retinitis pigmentosa that will receive the first day a single dose of cannabis (tetrahydrocannabinol:cannabidiol, THC:CBD, 1:40). On the second day the subjects will receive a single dose of cannabis (THC:CBD 1:1) and will be subjected to full ocular exam and visual function assessment. However, after the second day of treatment with cannabis, no further follow-up is planned in NCT03078309 trial, thus long-term effect of cannabis would not be analyzed in retinitis pigmentosa patients. To our knowledge no clinical studies are actually in progress on cannabinoid ligands and glaucoma.
13. Small interference-RNA
Small interference-RNAs (siRNA) are small nucleotides able to interfere with mRNA translation into protein. Two siRNAs reached clinical phases of development: SYL040012, which is able to silence the β2 adrenergic receptor (ADRB2); and QPI-1007 that is able to inhibit caspase-2. SYL040012 works as a beta-blocker and decreases IOP by inhibition of AH production [1]. In a preclinical study, SYL040012 decreased IOP of 20% below the baseline values in albino rabbits for about 7 days [140]. Phase 1 clinical trial on SYL040012 showed that the 600 µg/eye/day dose was more effective in decreasing IOP vs. placebo. Particularly, a subgroup of patients treated with 600 µg/eye/day showed a significant IOP decrease, 20% below the baseline values [141]. Low concentrations of SYL040012 were detected in plasma in a non-clinical pharmacokinetic study [140], while no detectable levels of SYL040012 were found in the clinical pharmacokinetic study [141]. In conclusion, SYL040012 can be considered the next-generation beta-blocker, because this siRNA is an effective IOP lowering agent with a better safety profile [1]. Results about a phase 2 trial of SYL040012 (NCT02250612, Bamosiran from Salentys S.A.) are not available yet, however, visual field evaluation was not included either within the primary or secondary endpoints of the study. QPI-1007 caspase-2 inhibitor was developed for treatment of both non-arthritic ischemic optic neuropathy (NAION) and glaucoma. Non-clinical studies showed strong evidences regarding the ability of siRNA caspase-2 inhibitor to increase RGCs survival [142, 143]. A study on QPI-1007 showed that intravitreal injection of the oligonucleotide was well tolerated in rats [144]. Furthermore, after intravenous infusion or bolus administration, QPI-1007 did not exert any microscopic and macroscopic modifications in rats, suggesting low QPI-1007 systemic toxicity [144]. In 2013, a phase I study evaluated the safety of QPI-1007, one year after the single dose intravitreal administration to NAION patients [NCT01064505]. Moreover, QPI-1007 safety and efficacy was evaluated in patients with acute angle-closure glaucoma [NCT01965106]; the study included visual field evaluation (primary endpoint) up to six months after the first administration [145].
14. Rho Kinase (ROCK) inhibitors
Cells of TM have smooth muscle-like properties [146-148]. Rho kinases (ROCK1 and ROCK2) are interesting pharmacological targets to manage glaucoma; these kinases can interact with an high number of proteins belonging to the actomyosin contractile system, named “contractome”, which was identified in various non-muscle contractile cells [149], including TM cells. ROCK is a protein kinase that regulates actin and myosin, which are proteins that are responsible for cellular contraction. ROCK activity also promotes the production of extracellular matrix proteins, that help the anchoring of cells to their substrate. In the trabecular meshwork the resistance to AH drainage is regulated by the contraction of TM cells and the production of extracellular matrix components. ROCK inhibitors (ROCKi) block TM cells contraction and reduce the production of extracellular matrix elements, thereby, ROCKi compounds increase aqueous humor outflow, thus decrease IOP.
Over the past few years, many studies have highlighted the essential role of the Rho and Rho-associated coiled-coil protein kinase (ROCK) pathway in the pathogenesis and treatment of glaucoma. Modulation of ROCK activity can be exploited not only for regulation of IOP, but ROCKi can be also used in glaucoma surgery; because ROCK inhibitors bear also anti-scarring activity [150]. A ROCK inhibitor (ROCKi) developed by Aerie Pharmaceuticals (0.02% netarsudil, Rhopressa®) is going to be soon approved by FDA, and it has been previously announced as breakthrough innovative therapy for treatment of glaucoma [1]. The clinical trial NCT02246764 was aimed at comparison of 0.02% netarsudil with 0.5% timolol maleate. This trial included within primary endpoints the ETDRs (Early Treatment Diabetic Retinopathy Study) visual acuity evaluation; however, the results of NCT02246764 are not available yet. Noteworthy, there are several reports that showed the neuroprotective properties of ROCK inhibitors, that were tested in several experimental models of glaucoma [151, 152]. The potential neuroprotective effects of ROCK inhibitors have been recently reviewed by Van de Velde and co-authors (2015) [152]. This review highlighted that ROCK inhibitors will probably be a breakthrough technology for treatment of glaucoma, not only for their strong efficacy in decreasing IOP, but also for their high potential neuroprotective properties, that include the induction of axonal regeneration [152]. Recently, Novack G.D. (2017) reported that Aerie updated the timing of its intended new drug application (NDA) filing of netarsudil (Rhopressa®), as glaucoma treatment [153]. However, a May the 15th 2017 press release announced the FDA acceptance of Rhopressa® NDA [154]. Netarsudil is the first compound belonging to a new “dual action” class of drugs, that are capable to decrease IOP by increasing AH outflow and also by decreasing AH in-flow. In fact, netarsudil can inhibit both ROCK and the norepinephrine transporter (NET) [155]. NET is involved in the presynaptic reuptake of norepinephrine, and inhibits by negative feedback the postsynaptic α2 adrenergic signaling. Therefore, inhibition of NET leads to prolongation of the α2 adrenergic signaling, which in turn reduces the production of AH; indeed, a NET inhibitors, such as netarsudil has a mechanism of action similar to the approved IOP lowering drug brimonidine (α2 agonist). Moreover, Aerie is developing Roclatan®, a combination of netarsudil with latanoprost; thus, Roclatan® efficacy is accounted to reduced AH production, increased outflow through the TM and the uveo-scleral pathway. We retrieved three clinical trial of Rhopressa® and Roclatan ® that included as secondary endpoint the visual field monitoring, with follow-up periods ranging from 3 to 12 months after treatment [NCT02674854, NCT02558374, NCT02558400]. While, the development of netarsudil is advanced in the Aerie pipeline, the compound ripasudil (Glanatec®)(Kowa Company ltd) reached the Japanese market in 2014 [156]. Kowa has licensed D. Western Therapeutics Institute, Inc.; which now has global rights on ripasudil, with exception of Japan. However, the agreement between Kowa and D. Western Therapeutics Institute is mostly undisclosed [156]. Ripasudil decreased significantly IOP in a phase III study (JapicCTI-111564); and the association of ripasudil with either timolol or latanoprost was more effective than timolol and latanoprost monotherapy; these results come out from two Phase III clinical trials JapicCTI-111701 and JapicCTI-111700, respectively [156]. Recently, repasudil, in association with a prostaglandin analog (undisclosed compound), was found to be effective in decreasing IOP in patients with exfoliative glaucoma [157]. We have found only one report on the putative neuroprotective effects of ripasudil, which protected RGCs of C57BL/6J mice subjected to optic nerve crush procedure [158]. In a recent preclinical study, ripasudil has been tested in two experimental animal models of diabetic retinopathy and retinopathy of prematurity, respectively [159]. This study may suggest a new therapeutic indication for Rock inhibitors, i.e. ripasudil.
LIM-kinase (LIMK, where LIM is the acronym of the three gene products Lin-11, Isl-1 and Mec-3) is a serine/threonine kinase, that phosphorylates cofilin and regulates actin cytoskeletal reorganization. LIMK, similarly to ROCK, was found to be involved in both remodeling of trabecular meshwork by cytoskeleton re-organization and cell contractility; therefore both LIMK and ROCK inhibition decrease IOP by enlarging the mesh of TM [160, 161]. LX7101 from Lexicon Pharmaceutical, a dual LIMK-ROCK2 inhibitor, reached the phase 1/2a phase [NCT01528111], however, no further development has been reported since 2015. Besides that, a recent study highlighted that LIMK inhibitors may exert a neuroprotective role on photoreceptors, in a model of retinal detachment, therefore, LIMKi development might be redirected to another therapeutic field, rather than glaucoma [162].
15. Market approved drugs and neuro-protective effects
Approved IOP lowering drugs have been further studied in order to claim some neuroprotective effects on RGCs [163]. Within approved drugs for glaucoma, the β-blockers betaxolol, timolol and levobetaxolol exerted retinal neuroprotection in experimental models of glaucoma, such as the ischemic reperfusion injury model. The mechanism of action, through which β-blockers might work as neuroprotectans, could be related to regulation of calcium channels or to induction of neurotrophic factor expression [163]. The effects of timolol on visual function were evaluated in the Low pressure Glaucoma Treatment Study (LoGTS 2011) [13]. The LoGTS 2011 was the main object of the recently published Cochrane Database systematic review, which analyzed clinical trials regarding neuroprotective agents and glaucoma. Particularly, the authors included in the analysis only randomized controlled trials in which topical or oral treatments were applied for neuroprotection in adults with open angle glaucoma (OAG) [13]. The LoGTS 2011 compared 0.2% brimonidine monotherapy with 0.5% timolol monotherapy and one endpoint of this study was the progression of visual field loss. The results of LoGTS 2011 study claimed that brimonidine treated group experienced a slower progression of visual field loss, in comparison to the timolol group. Brimonidine is an adrenergic α2 receptor agonist that decreases IOP mainly blocking aqueous production [1]. Several preclinical studies proved the neuroprotective effects of brimonidine on RGCs [164-168]. In particular, brimonidine promoted RGCs survival in a model of ocular hypertension by increasing p-BAD (Ser 136) levels and by promoting the Aβ non-amyloidogenic pathway (=increased levels of sAPPα) [169]. Besides the promising results on brimonidine the Cochrane systematic review highlighted that the LoGTS 2011 study could be affected by several bias, related to: i. the exclusion of participants after randomization; ii differences between the two study groups in rate of drop-out during the follow-up period; iii. improper handling of missing data [13]. Additionally, LoGTS 2011 study could be affected of bias in the final published reports, because the primary endpoint was changed between the first report in 2005 and the second paper in 2011 [13]. Furthermore, although evaluated, assessments of visual acuity, optic nerve structure and cup-disc ratio are not reported in the papers about LoGTS 2011 study [13].
The prostaglandin analog latanoprost is an approved IOP-lowering drug able to decrease IOP by facilitating AH outflow, mainly through the uveo-scleral pathway. Several non-clinical studies claimed that latanoprost exerts neuroprotective effects in the retina [170, 171]. A Japanese clinical trial evaluated long term effects (within 5 years) on normal tension glaucoma patients treated with latanoprost monotherapy, and this study reported that after 5 years the 68% of patients experienced no progression of glaucoma [172]. Furthermore, the “Observational Study of the Long-term Effect of Latanoprost in Normal Tension Glaucoma (NTG-X-PERT)” [NCT01209624] evaluated IOP and several parameters related to visual function in patients treated with latanoprost monotherapy; the study results were published only on clinicaltrial.gov. In this review, we have analyzed only one cumulative outcome, i.e. the “overall progression of glaucoma damage”. The “overall progression of glaucoma damage” was found in 25.8 patients over 469, that were included in the NTG-X-PERT, however, no statistical analysis of NTG-X-PERT results has been already reported. Worthy of note, the NTG-X-PERT was not included in the Cochrane Database systematic review (2017), because it was registered as an observational study [13].
CONCLUSION
The number of non-clinical and clinical studies on new candidates for glaucoma treatment is still growing, and some of these potential innovative drugs will be likely soon approved for clinical use. However, despite the promising results from non-clinical studies, retinal protection remains a “big question mark” both to neuroscientists and clinicians. So far, many molecules showed impressive effects in terms of retinal protection in several paradigms, but no one passed clinical trials. Non-clinical studies on neuroprotective agents have demonstrated significant effects in terms of functional endpoints such as PERG, visual field loss progression and so on. The reasons of that are not so clear, probably because non-clinical studies have relied on evaluation of early outcomes, while clinical trials rely on long follow-up, or perhaps, because most of non-clinical and clinical studies have used narrow time windows during drug treatment, or simply because we need to establish more accurate paradigms that recapitulate the whole retinal damage observed in the clinical condition. In conclusion, translating bench success to bedside has been frustrating for glaucoma scientists but should be, at the same time, a prick to keep going on that.
CONSENT FOR PUBLICATION
Not applicable.
Table 1.
List of compounds under development to treat glaucoma.
| Pharmacological Action | Name of Molecule(s) | IOP reduction Mechanism |
Neuroprotection
Mechanism |
Clinical Trial | Phase of Clinical Trial | Clinical Trial Outcomes |
|---|---|---|---|---|---|---|
| A1R agonist | trabodenoson | ↑ outflow | Inhibition of neuroinflammation | NCT02565173 | III | IOP |
| A2R agonist | OPA-6566 | ↑ outflow | N.D. | NCT01410188 | I/II | IOP |
| A3R agonist | 2-Cl-IB-MECA | ↓ Inflow ↑ outflow |
Inhibition of excitotoxicity | N.A. | N.A. | N.A. |
| A3R agonist | CF101 or IB-MECA, piclidenoson, |
↓ Inflow ↑ outflow |
Inhibition of excitotoxicity | NCT01033422 | II | IOP |
| A3R antagonist | PBF-677 | ↓ Inflow | N.D. | NCT02639975 | I | safety, PK parameters |
| P2X7 antagonist | A438079 | N.D. | Antiapoptotic ↓ [Ca 2+]intra |
N.A. | N.A. | N.A. |
| KATP opener | DNB-001 | ↑ outflow | Inhibition of excitotoxicity | NCT00683501 | I/II | IOP |
| NO releasing molecule/prostaglandin analog | Latanoprostene bunod or PF-03187207, BOL-303259-X, NCX-116, Vesneo™ |
↑ outflow | Regulation of optic nerve head blood flow downregulation of NMDA functions |
NCT00441883 | II | Visual function |
| δ-opiod agonist | SNC-121 | N.A. | Inhibition of neuroinflammation | N.A. | N.A. | N.A. |
| estrogens | 17β-estradiol and HE3286 | N.A. | Antiapoptotic Induction of BDNF |
N.A. | N.A. | N.A. |
| Neurotrophin | NGF | N.A. | Tissue trophism | NCT02855450 | I | Visual function |
| Neurotrophin | CNTF | N.A. | Tissue trophism | NCT01408472 | I | Visual function |
| Not defined | citicoline | N.A. | Inhibition of excitotoxicity | NCT01338389, NCT03046693 | Visual function | |
| HDAC inhibitor | Entinostat | N.A. | Induction of BDNF | N.A. | N.A. | N.A. |
| Dopaminergic agonist | Dopamine, 8-OH-DPAT, cabergoline | ↓ Inflow | Regulation of RGCs physiology |
NCT02706977, NCT02837640 |
Visual function | |
| Serotonergic ligands | AL-37807 | ↓ Inflow ↑ outflow |
Inhibition of excitotoxicity and neuroinflammation | NCT00372931 | II | IOP |
| Cannabinoids | HU-210 WIN55212-2 Cannabis |
↑ outflow | Inhibition of excitotoxicity and neuroinflammation |
NCT02080676, NCT03078309 |
Visual function | |
| Small interference RNA | SYL040012 | ↓ Inflow | N.A. | NCT02250612 | II | IOP |
| Small interference RNA | QPI-1007 | N.A. | Antiapoptotic | NCT01064505, NCT01965106 | I | Safety, optic nerve structure |
| Rho kinase and NET inhibitor | netarsudil | ↓ Inflow ↑ outflow |
Axonal regeneration | NCT02674854, NCT02558374, NCT02558400 | III | IOP, visual function |
| Pharmacological Action | Name of Molecule(s) | IOP reduction Mechanism |
Neuroprotection Mechanism |
Clinical Trial | Phase of Clinical Trial | Clinical Trial Outcomes |
| Rho kinase and NET inhibitor and prostaglandin analog | netarsudil latanoprost |
↓ Inflow ↑ outflow |
↓ Inflow and inhibition of neuroinflammation |
NCT02674854 | III | IOP, visual function |
| β-blocker | timolol | ↓ Inflow | Inhibition of excitotoxicity and induction of neurotrophic factors | LoGTS 2011 | IOP, visual function | |
| α2 adrenergic agonist | brimonidine | ↓ Inflow ↑ outflow |
Antiapoptotic | LoGTS 2011 | IOP, visual function | |
| prostaglandin analog | latanoprost | ↑ outflow | Antiapoptotic and inhibition of neuroinflammation | NTG-X-PERT | IOP, visual function |
ACKNOWLEDGEMENTS
Declared none.
CONFLICT OF INTEREST
The authors declare no conflict of interest, financial or otherwise.
REFERENCES
- 1.Bucolo C., Platania C.B., Reibaldi M., Bonfiglio V., Longo A., Salomone S., Drago F. Controversies in glaucoma: current medical treatment and drug development. Curr. Pharm. Des. 2015;21(32):4673–4681. doi: 10.2174/1381612821666150909095553. [http://dx.doi.org/10.2174/1381612821666150909095553]. [DOI] [PubMed] [Google Scholar]
- 2.Hoban K., Peden R., Megaw R., Halpin P., Tatham A.J. 24-hour contact lens sensor monitoring of intraocular pressure-related profiles in normal-tension glaucoma and rates of disease progression. Ophthalmic Res. 2017;57(4):208–215. doi: 10.1159/000455153. [http://dx.doi.org/10.1159/ 000455153]. [DOI] [PubMed] [Google Scholar]
- 3.Collaborative normal-tension glaucoma study group. The effectiveness of intraocular pressure reduction in the treatment of normal-tension glaucoma. Am. J. Ophthalmol. 1998;126(4):498–505. doi: 10.1016/s0002-9394(98)00272-4. [http://dx.doi.org/10.1016/S0002-9394(98)00272-4]. [DOI] [PubMed] [Google Scholar]
- 4.Morgan W.H., Balaratnasingam C., Lind C.R., Colley S., Kang M.H., House P.H., Yu D.Y. Cerebrospinal fluid pressure and the eye. Br. J. Ophthalmol. 2016;100(1):71–77. doi: 10.1136/bjophthalmol-2015-306705. [http://dx.doi.org/10. 1136/bjophthalmol-2015-306705]. [DOI] [PubMed] [Google Scholar]
- 5.Stowell C., Burgoyne C.F., Tamm E.R., Ethier C.R. Biomechanical aspects of axonal damage in glaucoma: A brief review. Exp. Eye Res. 2017;157:13–19. doi: 10.1016/j.exer.2017.02.005. [http://dx.doi.org/10.1016/j.exer. 2017.02.005]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Liu Y., Pang I.H. Challenges in the development of glaucoma neuroprotection therapy. Cell Tissue Res. 2013;353(2):253–260. doi: 10.1007/s00441-013-1584-z. [http://dx.doi.org/10.1007/s00441-013-1584-z]. [DOI] [PubMed] [Google Scholar]
- 7.Balendra S.I., Normando E.M., Bloom P.A., Cordeiro M.F. Advances in retinal ganglion cell imaging. Eye (Lond.) 2015;29(10):1260–1269. doi: 10.1038/eye.2015.154. [http://dx.doi.org/10.1038/eye.2015.154]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Hood D.C., Fortune B., Mavrommatis M.A., Reynaud J., Ramachandran R., Ritch R., Rosen R.B., Muhammad H., Dubra A., Chui T.Y. Details of glaucomatous damage are better seen on oct en face images than on oct retinal nerve fiber layer thickness maps. Invest. Ophthalmol. Vis. Sci. 2015;56(11):6208–6216. doi: 10.1167/iovs.15-17259. [http://dx.doi.org/10.1167/iovs.15-17259]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Porciatti V., Bosse B., Parekh P.K., Shif O.A., Feuer W.J., Ventura L.M. Adaptation of the steady-state perg in early glaucoma. J. Glaucoma. 2014;23(8):494–500. doi: 10.1097/IJG.0b013e318285fd95. [http://dx.doi.org/10. 1097/IJG.0b013e318285fd95]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Porciatti V., Feuer W.J., Monsalve P., Triolo G., Vazquez L., McSoley J., Ventura L.M. Head-down posture in glaucoma suspects induces changes in iop, systemic pressure, and perg that predict future loss of optic nerve tissue. J. Glaucoma. 2017;26(5):459–465. doi: 10.1097/IJG.0000000000000648. [http://dx.doi.org/10.1097/IJG.0000000000000648]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Banitt M.R., Ventura L.M., Feuer W.J., Savatovsky E., Luna G., Shif O., Bosse B., Porciatti V. Progressive loss of retinal ganglion cell function precedes structural loss by several years in glaucoma suspects. Invest. Ophthalmol. Vis. Sci. 2013;54(3):2346–2352. doi: 10.1167/iovs.12-11026. [http://dx.doi.org/10.1167/iovs.12-11026]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Osborne N.N. Recent clinical findings with memantine should not mean that the idea of neuroprotection in glaucoma is abandoned. Acta Ophthalmol. 2009;87(4):450–454. doi: 10.1111/j.1755-3768.2008.01459.x. [http://dx.doi.org/10. 1111/j.1755-3768.2008.01459.x]. [DOI] [PubMed] [Google Scholar]
- 13.Sena D.F., Lindsley K. Neuroprotection for treatment of glaucoma in adults. Cochrane Database Syst. Rev. 2017;1:CD006539. doi: 10.1002/14651858.CD006539.pub4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Levin L.A., Crowe M.E., Quigley H.A. Lasker/IRRF Initiative on Astrocytes and Glaucomatous Neurodegeneration Participants, Neuroprotection for glaucoma: Requirements for clinical translation. Exp. Eye Res. 2017;157:34–37. doi: 10.1016/j.exer.2016.12.005. [http://dx.doi.org/10.1016/j. exer.2016.12.005]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Nguyen C., Cone F.E., Nguyen T.D., Coudrillier B., Pease M.E., Steinhart M.R., Oglesby E.N., Jefferys J.L., Quigley H.A. Studies of scleral biomechanical behavior related to susceptibility for retinal ganglion cell loss in experimental mouse glaucoma. Invest. Ophthalmol. Vis. Sci. 2013;54(3):1767–1780. doi: 10.1167/iovs.12-10952. [http://dx.doi. org/10.1167/iovs.12-10952]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Quigley H.A., Pitha I.F., Welsbie D.S., Nguyen C., Steinhart M.R., Nguyen T.D., Pease M.E., Oglesby E.N., Berlinicke C.A., Mitchell K.L., Kim J., Jefferys J.J., Kimball E.C. Losartan treatment protects retinal ganglion cells and alters scleral remodeling in experimental glaucoma. PLoS One. 2015;10(10):e0141137. doi: 10.1371/journal.pone.0141137. [http://dx.doi.org/10.1371/journal.pone.0141137]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Coudrillier B., Pijanka J.K., Jefferys J.L., Goel A., Quigley H.A., Boote C., Nguyen T.D. Glaucoma-related changes in the mechanical properties and collagen micro-architecture of the human sclera. PLoS One. 2015;10(7):e0131396. doi: 10.1371/journal.pone.0131396. [http://dx.doi.org/ 10.1371/journal.pone.0131396]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Pijanka J.K., Spang M.T., Sorensen T., Liu J., Nguyen T.D., Quigley H.A., Boote C. Depth-dependent changes in collagen organization in the human peripapillary sclera. PLoS One. 2015;10(2):e0118648. doi: 10.1371/journal.pone.0118648. [http://dx.doi.org/10.1371/journal.pone.0118648]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Nucci C., Russo R., Martucci A., Giannini C., Garaci F., Floris R., Bagetta G., Morrone L.A. New strategies for neuroprotection in glaucoma, a disease that affects the central nervous system. Eur. J. Pharmacol. 2016;787:119–126. doi: 10.1016/j.ejphar.2016.04.030. [http://dx.doi.org/10.1016/j. ejphar.2016.04.030]. [DOI] [PubMed] [Google Scholar]
- 20.Sanderson J., Dartt D.A., Trinkaus-Randall V., Pintor J., Civan M.M., Delamere N.A., Fletcher E.L., Salt T.E., Grosche A., Mitchell C.H. Purines in the eye: recent evidence for the physiological and pathological role of purines in the RPE, retinal neurons, astrocytes, muller cells, lens, trabecular meshwork, cornea and lacrimal gland. Exp. Eye Res. 2014;127:270–279. doi: 10.1016/j.exer.2014.08.009. [http://dx.doi.org/ 10.1016/j.exer.2014.08.009]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Shearer T.W., Crosson C.E. Adenosine A1 receptor modulation of MMP-2 secretion by trabecular meshwork cells. Invest. Ophthalmol. Vis. Sci. 2002;43(9):3016–3020. [PubMed] [Google Scholar]
- 22.Perigolo-Vicente R., Ritt K., Pereira M.R., Torres P.M., Paes-de-Carvalho R., Giestal-de-Araujo E. Il-6 treatment increases the survival of retinal ganglion cells in vitro: The role of adenosine a1 receptor. Biochem. Biophys. Res. Commun. 2013;430(2):512–518. doi: 10.1016/j.bbrc.2012.12.004. [http://dx.doi.org/10.1016/j.bbrc.2012.12.004]. [DOI] [PubMed] [Google Scholar]
- 23.Serpa A., Pinto I., Bernardino L., Cascalheira J.F. Combined neuroprotective action of adenosine A1 and cannabinoid CB1 receptors against nmda-induced excitotoxicity in the hippocampus. Neurochem. Int. 2015;87:106–109. doi: 10.1016/j.neuint.2015.06.005. [http://dx.doi.org/10.1016/ j.neuint.2015.06.005]. [DOI] [PubMed] [Google Scholar]
- 24.Thomas T.P., Shih T.M. Stimulation of central A1 adenosine receptors suppresses seizure and neuropathology in a soman nerve agent seizure rat model. Toxicol. Mech. Methods. 2014;24(6):385–395. doi: 10.3109/15376516.2014.920450. [http://dx.doi.org/10.3109/15376516.2014.920450]. [DOI] [PubMed] [Google Scholar]
- 25.Hu S., Dong H., Zhang H., Wang S., Hou L., Chen S., Zhang J., Xiong L. Noninvasive limb remote ischemic preconditioning contributes neuroprotective effects via activation of adenosine A1 receptor and redox status after transient focal cerebral ischemia in rats. Brain Res. 2012;1459:81–90. doi: 10.1016/j.brainres.2012.04.017. [http://dx.doi.org/10.1016/ j.brainres.2012.04.017]. [DOI] [PubMed] [Google Scholar]
- 26.Jacobson K.A., Civan M.M. Ocular purine receptors as drug targets in the eye. J. Ocul. Pharmacol. Ther. 2016;32(8):534–547. doi: 10.1089/jop.2016.0090. [http://dx.doi.org/10.1089/jop.2016.0090]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Dikopf M.S., Vajaranant T.S., Edward D.P. Topical treatment of glaucoma: established and emerging pharmacology. Expert Opin. Pharmacother. 2017;18(9):885–898. doi: 10.1080/14656566.2017.1328498. [http://dx.doi.org/10.1080/ 14656566.2017.1328498]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Galvao J., Elvas F., Martins T., Cordeiro M.F., Ambrosio A.F., Santiago A.R. Adenosine A3 receptor activation is neuroprotective against retinal neurodegeneration. Exp. Eye Res. 2015;140:65–74. doi: 10.1016/j.exer.2015.08.009. [http://dx.doi.org/10.1016/j.exer.2015.08.009]. [DOI] [PubMed] [Google Scholar]
- 29.Avila M.Y., Stone R.A., Civan M.M. Knockout of A3 adenosine receptors reduces mouse intraocular pressure. Invest. Ophthalmol. Vis. Sci. 2002;43(9):3021–3026. [PubMed] [Google Scholar]
- 30.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(14):3775–3779. doi: 10.1016/j.bmcl.2004.04.099. [http://dx.doi.org/10.1016/j.bmcl.2004.04.099]. [DOI] [PubMed] [Google Scholar]
- 31.Avni I., Garzozi H.J., Barequet I.S., Segev F., Varssano D., Sartani G., Chetrit N., Bakshi E., Zadok D., Tomkins O., Litvin G., Jacobson K.A., Fishman S., Harpaz Z., Farbstein M., Yehuda S.B., Silverman M.H., Kerns W.D., Bristol D.R., Cohn I., Fishman P. Treatment of dry eye syndrome with orally administered CF101: data from a phase 2 clinical trial. Ophthalmology. 2010;117(7):1287–1293. doi: 10.1016/j.ophtha.2009.11.029. [http://dx.doi.org/10.1016/j.ophtha.2009. 11.029]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Jacobson K.A., von Lubitz D.K., Daly J.W., Fredholm B.B. Adenosine receptor ligands: differences with acute versus chronic treatment. Trends Pharmacol. Sci. 1996;17(3):108–113. doi: 10.1016/0165-6147(96)10002-x. [http:// dx.doi.org/10.1016/0165-6147(96)10002-X]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Zhang X.L., Zhang M., Laties A.M., Mitchell C.H. 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(2):566–575. doi: 10.1111/j.1471-4159.2006.03900.x. [http://dx.doi.org/10. 1111/j.1471-4159.2006.03900.x]. [DOI] [PubMed] [Google Scholar]
- 34.Zhang M., Budak M.T., Lu W., Khurana T.S., Zhang X., Laties A.M., Mitchell C.H. Identification of the A3 adenosine receptor in rat retinal ganglion cells. Mol. Vis. 2006;12:937–948. [PubMed] [Google Scholar]
- 35.Hu H., Lu W., Zhang M., Zhang X., Argall A.J., Patel S., Lee G.E., Kim Y.C., Jacobson K.A., Laties A.M., Mitchell C.H. Stimulation of the P2X7 receptor kills rat retinal ganglion cells in vivo. Exp. Eye Res. 2010;91(3):425–432. doi: 10.1016/j.exer.2010.06.017. [http://dx.doi.org/10. 1016/j.exer.2010.06.017]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Sakamoto K., Endo K., Suzuki T., Fujimura K., Kurauchi Y., Mori A., Nakahara T., Ishii K. P2X7 receptor antagonists protect against n-methyl-d-aspartic acid-induced neuronal injury in the rat retina. Eur. J. Pharmacol. 2015;756:52–58. doi: 10.1016/j.ejphar.2015.03.008. [http://dx.doi.org/10. 1016/j.ejphar.2015.03.008]. [DOI] [PubMed] [Google Scholar]
- 37.Lim J.C., Lu W.A., Beckel J.M., Mitchell C.H. Neuronal release of cytokine IL-3 triggered by mechanosensitive autostimulation of the P2X7 receptor is neuroprotective. Front. Cell. Neurosci. 2016;10:270. doi: 10.3389/fncel.2016.00270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Lu W.N., Albalawi F., Beckel J.M., Lim J.C., Laties A.M., Mitchell C.H. The P2X7 receptor links mechanical strain to cytokine il-6 up-regulation and release in neurons and astrocytes. J. Neurochem. 2017;141(3):436–448. doi: 10.1111/jnc.13998. [http://dx.doi.org/10.1111/jnc. 13998]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Xue B., Xie Y., Xue Y., Hu N., Zhang G., Guan H., Ji M. Involvement of P2X7 receptors in retinal ganglion cell apoptosis induced by activated muller cells. Exp. Eye Res. 2016;153:42–50. doi: 10.1016/j.exer.2016.10.005. [http://dx.doi.org/10.1016/j.exer.2016.10.005]. [DOI] [PubMed] [Google Scholar]
- 40.Beckel J.M., Argall A.J., Lim J.C., Xia J.S., Lu W.N., Coffey E.E., Macarak E.J., Shahidullah M., Delamere N.A., Zode G.S., Sheffield V.C., Shestopalov V.I., Laties A.M., Mitchell C.H. Mechanosensitive release of adenosine 5 '-triphosphate through pannexin channels and mechanosensitive upregulation of pannexin channels in optic nerve head astrocytes: A mechanism for purinergic involvement in chronic strain. Glia. 2014;62(9):1486–1501. doi: 10.1002/glia.22695. [http://dx.doi.org/10.1002/glia.22695]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Niyadurupola N., Sidaway P., Ma N., Rhodes J.D., Broadway D.C., Sanderson J. P2X7 receptor activation mediates retinal ganglion cell death in a human retina model of ischemic neurodegeneration. Invest. Ophthalmol. Vis. Sci. 2013;54(3):2163–2170. doi: 10.1167/iovs.12-10968. [http://dx.doi.org/10.1167/iovs.12-10968]. [DOI] [PubMed] [Google Scholar]
- 42.Perez de Lara M.J., Guzman-Aranguez A., de la Villa P., Diaz-Hernandez J.I., Miras-Portugal M.T., Pintor J. Increased levels of extracellular ATP in glaucomatous retinas: Possible role of the vesicular nucleotide transporter during the development of the pathology. Mol. Vis. 2015;21:1060–1070. [PMC free article] [PubMed] [Google Scholar]
- 43.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. [http://dx.doi.org/10.1016/j.neulet.2012.11.060]. [DOI] [PubMed] [Google Scholar]
- 44.Chowdhury U.R., Bahler C.K., Hann C.R., Chang M., Resch Z.T., Romero M.F., Fautsch M.P. Atp-sensitive potassium (KATP) channel activation decreases intraocular pressure in the anterior chamber of the eye. Invest. Ophthalmol. Vis. Sci. 2011;52(9):6435–6442. doi: 10.1167/iovs.11-7523. [http://dx.doi.org/10.1167/iovs.11-7523]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Chowdhury U.R., Holman B.H., Fautsch M.P. ATP-sensitive potassium (KATP) channel openers diazoxide and nicorandil lower intraocular pressure in vivo. Invest. Ophthalmol. Vis. Sci. 2013;54(7):4892–4899. doi: 10.1167/iovs.13-11872. [http://dx.doi.org/10.1167/iovs.13-11872]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Choi A., Choi J.S., Yoon Y.J., Kim K.A., Joo C.K. KR-31378, a potassium-channel opener, induces the protection of retinal ganglion cells in rat retinal ischemic models. J. Pharmacol. Sci. 2009;109(4):511–517. doi: 10.1254/jphs.fp0072067. [http://dx.doi.org/10.1254/jphs.FP0072067]. [DOI] [PubMed] [Google Scholar]
- 47.Roy Chowdhury U., Dosa P.I., Fautsch M.P. ATP sensitive potassium channel openers: a new class of ocular hypotensive agents. Exp. Eye Res. 2017;158:85–93. doi: 10.1016/j.exer.2016.04.020. [http://dx.doi.org/10.1016/j.exer. 2016.04.020]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Puglia C., Offerta A., Carbone C., Bonina F., Pignatello R., Puglisi G. Lipid nanocarriers (LNC) and their applications in ocular drug delivery. Curr. Med. Chem. 2015;22(13):1589–1602. doi: 10.2174/0929867322666150209152259. [http://dx.doi.org/10.2174/0929867322666150209152259]. [DOI] [PubMed] [Google Scholar]
- 49.Leonardi A., Bucolo C., Romano G.L., Platania C.B.M., Drago F., Puglisi G., Pignatello R. Influence of different surfactants on the technological properties and in vivo ocular tolerability of lipid nanoparticles. Int. J. Pharm. 2014;470(1-2):133–140. doi: 10.1016/j.ijpharm.2014.04.061. [http://dx. doi.org/10.1016/j.ijpharm.2014.04.061]. [DOI] [PubMed] [Google Scholar]
- 50.Bucolo C., Drago F. Carbon monoxide and the eye: implications for glaucoma therapy. Pharmacol. Ther. 2011;130(2):191–201. doi: 10.1016/j.pharmthera.2011.01.013. [http://dx.doi.org/10.1016/j.pharmthera.2011.01.013]. [DOI] [PubMed] [Google Scholar]
- 51.Salomone S., Foresti R., Villari A., Giurdanella G., Drago F., Bucolo C. Regulation of vascular tone in rabbit ophthalmic artery: cross talk of endogenous and exogenous gas mediators. Biochem. Pharmacol. 2014;92(4):661–668. doi: 10.1016/j.bcp.2014.10.011. [http://dx.doi.org/10.1016/j.bcp. 2014.10.011]. [DOI] [PubMed] [Google Scholar]
- 52.Takir S., Gurel-Gurevin E., Toprak A., Demirci-Tansel C., Uydes-Dogan B.S. The elevation of intraocular pressure is associated with apoptosis and increased immunoreactivity for nitric oxide synthase in rat retina whereas the effectiveness of retina derived relaxing factor is unaffected. Exp. Eye Res. 2016;145:401–411. doi: 10.1016/j.exer.2016.03.002. [http://dx.doi.org/10.1016/j.exer.2016.03.002]. [DOI] [PubMed] [Google Scholar]
- 53.Nucci C., Tartaglione R., Rombola L., Morrone L.A., Fazzi E., Bagetta G. Neurochemical evidence to implicate elevated glutamate in the mechanisms of high intraocular pressure (IOP)-induced retinal ganglion cell death in rat. Neurotoxicology. 2005;26(5):935–941. doi: 10.1016/j.neuro.2005.06.002. [http://dx.doi.org/10.1016/j.neuro.2005.06.002]. [DOI] [PubMed] [Google Scholar]
- 54.Schmidl D., Boltz A., Kaya S., Palkovits S., Told R., Napora K.J., Cherecheanu A.P., Werkmeister R.M., Garhofer G., Schmetterer L. Role of nitric oxide in optic nerve head blood flow regulation during an experimental increase in intraocular pressure in healthy humans. Exp. Eye Res. 2013;116:247–253. doi: 10.1016/j.exer.2013.09.008. [http://dx. doi.org/10.1016/j.exer.2013.09.008]. [DOI] [PubMed] [Google Scholar]
- 55.Cavet M.E., Vittitow J.L., Impagnatiello F., Ongini E., Bastia E. Nitric oxide (no): An emerging target for the treatment of glaucoma. Invest. Ophthalmol. Vis. Sci. 2014;55(8):5005–5015. doi: 10.1167/iovs.14-14515. [http://dx.doi.org/10.1167/iovs.14-14515]. [DOI] [PubMed] [Google Scholar]
- 56.Karim M.Z., Sawada A., Mizuno K., Kawakami H., Ishida K., Yamamoto T. Neuroprotective effect of nipradilol [3,4-dihydro-8-(2-hydroxy-3-isopropylamino)-propoxy-3-nitroxy-2H-1-benzopyran] in a rat model of optic nerve degeneration. J. Glaucoma. 2009;18(1):26–31. doi: 10.1097/IJG.0b013e3181752c6f. [http://dx.doi.org/10.1097/IJG.0b013e3181752c6f]. [DOI] [PubMed] [Google Scholar]
- 57.Miyamoto N., Izumi H., Miyamoto R., Kubota T., Tawara A., Sasaguri Y., Kohno K. Nipradilol and timolol induce Foxo3a and Peroxiredoxin 2 expression and protect trabecular meshwork cells from oxidative stress. Invest. Ophthalmol. Vis. Sci. 2009;50(6):2777–2784. doi: 10.1167/iovs.08-3061. [http://dx.doi.org/10.1167/iovs.08-3061]. [DOI] [PubMed] [Google Scholar]
- 58.Araie M., Shirato S., Yamazaki Y., Kitazawa Y., Ohashi Y., Grp N-T.S. Clinical efficacy of topical nipradilol and timolol on visual field performance in normal-tension glaucoma: a multicenter, randomized, double-masked comparative study. Jpn. J. Ophthalmol. 2008;52(4):255–264. doi: 10.1007/s10384-008-0540-z. [http://dx.doi.org/10.1007/s10384-008-0540-z]. [DOI] [PubMed] [Google Scholar]
- 59.Heyne G.W., Kiland J.A., Kaufman P.L., Gabelt B.T. Effect of nitric oxide on anterior segment physiology in monkeys. Invest. Ophthalmol. Vis. Sci. 2013;54(7):5103–5110. doi: 10.1167/iovs.12-11491. [http://dx.doi.org/ 10.1167/iovs.12-11491]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Weinreb R.N., Ong T., Sforzolini B.S., Vittitow J.L., Singh K., Kaufman P.L. A randomised, controlled comparison of latanoprostene bunod and latanoprost 0.005% in the treatment of ocular hypertension and open angle glaucoma: The VOYAGER study. Br. J. Ophthalmol. 2015;99(6):738–745. doi: 10.1136/bjophthalmol-2014-305908. [http://dx.doi.org/10.1136/ bjophthalmol-2014-305908]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Garcia G.A., Ngai P., Mosaed S., Lin K.Y. Critical evaluation of latanoprostene bunod in the treatment of glaucoma. Clin. Ophthalmol. 2016;10:2035–2050. doi: 10.2147/OPTH.S103985. [http://dx.doi.org/10.2147/OPTH.S103985]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Stagni E., Bucolo C., Motterlini R., Drago F. Morphine-induced ocular hypotension is modulated by nitric oxide and carbon monoxide: Role of mu3 receptors. J. Ocul. Pharmacol. Ther. 2010;26(1):31–35. doi: 10.1089/jop.2009.0081. [http://dx.doi.org/10.1089/jop.2009.0081]. [DOI] [PubMed] [Google Scholar]
- 63.Bonfiglio V., Bucolo C., Camillieri G., Drago F. Possible involvement of nitric oxide in morphine-induced miosis and reduction of intraocular pressure in rabbits. Eur. J. Pharmacol. 2006;534(1-3):227–232. doi: 10.1016/j.ejphar.2006.01.045. [http://dx.doi.org/10.1016/j.ejphar.2006.01.045]. [DOI] [PubMed] [Google Scholar]
- 64.Abdul Y., Akhter N., Husain S. Delta-opioid agonist SNC-121 protects retinal ganglion cell function in a chronic ocular hypertensive rat model. Invest. Ophthalmol. Vis. Sci. 2013;54(3):1816–1828. doi: 10.1167/iovs.12-10741. [http://dx.doi.org/10.1167/iovs.12-10741]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Husain S., Abdul Y., Singh S., Ahmad A., Husain M. Regulation of nitric oxide production by delta-opioid receptors during glaucomatous injury. PLoS One. 2014;9(10):e110397. doi: 10.1371/journal.pone.0110397. [http://dx.doi.org/10.1371/journal.pone.0110397]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Riazi-Esfahani M., Kiumehr S., Asadi-Amoli F., Lashay A.R., Dehpour A.R. Morphine pretreatment provides histologic protection against ischemia-reperfusion injury in rabbit retina. Retina J. Ret. Vit. Dis. 2008;28(3):511–517. doi: 10.1097/IAE.0b013e31815960c3. [DOI] [PubMed] [Google Scholar]
- 67.Husain S., Potter D.E., Crosson C.E. Opioid receptor-activation: retina protected from ischemic injury. Invest. Ophthalmol. Vis. Sci. 2009;50(8):3853–3859. doi: 10.1167/iovs.08-2907. [http://dx.doi.org/10.1167/iovs.08-2907]. [DOI] [PubMed] [Google Scholar]
- 68.Akhter N., Nix M., Abdul Y., Singh S., Husain S. Delta-opioid receptors attenuate TNF-alpha-induced MMP-2 secretion from human onh astrocytes. Invest. Ophthalmol. Vis. Sci. 2013;54(10):6605–6611. doi: 10.1167/iovs.13-12196. [http://dx.doi.org/10.1167/iovs.13-12196]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Romano G.L., Platania C.B., Forte S., Salomone S., Drago F., Bucolo C. Microrna target prediction in glaucoma. 2015. [DOI] [PubMed] [Google Scholar]
- 70.Williams P.A., Marsh-Armstrong N., Howell G.R. Lasker/IRRF initiative on astrocytes and glaucomatous neurodegeneration participants, neuroinflammation in glaucoma: A new opportunity. Exp. Eye Res. 2017;157:20–27. doi: 10.1016/j.exer.2017.02.014. [http://dx.doi.org/10.1016/j.exer. 2017.02.014]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Engler-Chiurazzi E.B., Singh M., Simpkins J.W. Reprint of: From the 90's to now: A brief historical perspective on more than two decades of estrogen neuroprotection. Brain Res. 2016;1645:79–82. doi: 10.1016/j.brainres.2016.06.016. [http://dx.doi.org/10.1016/j.brainres.2016.06.016]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Zhou X.H., Li F., Ge J., Sarkisian S.R., Tomita H., Zaharia A., Chodosh J., Cao W. Retinal ganglion cell protection by 17-beta-estradiol in a mouse model of inherited glaucoma. Dev. Neurobiol. 2007;67(5):603–616. doi: 10.1002/dneu.20373. [http://dx.doi.org/10.1002/dneu.20373]. [DOI] [PubMed] [Google Scholar]
- 73.Nakazawa T., Takahashi H., Shimura M. Estrogen has a neuroprotective effect on axotomized RGCs through ERK signal transduction pathway. Brain Res. 2006;1093:141–149. doi: 10.1016/j.brainres.2006.03.084. [http://dx. doi.org/10.1016/j.brainres.2006.03.084]. [DOI] [PubMed] [Google Scholar]
- 74.Kumar D.M., Perez E., Cai Z.Y., Aoun P., Brun-Zinkernagel A.M., Covey D.F., Simpkins J.W., Agarwal N. Role of nonfeminizing estrogen analogues in neuroprotection of rat retinal ganglion cells against glutamate-induced cytotoxicity. Free Radic. Biol. Med. 2005;38(9):1152–1163. doi: 10.1016/j.freeradbiomed.2004.12.007. [http://dx.doi.org/10.1016/ j.freeradbiomed.2004.12.007]. [DOI] [PubMed] [Google Scholar]
- 75.Russo R., Cavaliere F., Watanabe C., Nucci C., Bagetta G., Corasaniti M.T., Sakurada S., Morrone L.A. 2008. [DOI] [PubMed] [Google Scholar]
- 76.Prokai-Tatrai K., Xin H., Nguyen V., Szarka S., Blazics B., Prokai L., Koulen P. 17 beta-estradiol eye drops protect the retinal ganglion cell layer and preserve visual function in an in vivo model of glaucoma. Mol. Pharm. 2013;10(8):3253–3261. doi: 10.1021/mp400313u. [http://dx.doi. org/10.1021/mp400313u]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Dewundara S.S., Wiggs J.L., Sullivan D.A., Pasquale L.R. Is estrogen a therapeutic target for glaucoma? Semin. Ophthalmol. 2016;31(1-2):140–146. doi: 10.3109/08820538.2015.1114845. [http://dx.doi.org/10.3109/08820538.2015. 1114845]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Ahlem C.N., Kennedy M.R., Page T.M., Reading C.L., White S.K., McKenzie J.J., Cole P.I., Stickney D.R., Frincke J.M. Studies of the pharmacology of 17 alpha-ethynyl-androst-5-ene-3 beta,7 beta,17 beta-triol, a synthetic anti-inflammatory androstene. Int. J. Clin. Exp. Med. 2011;4(2):119–135. [PMC free article] [PubMed] [Google Scholar]
- 79.Arbo B.D., Benetti F., Ribeiro M.F. Astrocytes as a target for neuroprotection: modulation by progesterone and dehydroepiandrosterone. Prog. Neurobiol. 2016;144:27–47. doi: 10.1016/j.pneurobio.2016.03.010. [http://dx.doi. org/10.1016/j.pneurobio.2016.03.010]. [DOI] [PubMed] [Google Scholar]
- 80.El Bitar F., Meunier J., Villard V., Almeras M., Krishnan K., Covey D.F., Maurice T., Akwa Y. Neuroprotection by the synthetic neurosteroid enantiomers ent-PREGS and ent-DHEAS against a beta(25-35) peptide-induced toxicity in vitro and in vivo in mice. Psychopharmacology (Berl.) 2014;231(17):3293–3312. doi: 10.1007/s00213-014-3435-3. [http://dx.doi.org/10.1007/s00213-014-3435-3]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Kokona D., Charalampopoulos I., Pediaditakis I., Gravanis A., Thermos K. The neurosteroid dehydroepiandrosterone (DHEA) protects the retina from AMPA-induced excitotoxicity: NGF TrkA receptor involvement. Neuropharmacology. 2012;62(5-6):2106–2117. doi: 10.1016/j.neuropharm.2012.01.006. [http://dx.doi.org/10.1016/j.neuropharm.2012.01.006]. [DOI] [PubMed] [Google Scholar]
- 82.Luppi C., Fioravanti M., Bertolini B., Inguscio M., Grugnetti A., Guerriero F., Rovelli C., Cantoni F., Guagnano P., Marazzi E., Rolfo E., Ghianda D., Levante D., Guerrini C., Bonacasa R., Solerte S.B. Growth factors decrease in subjects with mild to moderate Alzheimer’s disease (AD): potential correction with dehydroepiandrosterone-sulphate (DHEAS). Arch. Gerontol. Geriatr. 2009;49:173–184. doi: 10.1016/j.archger.2009.09.027. [http://dx.doi.org/10.1016/j.archger.2009.09.027]. [DOI] [PubMed] [Google Scholar]
- 83.Charalampopoulos I., Alexaki V.I., Lazaridis I., Dermitzaki E., Avlonitis N., Tsatsanis C., Calogeropoulou T., Margioris A.N., Castanas E., Gravanis A. G protein-associated, specific membrane binding sites mediate the neuroprotective effect of dehydroepiandrosterone. FASEB J. 2006;20(1):577. doi: 10.1096/fj.05-5078fje. [http://dx.doi.org/10. 1096/fj.05-5078fje]. [DOI] [PubMed] [Google Scholar]
- 84.Khan R.S., Dine K., Luna E., Ahlem C., Shindler K.S. HE3286 reduces axonal loss and preserves retinal ganglion cell function in experimental optic neuritis. Invest. Ophthalmol. Vis. Sci. 2014;55(9):5744–5751. doi: 10.1167/iovs.14-14672. [http://dx.doi.org/10.1167/iovs.14-14672]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Lambert W.S., Carlson B.J., Formichella C.R., Sappington R.M., Ahlem C., Calkins D.J. Oral delivery of a synthetic sterol reduces axonopathy and inflammation in a rodent model of glaucoma. Front. Neurosci. 2017;11:45. doi: 10.3389/fnins.2017.00045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Johnson E.C., Guo Y., Cepurna W.O., Morrison J.C. Neurotrophin roles in retinal ganglion cell survival: lessons from rat glaucoma models. Exp. Eye Res. 2009;88(4):808–815. doi: 10.1016/j.exer.2009.02.004. [http://dx. doi.org/10.1016/j.exer.2009.02.004]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Oddone F., Roberti G., Micera A., Busanello A., Bonini S., Quaranta L., Agnifili L., Manni G. Exploring serum levels of brain derived neurotrophic factor and nerve growth factor across glaucoma stages. PLoS One. 2017;12(1):e0168565. doi: 10.1371/journal.pone.0168565. [http://dx.doi. org/10.1371/journal.pone.0168565]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Dekeyster E., Geeraerts E., Buyens T., Van den Haute C., Baekelandt V., De Groef L., Salinas-Navarro M., Moons L. Tackling glaucoma from within the brain: an unfortunate interplay of BDNF and TrkB. PLoS One. 2015;10(11):e0142067. doi: 10.1371/journal.pone.0142067. [http://dx.doi.org/ 10.1371/journal.pone.0142067]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Kimura A., Namekata K., Guo X.L., Harada C., Harada T. Neuroprotection, growth factors and bdnf-trkb signalling in retinal degeneration. Int. J. Mol. Sci. 2016;17(9) doi: 10.3390/ijms17091584. [http://dx.doi.org/ 10.3390/ijms17091584]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Bei F.F., Lee H.H.C., Liu X.F., Gunner G., Jin H., Ma L., Wang C., Hou L.J., Hensch T.K., Frank E., Sanes J.R., Chen C.F., Fagiolini M., He Z.G. Restoration of visual function by enhancing conduction in regenerated axons. Cell. 2016;164(1-2):219–232. doi: 10.1016/j.cell.2015.11.036. [http://dx.doi.org/10.1016/j.cell.2015.11.036]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Russo R., Adornetto A., Cavaliere F., Varano G.P., Rusciano D., Morrone L.A., Corasaniti M.T., Bagetta G., Nucci C. Intravitreal injection of forskolin, homotaurine, and L-carnosine affords neuroprotection to retinal ganglion cells following retinal ischemic injury. Mol. Vis. 2015;21:718–729. [PMC free article] [PubMed] [Google Scholar]
- 92.Bae O.N., Serfozo K., Baek S.H., Lee K.Y., Dorrance A., Rumbeiha W., Fitzgerald S.D., Farooq M.U., Naravelta B., Bhatt A., Majid A. Safety and efficacy evaluation of carnosine, an endogenous neuroprotective agent for ischemic stroke. Stroke. 2013;44(1):205–212. doi: 10.1161/STROKEAHA.112.673954. [http://dx.doi.org/10.1161/STROKEAHA.112.673954]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Wu S.C., Yue Y., Tian H., Tao L., Wang Y.T., Xiang J., Wang S., Ding H. Tramiprosate protects neurons against ischemic stroke by disrupting the interaction between PSD95 and nNOS. Neuropharmacology. 2014;83:107–117. doi: 10.1016/j.neuropharm.2014.04.010. [http://dx.doi.org/10.1016/j. neuropharm.2014.04.010]. [DOI] [PubMed] [Google Scholar]
- 94.Marchena M., Villarejo-Zori B., Zaldivar-Diez J., Palomo V., Gil C., Hernandez-Sanchez C., Martinez A., de la Rosa E.J. Small molecules targeting glycogen synthase kinase 3 as potential drug candidates for the treatment of retinitis pigmentosa. J. Enzyme Inhib. Med. Chem. 2017;32(1):522–526. doi: 10.1080/14756366.2016.1265522. [http://dx.doi.org/10. 1080/14756366.2016.1265522]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Masri B., Morin N., Cornu M., Knibiehler B., Audigier Y. Apelin (65-77) activates p70 S6 kinase and is mitogenic for umbilical endothelial cells. FASEB J. 2004;18(12):1909–1911. doi: 10.1096/fj.04-1930fje. [http://dx. doi.org/10.1096/fj.04-1930fje]. [DOI] [PubMed] [Google Scholar]
- 96.Ishimaru Y., Sumino A., Kajioka D., Shibagaki F., Yamamuro A., Yoshioka Y., Maeda S. Apelin protects against NMDA-induced retinal neuronal death via an APJ receptor by activating Akt and ERK1/2, and suppressing TNF-alpha expression in mice. J. Pharmacol. Sci. 2017;133(1):34–41. doi: 10.1016/j.jphs.2016.12.002. [http://dx.doi.org/10. 1016/j.jphs.2016.12.002]. [DOI] [PubMed] [Google Scholar]
- 97.Juhl C., Els-Heindl S., Schonauer R., Redlich G., Haaf E., Wunder F., Riedl B., Burkhardt N., Beck-Sickinger A.G., Bierer D. Development of potent and metabolically stable APJ ligands with high therapeutic potential. ChemMedChem. 2016;11(21):2378–2384. doi: 10.1002/cmdc.201600307. [http://dx.doi.org/10.1002/cmdc.201600307]. [DOI] [PubMed] [Google Scholar]
- 98.Murza A., Sainsily X., Cote J., Bruneau-Cossette L., Besserer-Offroy E., Longpre J.M., Leduc R., Dumaine R., Lesur O., Auger-Messier M., Sarret P., Marsault E. Structure-activity relationship of novel macrocyclic biased apelin receptor agonists. Org. Biomol. Chem. 2017;15(2):449–458. doi: 10.1039/c6ob02247b. [http://dx.doi.org/10. 1039/C6OB02247B]. [DOI] [PubMed] [Google Scholar]
- 99.Narayanan S., Maitra R., Deschamps J.R., Bortoff K., Thomas J.B., Zhang Y.Y., Warner K., Vasukuttan V., Decker A., Runyon S.P. Discovery of a novel small molecule agonist scaffold for the APJ receptor. Bioorg. Med. Chem. 2016;24(16):3758–3770. doi: 10.1016/j.bmc.2016.06.018. [http://dx.doi.org/10.1016/j.bmc.2016.06.018]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Oshitari T., Yoshida-Hata N., Yamamoto S. Effect of neurotrophic factors on neuronal apoptosis and neurite regeneration in cultured rat retinas exposed to high glucose. Brain Res. 2010;1346:43–51. doi: 10.1016/j.brainres.2010.05.073. [http://dx.doi.org/10.1016/j.brainres.2010.05.073]. [DOI] [PubMed] [Google Scholar]
- 101.Matteucci A., Varano M., Gaddini L., Mallozzi C., Villa M., Pricci F., Malchiodi-Albedi F. Neuroprotective effects of citicoline in in vitro models of retinal neurodegeneration. Int. J. Mol. Sci. 2014;15(4):6286–6297. doi: 10.3390/ijms15046286. [http://dx.doi.org/10.3390/ijms 15046286]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Parisi V., Centofanti M., Ziccardi L., Tanga L., Michelessi M., Roberti G., Manni G. Treatment with citicoline eye drops enhances retinal function and neural conduction along the visual pathways in open angle glaucoma. Graefes Arch. Clin. Exp. Ophthalmol. 2015;253(8):1327–1340. doi: 10.1007/s00417-015-3044-9. [http://dx.doi.org/10.1007/ s00417-015-3044-9]. [DOI] [PubMed] [Google Scholar]
- 103.Parisi V., Coppola G., Centofanti M., Oddone F., Angrisani A.M., Ziccardi L., Ricci B., Quaranta L., Manni G. Evidence of the neuroprotective role of citicoline in glaucoma patients. 2008. [DOI] [PubMed] [Google Scholar]
- 104.Virno M., Pecori-Giraldi J., Liguori A., De Gregorio F. The protective effect of citicoline on the progression of the perimetric defects in glaucomatous patients (perimetric study with a 10-year follow-up). Acta Ophthalmol. Scand. Suppl. 2000;(232):56–57. doi: 10.1111/j.1600-0420.2000.tb01107.x. [http://dx.doi.org/10.1111/j.1600-0420.2000.tb01107.x]. [DOI] [PubMed] [Google Scholar]
- 105.Pelzel H.R., Schlamp C.L., Nickells R.W. Histone H4 deacetylation plays a critical role in early gene silencing during neuronal apoptosis. BMC Neurosci. 2010;•••:11. doi: 10.1186/1471-2202-11-62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Pelzel H.R., Schlamp C.L., Waclawski M., Shaw M.K., Nickells R.W. Silencing of Fem1c(R3) gene expression in the DBA/2j mouse precedes retinal ganglion cell death and is associated with histone deacetylase activity. Invest. Ophthalmol. Vis. Sci. 2012;53(3):1428–1435. doi: 10.1167/iovs.11-8872. [http://dx.doi.org/10.1167/iovs.11-8872]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Kimura A., Namekata K., Guo X.L., Noro T., Harada C., Harada T. Valproic acid prevents NMDA-induced retinal ganglion cell death via stimulation of neuronal trkb receptor signaling. Am. J. Pathol. 2015;185(3):756–764. doi: 10.1016/j.ajpath.2014.11.005. [http://dx.doi.org/10.1016/j. ajpath.2014.11.005]. [DOI] [PubMed] [Google Scholar]
- 108.Biermann J., Boyle J., Pielen A., Lagreze W.A. Histone deacetylase inhibitors sodium butyrate and valproic acid delay spontaneous cell death in purified rat retinal ganglion cells. Mol. Vis. 2011;17(44-46):395–403. [PMC free article] [PubMed] [Google Scholar]
- 109.Prince H.M., Bishton M.J., Harrison S.J. Clinical studies of histone deacetylase inhibitors. Clin. Cancer Res. 2009;15(12):3958–3969. doi: 10.1158/1078-0432.CCR-08-2785. [http://dx.doi.org/10.1158/1078-0432.CCR-08-2785]. [DOI] [PubMed] [Google Scholar]
- 110.Knipstein J., Gore L. Entinostat for treatment of solid tumors and hematologic malignancies. Expert Opin. Investig. Drugs. 2011;20(10):1455–1467. doi: 10.1517/13543784.2011.613822. [http://dx.doi.org/10.1517/13543784.2011. 613822]. [DOI] [PubMed] [Google Scholar]
- 111.Chindasub P., Lindsey J.D., Duong-Polk K., Leung C.K., Weinreb R.N. Inhibition of histone deacetylases 1 and 3 protects injured retinal ganglion cells. Invest. Ophthalmol. Vis. Sci. 2013;54(1):96–102. doi: 10.1167/iovs.12-10850. [http://dx.doi.org/10.1167/iovs.12-10850]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Schmitt H.M., Schlamp C.L., Nickells R.W. Role of HDACs in optic nerve damage-induced nuclear atrophy of retinal ganglion cells. Neurosci. Lett. 2016;625:11–15. doi: 10.1016/j.neulet.2015.12.012. [http://dx.doi.org/10.1016/ j.neulet.2015.12.012]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Bucolo C., Leggio G.M., Maltese A., Castorina A., D’Agata V., Drag F. Dopamine-(3) receptor modulates intraocular pressure: Implications for glaucoma. Biochem. Pharmacol. 2012;83(5):680–686. doi: 10.1016/j.bcp.2011.11.031. [http://dx.doi.org/10.1016/j.bcp.2011.11.031]. [DOI] [PubMed] [Google Scholar]
- 114.Platania C.B.M., Salomone S., Leggio G.M., Drago F., Bucolo C. Homology modeling of dopamine D-2 and D-3 receptors: molecular dynamics refinement and docking evaluation. PLoS One. 2012;7(9):e44316. doi: 10.1371/journal.pone.0044316. [http://dx.doi.org/10.1371/journal.pone. 0044316]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Platania C.B.M., Leggio G.M., Drago F., Salomone S., Bucolo C. Regulation of intraocular pressure in mice: structural analysis of dopaminergic and serotonergic systems in response to cabergoline. Biochem. Pharmacol. 2013;86(9):1347–1356. doi: 10.1016/j.bcp.2013.08.010. [http://dx.doi.org/ 10.1016/j.bcp.2013.08.010]. [DOI] [PubMed] [Google Scholar]
- 116.Jackson G.R., Owsley C. Visual dysfunction, neurodegenerative diseases, and aging. Neurol. Clin. 2003;21(3):709. doi: 10.1016/s0733-8619(02)00107-x. [http://dx.doi. org/10.1016/S0733-8619(02)00107-X]. [DOI] [PubMed] [Google Scholar]
- 117.Bodis-Wollner I., Tzelepi A. The push-pull action of dopamine on spatial tuning of the monkey retina: The effects of dopaminergic deficiency and selective D-1 and D-2 receptor ligands on the pattern electroretinogram. Vision Res. 1998;38(10):1479–1487. doi: 10.1016/s0042-6989(98)00028-5. [http://dx.doi.org/10.1016/S0042-6989(98)00028-5]. [DOI] [PubMed] [Google Scholar]
- 118.Li Q., Wu N., Cui P., Gao F., Qian W.J., Miao Y.Y., Sun X.H., Wang Z.F. Suppression of outward K+ currents by activating dopamine D1 receptors in rat retinal ganglion cells through PKA and CaMKII signaling pathways. Brain Res. 2016;1635:95–104. doi: 10.1016/j.brainres.2016.01.039. [http://dx.doi.org/10.1016/j.brainres.2016.01.039]. [DOI] [PubMed] [Google Scholar]
- 119.Yedlapudi D., Joshi G.S., Luo D., Todi S.V., Dutta A.K. Inhibition of alpha-synuclein aggregation by multifunctional dopamine agonists assessed by a novel in vitro assay and an in vivo drosophila synucleinopathy model. Sci. Rep. 2016;6:38510. doi: 10.1038/srep38510. [http://dx. doi.org/10.1038/srep38510]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Luo D., Sharma H., Yedlapudi D., Antonio T., Reith M.E.A., Dutta A.K. Novel multifunctional dopamine D-2/D-3 receptors agonists with potential neuroprotection and anti-alpha synuclein protein aggregation properties. Bioorg. Med. Chem. 2016;24(21):5088–5102. doi: 10.1016/j.bmc.2016.08.021. [http://dx.doi.org/10.1016/j.bmc.2016.08.021]. [DOI] [PubMed] [Google Scholar]
- 121.Reichelt D., Radad K., Moldzio R., Rausch W.D., Reichmann H., Gille G. Comparable neuroprotective effects of pergolide and pramipexole on ferrous sulfate-induced dopaminergic cell death in cell culture. CNS Neurol. Disord. Drug Targets. 2016;15(10):1325–1332. doi: 10.2174/1871527315666160801145442. [http://dx.doi.org/10.2174/1871527315666160801145442]. [DOI] [PubMed] [Google Scholar]
- 122.London A., Benhar I., Schwartz M. The retina as a window to the brain-from eye research to cns disorders. Nat. Rev. Neurol. 2013;9(1):44–53. doi: 10.1038/nrneurol.2012.227. [http://dx.doi.org/10.1038/nrneurol.2012.227]. [DOI] [PubMed] [Google Scholar]
- 123.Sharif N.A., Senchyna M. Serotonin receptor subtype mRNA expression in human ocular tissues, determined by RT-PCR. Mol. Vis. 2006;12(117):1040–1047. [PubMed] [Google Scholar]
- 124.May J.A., Sharif N.A., McLaughlin M.A., Chen H.H., Severns B.S., Kelly C.R., Holt W.F., Young R., Glennon R.A., Hellberg M.R., Dean T.R. Ocular hypotensive response in nonhuman primates of (8R)-1[(2s)-2-aminopropyl]-8,9-dihydro-7H-pyrano[2,3-g]indazol-8-ol a selective 5-HT2 receptor agonist. J. Med. Chem. 2015;58(22):8818–8833. doi: 10.1021/acs.jmedchem.5b00857. [http://dx.doi.org/10.1021/acs.jmedchem. 5b00857]. [DOI] [PubMed] [Google Scholar]
- 125.Sharif N.A. Serotonin-2 receptor agonists as novel ocular hypotensive agents and their cellular and molecular mechanisms of action: novel drug targets for glaucoma treatment. Curr. Drug Targets. 2010;11(8):978–993. doi: 10.2174/138945010791591278. [http://dx.doi.org/10.2174/138945010791591278]. [DOI] [PubMed] [Google Scholar]
- 126.Osborne N.N., Wood J.P.M., Melena J., Chao H.M., Nash M.S., Bron A.J., Chidlow G. 5-hydroxytryptamine(1A) agonists: Potential use in glaucoma. Evidence from animal studies. Eye (Lond.) 2000;14:454–463. doi: 10.1038/eye.2000.131. [http://dx.doi.org/10.1038/eye.2000.131]. [DOI] [PubMed] [Google Scholar]
- 127.Marinova Z., Walitza S., Grunblatt E. 5-HT2A serotonin receptor agonist doi alleviates cytotoxicity in neuroblastoma cells: Role of the erk pathway. Prog. Neuropsychopharmacol. Biol. Psychiatry. 2013;44:64–72. doi: 10.1016/j.pnpbp.2013.01.017. [http://dx.doi.org/10.1016/j.pnpbp.2013.01.017]. [DOI] [PubMed] [Google Scholar]
- 128.Johansen F.F., Hasseldam H., Smith M.N., Rasmussen R.S. Drug-induced hypothermia by 5HT1A agonists provide neuroprotection in experimental stroke: New perspectives for acute patient treatment. J. Stroke Cerebrovasc. Dis. 2014;23(10):2879–2887. doi: 10.1016/j.jstrokecerebrovasdis.2014.07.019. [http://dx.doi.org/10.1016/j.jstrokecerebrovasdis.2014.07.019]. [DOI] [PubMed] [Google Scholar]
- 129.Yoles E., Belkin M., Schwartz M. Hu-211, a nonpsychotropic cannabinoid, produces short- and long-term neuroprotection after optic nerve axotomy. J. Neurotrauma. 1996;13(1):49–57. doi: 10.1089/neu.1996.13.49. [http:// dx.doi.org/10.1089/neu.1996.13.49]. [DOI] [PubMed] [Google Scholar]
- 130.Eshhar N., Striem S., Kohen R., Tirosh O., Biegon A. Neuroprotective and antioxidant activities of HU-211, a novel NMDA receptor antagonist. Eur. J. Pharmacol. 1995;283(1-3):19–29. doi: 10.1016/0014-2999(95)00271-l. [http://dx.doi.org/10.1016/0014-2999(95)00271-L]. [DOI] [PubMed] [Google Scholar]
- 131.Juttler E., Potrovita I., Tarabin V., Prinz S., Tuan D.S., Fink G., Schwaninger M. The cannabinoid dexanabinol is an inhibitor of the nuclear factor-kappa b (NF-kappa B). Neuropharmacology. 2004;47(4):580–592. doi: 10.1016/j.neuropharm.2004.05.009. [http://dx.doi.org/10.1016/j.neuropharm. 2004.05.009]. [DOI] [PubMed] [Google Scholar]
- 132.Lax P., Esquiva G., Altavilla C., Cuenca N. Neuroprotective effects of the cannabinoid agonist HU210 on retinal degeneration. Exp. Eye Res. 2014;120:175–185. doi: 10.1016/j.exer.2014.01.019. [http://dx.doi.org/10.1016/j. exer.2014.01.019]. [DOI] [PubMed] [Google Scholar]
- 133.Crandall J., Matragoon S., Khalifa Y.M., Borlongan C., Tsai N.T., Caldwell R.B., Liou G.I. Neuroprotective and intraocular pressure-lowering effects of (-)delta9-tetrahydrocannabinol in a rat model of glaucoma. Ophthalmic Res. 2007;39(2):69–75. doi: 10.1159/000099240. [http://dx.doi.org/10.1159/000099240]. [DOI] [PubMed] [Google Scholar]
- 134.Adelli G.R., Bhagav P., Taskar P., Hingorani T., Pettaway S., Gul W., ElSohly M.A., Repka M.A., Majumdar S. Development of a delta9-tetrahydrocannabinol amino acid-dicarboxylate prodrug with improved ocular bioavailability. Invest. Ophthalmol. Vis. Sci. 2017;58(4):2167–2179. doi: 10.1167/iovs.16-20757. [http://dx.doi.org/10.1167/iovs.16-20757]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Panahi Y., Manayi A., Nikan M., Vazirian M. The arguments for and against cannabinoids application in glaucomatous retinopathy. Biomed. Pharmacother. 2017;86:620–627. doi: 10.1016/j.biopha.2016.11.106. [http://dx.doi.org/10. 1016/j.biopha.2016.11.106]. [DOI] [PubMed] [Google Scholar]
- 136.Sappington R.M., Calkins D.J. Contribution of trpv1 to microglia-derived il-6 and nf kappa b translocation with elevated hydrostatic pressure. Invest. Ophthalmol. Vis. Sci. 2008;49(7):3004–3017. doi: 10.1167/iovs.07-1355. [http://dx.doi.org/10.1167/iovs.07-1355]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Ward N.J., Ho K.W., Lambert W.S., Weitlauf C., Calkins D.J. Absence of transient receptor potential vanilloid-1 accelerates stress-induced axonopathy in the optic projection. J. Neurosci. 2014;34(9):3161–3170. doi: 10.1523/JNEUROSCI.4089-13.2014. [http://dx.doi.org/10.1523/JNEUROSCI. 4089-13.2014]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Weitlauf C., Ward N.J., Lambert W.S., Sidorova T.N., Ho K.W., Sappington R.M., Calkins D.J. Short-term increases in transient receptor potential vanilloid-1 mediate stress-induced enhancement of neuronal excitation. J. Neurosci. 2014;34(46):15369–15381. doi: 10.1523/JNEUROSCI.3424-14.2014. [http://dx.doi.org/10.1523/JNEUROSCI.3424-14.2014]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Sappington R.M., Sidorova T., Ward N.J., Chakravarthy R., Ho K.W., Calkins D.J. Activation of transient receptor potential vanilloid-1 (TRPV1) influences how retinal ganglion cell neurons respond to pressure-related stress. Channels. 2015;9(2):102–113. doi: 10.1080/19336950.2015.1009272. [http://dx.doi.org/10.1080/19336950.2015.1009272]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Martinez T., Gonzalez M.V., Roehl I., Wright N., Paneda C., Jimenez A.I. In vitro and in vivo efficacy of SYL040012, a novel sirna compound for treatment of glaucoma. Mol. Ther. 2014;22(1):81–91. doi: 10.1038/mt.2013.216. [http://dx.doi.org/10.1038/mt.2013.216]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Moreno-Montanes J., Sadaba B., Ruz V., Gomez-Guiu A., Zarranz J., Gonzalez M.V., Paneda C., Jimenez A.I. Phase I clinical trial of SYL040012, a small interfering rna targeting beta-adrenergic receptor 2, for lowering intraocular pressure. Mol. Ther. 2014;22(1):226–232. doi: 10.1038/mt.2013.217. [http://dx.doi.org/10.1038/mt.2013.217]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Ahmed Z., Kalinski H., Berry M., Almasieh M., Ashush H., Slager N., Brafman A., Spivak I., Prasad N., Mett I., Shalom E., Alpert E., Di Polo A., Feinstein E., Logan A. Ocular neuroprotection by siRNA targeting caspase-2. Cell Death Dis. 2011;2:e173. doi: 10.1038/cddis.2011.54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Vigneswara V., Berry M., Logan A., Ahmed Z. Pharmacological inhibition of caspase-2 protects axotomised retinal ganglion cells from apoptosis in adult rats. PLoS One. 2012;7(12):e53473. doi: 10.1371/journal.pone.0053473. [http://dx.doi.org/10.1371/journal.pone.0053473]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Solano E.C.R., Kornbrust D.J., Beaudry A., Foy J.W.D., Schneider D.J., Thompson J.D. Toxicological and pharmacokinetic properties of QPI-1007, a chemically modified synthetic siRNA targeting caspase 2 mRNA, following intravitreal injection. Nucleic Acid Ther. 2014;24(4):258–266. doi: 10.1089/nat.2014.0489. [http://dx.doi.org/ 10.1089/nat.2014.0489]. [DOI] [PubMed] [Google Scholar]
- 145.Chen Y.J., Tai M.C., Cheng J.H., Chen J.T., Chen Y.H., Lu D.W. The longitudinal changes of the visual field in an asian population with primary angle-closure glaucoma with and without an acute attack. J. Ocul. Pharmacol. Ther. 2012;28(5):529–535. doi: 10.1089/jop.2012.0006. [http://dx.doi.org/10.1089/jop.2012.0006]. [DOI] [PubMed] [Google Scholar]
- 146.Steinhausen K., Stumpff F., Strauss O., Thieme H., Wiederholt M. Influence of muscarinic agonists and tyrosine kinase inhibitors on l-type Ca2+ channels in human and bovine trabecular meshwork cells. Exp. Eye Res. 2000;70(3):285–293. doi: 10.1006/exer.1999.0785. [http://dx.doi.org/10. 1006/exer.1999.0785]. [DOI] [PubMed] [Google Scholar]
- 147.Stumpff F., Wiederholt M. Regulation of trabecular meshwork contractility. Ophthalmologica. 2000;214(1):33–53. doi: 10.1159/000027471. [http://dx. doi.org/10.1159/000027471]. [DOI] [PubMed] [Google Scholar]
- 148.Cellini M., Versura P., Trere D., Campos E.C. Effects of endothelin-1 on human trabecular meshwork cell contraction - an in vitro cell culture model. Ophthalmic Res. 2005;37(1):43–49. doi: 10.1159/000083021. [http://dx.doi.org/10.1159/000083021]. [DOI] [PubMed] [Google Scholar]
- 149.Zaidel-Bar R., Guo Z.H., Luxenburg C. The contractome - a systems view of actomyosin contractility in non-muscle cells. J. Cell Sci. 2015;128(12):2209–2217. doi: 10.1242/jcs.170068. [http://dx.doi.org/10.1242/ jcs.170068]. [DOI] [PubMed] [Google Scholar]
- 150.Van de Velde S., Van Bergen T., Vandewalle E., Kindt N., Castermans K., Moons L., Stalmans I. Rho kinase inhibitor AMA0526 improves surgical outcome in a rabbit model of glaucoma filtration surgery. 2015. [DOI] [PubMed] [Google Scholar]
- 151.Shaw P.X., Sang A., Wang Y., Ho D., Douglas C., Dia L., Goldberg J.L. Topical administration of a ROCK/NET inhibitor promotes retinal ganglion cell survival and axon regeneration after optic nerve injury. Exp. Eye Res. 2017;158:33–42. doi: 10.1016/j.exer.2016.07.006. [http://dx.doi. org/10.1016/j.exer.2016.07.006]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Van de Velde S., De Groef L., Stalmans I., Moons L., Van Hove I. Towards axonal regeneration and neuroprotection in glaucoma: Rho kinase inhibitors as promising therapeutics. Prog. Neurobiol. 2015;131:105–119. doi: 10.1016/j.pneurobio.2015.06.002. [http://dx.doi.org/10.1016/j.pneurobio. 2015.06.002]. [DOI] [PubMed] [Google Scholar]
- 153.Novack G.D. Eyes on new product development. J. Ocul. Pharmacol. Ther. 2017;33(2):65. doi: 10.1089/jop.2017.29024.gdn. [http://dx.doi.org/10.1089/jop. 2017.29024.gdn]. [DOI] [PubMed] [Google Scholar]
- 154. http://www.reuters.com/article/brief-aerie-pharmaceuticals-announces-fd-idUSASA09PN3
- 155.Wang R.F., Williamson J.E., Kopczynski C., Serle J.B. Effect of 0.04% AR-13324, a ROCK, and norepinephrine transporter inhibitor, on aqueous humor dynamics in normotensive monkey eyes. J. Glaucoma. 2015;24(1):51–54. doi: 10.1097/IJG.0b013e3182952213. [http://dx.doi.org/10.1097/ IJG.0b013e3182952213]. [DOI] [PubMed] [Google Scholar]
- 156.Garnock-Jones K. Ripasudil: First global approval. Drugs. 2014;74(18):2211–2215. doi: 10.1007/s40265-014-0333-2. [http://dx.doi.org/10.1007/s40265-014-0333-2]. [DOI] [PubMed] [Google Scholar]
- 157.Matsumura R., Inoue T., Matsumura A., Tanihara H. Efficacy of ripasudil as a second-line medication in addition to a prostaglandin analog in patients with exfoliation glaucoma: a pilot study. Clin. Drug Investig. 2017;37(6):535–539. doi: 10.1007/s40261-017-0509-0. [http://dx.doi.org/10.1007/ s40261-017-0509-0]. [DOI] [PubMed] [Google Scholar]
- 158.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(11):7126–7136. doi: 10.1167/iovs.13-13842. [http://dx.doi.org/10.1167/iovs.13-13842]. [DOI] [PubMed] [Google Scholar]
- 159.Yamaguchi M., Nakao S., Arita R., Kaizu Y., Arima M., Zhou Y.D., Kita T., Yoshida S., Kimura K., Isobe T., Kaneko Y., Sonoda K., Ishibashi T. Vascular normalization by ROCK inhibitor: Therapeutic potential of ripasudil (K-115) eye drop in retinal angiogenesis and hypoxia. Invest. Ophthalmol. Vis. Sci. 2016;57(4):2264–2276. doi: 10.1167/iovs.15-17411. [http://dx.doi.org/10.1167/iovs.15-17411]. [DOI] [PubMed] [Google Scholar]
- 160.Harrison B.A., Whitlock N.A., Voronkov M.V., Almstead Z.Y., Gu K.J., Mabon R., Gardyan M., Hamman B.D., Allen J., Gopinathan S., McKnight B., Crist M., Zhang Y.L., Liu Y., Courtney L.F., Key B., Zhou J., Patel N., Yates P.W., Liu Q.Y., Wilson A.G.E., Kimball S.D., Crosson C.E., Rice D.S., Rawlins D.B. Novel class of Lim-kinase 2 inhibitors for the treatment of ocular hypertension and associated glaucoma. J. Med. Chem. 2009;52(21):6515–6518. doi: 10.1021/jm901226j. [http://dx.doi.org/10.1021/jm901226j]. [DOI] [PubMed] [Google Scholar]
- 161.Zhang M., Maddala R., Rao P.V. Novel molecular insights into RhoA GTPase-induced resistance to aqueous humor outflow through the trabecular meshwork. Am. J. Physiol. Cell Physiol. 2008;295(5):C1057–C1070. doi: 10.1152/ajpcell.00481.2007. [http://dx.doi.org/10.1152/ajpcell. 00481.2007]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Wang W.W., Townes-Anderson E. Lim kinase, a newly identified regulator of presynaptic remodeling by rod photoreceptors after injury. Invest. Ophthalmol. Vis. Sci. 2015;56(13):7847–7858. doi: 10.1167/iovs.15-17278. [http://dx.doi.org/10.1167/iovs.15-17278]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Pfeiffer N., Lamparter J., Gericke A., Grus F.H., Hoffmann E.M., Wahl J. Neuroprotection of medical IOP-lowering therapy. Cell Tissue Res. 2013;353(2):245–251. doi: 10.1007/s00441-013-1671-1. [http://dx.doi.org/10.1007/ s00441-013-1671-1]. [DOI] [PubMed] [Google Scholar]
- 164.Galindo-Romero C., Harun-Or-Rashid M., Jimenez-Lopez M., Vidal-Sanz M., Agudo-Barriuso M., Hallbook F. Neuroprotection by alpha 2-adrenergic receptor stimulation after excitotoxic retinal injury: A study of the total population of retinal ganglion cells and their distribution in the chicken retina. PLoS One. 2016;11(9):e0161862. doi: 10.1371/journal.pone.0161862. [http://dx.doi.org/10.1371/journal.pone.0161862]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Lindsey J.D., Duong-Polk K.X., Hammond D., Chindasub P., Leung C.K.S., Weinreb R.N. Differential protection of injured retinal ganglion cell dendrites by brimonidine. Invest. Ophthalmol. Vis. Sci. 2015;56(3):1789–1804. doi: 10.1167/iovs.14-13892. [http://dx.doi.org/10.1167/iovs. 14-13892]. [DOI] [PubMed] [Google Scholar]
- 166.Guo X.L., Namekata K., Kimura A., Noro T., Azuchi Y., Semba K., Harada C., Yoshida H., Mitamura Y., Harada T. Brimonidine suppresses loss of retinal neurons and visual function in a murine model of optic neuritis. Neurosci. Lett. 2015;592:27–31. doi: 10.1016/j.neulet.2015.02.059. [http://dx.doi.org/10.1016/j.neulet.2015.02.059]. [DOI] [PubMed] [Google Scholar]
- 167.Lee D., Kim K.Y., Noh Y.H., Chai S., Lindsey J.D., Ellisman M.H., Weinreb R.N., Ju W.K. Brimonidine blocks glutamate excitotoxicity-induced oxidative stress and preserves mitochondrial transcription factor a in ischemic retinal injury. PLoS One. 2012;7(10):e47098. doi: 10.1371/journal.pone.0047098. [http://dx.doi.org/10.1371/journal.pone.0047098]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Pinar-Sueiro S., Urcola H., Rivas M.A., Vecino E. Prevention of retinal ganglion cell swelling by systemic brimonidine in a rat experimental glaucoma model. Clin. Experiment. Ophthalmol. 2011;39(8):799–807. doi: 10.1111/j.1442-9071.2011.02573.x. [http://dx.doi.org/10.1111/j.1442-9071.2011.02573.x]. [DOI] [PubMed] [Google Scholar]
- 169.Nizari S., Guo L., Davis B.M., Normando E.M., Galvao J., Turner L.A., Bizrah M., Dehabadi M., Tian K.L., Cordeiro M.F. Non-amyloidogenic effects of alpha 2 adrenergic agonists: implications for brimonidine-mediated neuroprotection. Cell Death Dis. 2016;7:e2514. doi: 10.1038/cddis.2016.397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Yamagishi R., Aihara M., Araie M. Neuroprotective effects of prostaglandin analogues on retinal ganglion cell death independent of intraocular pressure reduction. Exp. Eye Res. 2011;93(3):265–270. doi: 10.1016/j.exer.2011.06.022. [http://dx.doi.org/10.1016/j.exer.2011.06.022]. [DOI] [PubMed] [Google Scholar]
- 171.Vidal L., Diaz F., Villena A., Moreno M., Campos J.G., de Vargas I.P. Reaction of muller cells in an experimental rat model of increased intraocular pressure following timolol, latanoprost and brimonidine. Brain Res. Bull. 2010;82(1-2):18–24. doi: 10.1016/j.brainresbull.2010.02.011. [http://dx. doi.org/10.1016/j.brainresbull.2010.02.011]. [DOI] [PubMed] [Google Scholar]
- 172.Kashiwagi K., Tsumura T., Tsukahara S. Long-term effects of latanoprost monotherapy on intraocular pressure in japanese glaucoma patients. J. Glaucoma. 2008;17(8):662–666. doi: 10.1097/IJG.0b013e318166656d. [http://dx.doi. org/10.1097/IJG.0b013e318166656d]. [DOI] [PubMed] [Google Scholar]

