Open access

Neuroprotection in Glaucoma

Written By

Sotiria Palioura and Demetrios G. Vavvas

Submitted: 11 April 2012 Published: 17 April 2013

DOI: 10.5772/54294

From the Edited Volume

Glaucoma - Basic and Clinical Aspects

Edited by Shimon Rumelt

Chapter metrics overview

2,481 Chapter Downloads

View Full Metrics

1. Introduction

Glaucoma is a distinctive group of optic neuropathies characterized by progressive degeneration of neuronal tissue due to death of retinal ganglion cells, with accompanying gradual visual field loss. [1, 2] It is the leading cause of irreversible blindness worldwide [3] and complex genetic and environmental risk factors have been implicated in its progression. [4-7] Neuroprotection for glaucoma refers to any intervention that aims either to prevent optic nerve damage and retinal ganglion cell death or to preserve already diseased neuronal tissue and its function, with the ultimate goal of maintaining vision. Thus, neuroprotective agents can be thought of as pharmacological antagonists of intracellular injury and death pathways.

Agents that lower the intra-ocular pressure (IOP) have been shown to slow glaucoma progression in several controlled clinical trials and even arrest the progression in some cases [8-10], yet their effectiveness is limited in preventing retinal ganglion cell loss. Retinal ganglion cell damage in glaucoma is not confined to the neurons that are insulted primarily, but neighboring neurons are injured secondarily as well. [11] Therefore, efforts that focus on discovering alternative therapeutic approaches independent of IOP reduction have placed neuroprotective treatment modalities at the frontiers of glaucoma research.

Advertisement

2. Apoptosis and necrosis in glaucoma

Apoptosis and necrosis constitute the two major pathways to cell death. [12] In 1972, Kerr, Wyllie and Currie used the Greek term ‘apoptosis’ (from the Greek: dropping off of petals from plants) to describe a specific morphological aspect of cell death. [13] Apoptosis is accompanied by rounding-up of the cell, reduction of cellular volume, chromatin condensation, and engulfment by resident phagocytes. Apoptosis is the best-characterized type of programmed cell death, and these morphological changes are largely mediated by the activation of the caspase family of cysteine proteases. [14] In contrast, ‘necrosis’ (from the Greek: death) is associated with a gain in cell volume, swelling of organelles, plasma membrane rupture and subsequent release of intracellular contents with ensuing inflammation. Until recently necrosis had been considered a passive, unregulated form of cell death. New evidence indicates that some forms of necrosis can be induced by regulated signal transduction pathways such as those mediated by receptor interacting protein kinases (RIP Kinases). RIP kinases cross talk with caspases and lie downstream of cell death signals such as the Fas Ligand or the Tumor Necrosis Factor-α (TNF-α). [15] This programmed form of necrosis is termed programmed necrosis or necroptosis. [12, 16, 17]

Figure 1.

The extrinsic pathway is initiated by binding of death ligands such as TNF-α and Fas ligand to their cell-surface death receptors such as TNF receptor and Fas. The death domains of these receptors recruit adaptor molecules like FADD and caspase-8, which leads to the activation of caspase-8. Activated caspase-8 cleaves the effector caspases such as caspase-3, thereby activating them and inducing apoptosis. The extrinsic pathway interacts with the intrinsic pathway via caspase-8-mediated cleavage of Bid. The intrinsic pathway is initiated by release of mitochondrial intermembrane proteins such as cytochrome c and Smac/Diablo into the cytosol. Released cytochrome c forms an apoptosome with Apaf-1 and caspase-9, which leads to caspase-9 activation. Smac/Diablo enhances caspase activation through the neutralization of IAP proteins.

Cysteine aspartate-specific proteases or caspases are central to the execution of apoptosis. Their activation occurs mainly through two distinct pathways: extrinsic and intrinsic (Fig. 1). [18] The extrinsic pathway is initiated by binding of extracellular death ligands such as TNF-α and Fas ligand to their cell-surface death receptors, TNF receptor and Fas. [19] The death domains of these receptors recruit adaptor molecules like Fas-associated death domain (FADD) and caspase-8, forming the death inducing signaling complex (DISC). [20] The formation of DISC leads to activation of caspase-8, which in turn mediates cleavage of effector caspases. The extrinsic pathway can cross-talk with the intrinsic pathway through caspase-8-mediated cleavage of Bid, aBH3-only member of the Bcl-2 family of proteins. [21, 22] Bid cleavage releases a truncated fragment that triggers the release of mitochondrial proteins, thereby initiating the intrinsic caspase cascade as described below.

The intrinsic pathway is mediated by mitochondria. [23] In response to intracellular and environmental stress, mitochondria release inter-membrane proteins such as cytochrome c and second mitochondria-derived activator of caspases (Smac)/direct inhibitor of apoptosis-binding protein with low pI (Diablo) into the cytosol. Released cytochrome c triggers the formation of an apoptosome along with apoptotic protease activating factor-1 (Apaf-1) and caspase-9 in the presence of ATP, which leads to caspase-9 activation. [24] Smac/Diablo enhances caspase activation through the neutralization of inhibitor of apoptosis (IAP) (Fig. 1). [25, 26]

Necrosis is mainly regulated by a set of protein Kinases called RIP Kinases. RIP1 switches its function to a regulator of cell death when it is deubiquitinated by A20 or cylindromatosis (CYLD). [27, 28] Deubiquitination of RIP1 abolishes its ability to activate NF-κB after TNF-α stimulation, and leads to the formation of cytosolic DISC with FADD and caspase-8, the so-called complex II. [29] As described above in caspase signaling, DISC formation leads to caspase-8 activation and subsequent apoptosis. In contrast to TNF signaling, Fas directly recruits RIP1, FADD and caspase-8 to the plasma membrane and forms DISC (Fig. 2). [30] During apoptosis, RIP1 is cleaved and inactivated by caspases. [31] Although many cell lines are protected against death receptor-induced apoptosis by use of pan-caspase inhibitors, Vercammen and others found that, in mouse L929 fibrosarcoma cells, caspase inhibition does not prevent TNF- or Fas-induced cell death and the cells acquire a necrotic morphology. [32, 33] In 2000, Holler and others discovered that RIP1 kinase is a key molecule that induces necrotic cell death mediated by death receptors. [34]

In 2005, Degterev, Yuan, and others using chemical library screening, identified small compounds named necrostatins that specifically inhibit death receptor- mediated necrosis. [16] Necrostatins have been shown to specifically inhibit RIP1 kinase phosphorylation during necrosis without affecting death receptor-induced NF-κB activation. [35] RIP1 kinase activity appears to be important for necrosome formation, as necrostatin-1 abolishes the formation of the RIP1-RIP3 complex and RIP3 kinase phosphorylation during necrosis. [36, 37] Cho and others propose that another unknown kinase activated by RIP1 may mediate RIP3 phosphorylation, based on the findings that ectopically expressed RIP1 does not phosphorylate RIP3. [36] The activities of RIP1 and RIP3 may be mutually regulated in a necrosome signaling complex. RIPK activation leads likely to increased reactive oxygen species (ROS) production. Activated RIP3 interacts with metabolic enzymes such as glycogen phosphorylase (PYGL), glutamate-ammonia ligase (GLUL) and glutamate dehydrogenase 1 (GLUD1). [38] Activation of these enzymes eventually stimulates the Krebs cycle and oxidative phosphorylation, thereby increasing mitochondrial ROS production. Secondly, after TNF-α stimulation, RIP1 forms a complex with TNFR, Riboflavin kinase, and NADPH oxidase 1. [39, 40] NADPH oxidase is the best characterized non-mitochondrial source of ROS and forms a membrane bound enzyme complex with p22phox and Rac. [41] Thirdly, RIP1 kinase activates autophagic degradation of catalase, which converts hydrogen peroxide to water and oxygen, thereby increasing ROS accumulation. [42] More recently, activation of the necrosome has shown to interact with the mixed lineage kinase domain-like (MLKL) and phosphoglycerate mutase 5 (PGAM5) resulting in the fusion of mitochondria and necrotic cell death. [43, 44]

Figure 2.

Schematic of the RIPK signaling pathway.
A, In response to TNF-α stimulation, RIP1 is recruited to TNFR and forms a membrane associated complex I with TRADD, TRAF2/5 and cIAP1/2, which in turn leads to polyubiquitination of RIP1 and pro-survival NF-κB activation. 
B, RIP1 switches function to a regulator of cell death when RIP1 is unubiquitinated by A20 or CYLD. Deubiquitination of RIP1 leads to the formation of cytosolic DISC with FADD and caspase-8, the so-called complex II. In contrast to TNF signaling, Fas stimulation directly forms DISC. Activation of caspase-8 in DISC leads to apoptosis induction. During apoptosis, RIP1 is cleaved and inactivated by caspase-8. C, In conditions where caspases are blocked or cannot be activated efficiently, RIP1 binds to RIP3, and both RIP1 and RIP3 kinases are phosphorylated at the RIP1-RIP3 complex. RIP1 kinase phosphorylation is critical for necrosis induction. In response to TNF-α, RIP1 binds to NADPH oxidase 1 and produces superoxide. Activated RIP3 binds to PYGL, GLUL and GLUD1 and increases the production of mitochondrial ROS. ROS overproduction leads to mitochondrial dysfunction, resulting in the release of mitochondrial pro-death proteins. Activation of the necrosome has been shown to interact with mixed lineage kinase domain-like (MLKL) and phosphoglycerate mutase 5 (PGAM5) resulting in the fusion of mitochondria and necrotic cell death

Figure 3.

Schematic of changes in animal models of high IOP mediated optic nerve damage. A, In normal IOP microglia are quiescent and cells are in normal state. B, Elevated IOP leads to increased numbers of activated microglia with amoeboid morphology around the optic nerve head. These microglia appear to secrete TNF-α leading to RGC death. Other molecules, including FasL on microglia, nitric oxide (NO), and reactive oxygen species (ROS) may also play a role in RGC death. Changes in blood supply and ischemia also contribute to the death of RGCs.

In chronic glaucoma, apoptosis of retinal ganglion cells has been shown as the main pathway to cell death. [2, 45, 46] The exact mechanism though is not clear. Since a significant proportion of patients who suffer from glaucoma have high IOP, it has been hypothesized that high IOP induces stress to retinal ganglion cells either directly [47, 48] or indirectly to their axons at the lamina cribrosa [49] thus leading to apoptosis. However, although high IOP has been thought to be the main causative factor, the fact that glaucoma can occur in the presence of IOP within the normal range, while can be absent in a subset of subjects with high IOP indicates that the underlying etiology of this disease remains unknown and in essence fail to fully fulfill Koch’s postulates [50, 51].

Mechanisms believed to cause stress to retinal ganglion cells and to initiate the apoptotic cascade include: biomechanical stress [52, 53], excitotoxicity [54-57], tissue hypoxia [58, 59], altered nutritional blood supply [60, 61], mitochondrial dysfunction [62-65], Müller glial cell activation [66], protein misfolding [67-69], oxidative stress [70, 71], dysfunctional autoimmunity [72], neurotrophin deprivation [73, 74], and inflammation. [75, 76]

Advertisement

3. Animal models

Animal experimental models in glaucoma research are produced by inducing either an elevation in intraocular pressure or damage to the axons of retinal ganglion cells. [77] Several methods have been employed to raise intraocular pressure in animal models. They range from obliteration of episcleral vessels [78] to iatrogenic sclerosis of the trabecular meshwork (laser-induced [79] or through retroinjection of hypertonic saline into the limbal plexus [80]) or to mechanical obstruction of the trabecular meshwork with polystyrene beads. [81] Direct damage to retinal ganglion cell axons can be achieved via axotomy or crushing of axons. Retinal ganglion cell death occurs in 1-2 weeks from the time of optic nerve transection or crushing of axons in a fairly predictable fashion. [82] Interestingly, there is a specific mouse strain (DBA/J2) which inherently produces a much slower degeneration of retinal ganglion cells; a process thought to more closely mimic human disease than other induced mouse models of glaucoma. [83] Mutations in the transmembrane glycoprotein Gpnmb (premature stop codon at position 150 -GpnmbR150X) and the presence of the Tyrp1b gene allele (the b mutation is in a heme-associated domain and renders the protein susceptible to rapid proteolytic degradation) in the DBA/2J mouse model are thought to cause pigment dispersion and iris atrophy respectively. Both proteins are highly expressed in melanocytes and are involved in melanin and cell growth regulation. The decreased levels of Gpnmb and Tyrp1 lead to cell death and abnormal melanin content release that deposits onto the trabecular meshwork resulting in elevated IOP. [84] Of note, these mutations have not been shown to cause glaucoma in patients.

The ideal animal model should manifest slow focal injury to retinal ganglion cell axons at the optic nerve head that leads to sectoral death of retinal ganglion cells without loss of other retinal neurons. [77] The currently used animal models for glaucoma research are far from ideal and their limitations are partly responsible for the failure to translate results from the bench to the bedside. For the elevated IOP models, it is clear that there are different susceptibilities of retinal ganglion cell damage among different species (mice, rats, monkeys) and among different strains or age of the same species. [51, 85, 86] For the axotomy models, acute damage to retinal ganglion cell axons is different from the slow progression seen clinically in glaucoma, and thus it remains unclear whether studies with these models can safely reproduce glaucomatous damage as it occurs in humans. [82] For the DBA/2J mouse, there is significant variability in glaucoma progression among animals and between eyes of the same animal, which renders any comparison study particularly difficult. [77]

An inherent limitation of using animal models for the study of any human disease process is that animal models often lack the heterogeneity, compounded comorbidities, and polypharmacy that are present in human pathologic conditions. Moreover, it is difficult to extrapolate from animal studies what the appropriate dose of a putative neuroprotective agent would be in human subjects since pre-clinical studies rarely assess for a dose-response curve, therapeutic index, and central nervous system penetration. [87, 88]

Advertisement

4. Success in the lab

Efforts in pre-clinical studies have targeted the various mechanisms that produce axonal degeneration and retinal ganglion cell death and have led to the discovery of an array of neuroprotective agents. [89] First, apoptosis of retinal ganglion cells has been inhibited in the lab through the use of anti-excitotoxic agents that primarily inhibit or interfere with the glutamate excitotoxic cascade. [90] Glutamate is a natural neurotransmitter that is required by the organism for proper cell signaling including retinal ganglion cells. Glutamate acts through many types of glutamate receptors/ion channels. One of these receptors/ion channels is the N-methyl-D-aspartate (NMDA) type, which leads to calcium flux upon activation. Persistent activation of this channel by glutamate or NDMA leads to excitotoxicity. Glutamate induced excitotoxicity has been shown in some but not all animal models and elevated glutamate levels have been detected in some but not all studies (reviewed in [91]). Memantine is an uncompetitive antagonist of the N-methyl-D-aspartate (NMDA) type of glutamate receptor/channel. It can only bind to this ion channel in its “open” state, that is after glutamate has already bound to its receptor and has caused the channel to open. [92] Studies on several animal models of glaucoma have supported the neuroprotective role of memantine on retinal ganglion cells. [93]. Activated glutamate receptor leads to increased calcium flux. Calcium levels are important in many neuronal signaling events and aberrant calcium levels are thought to be important mediators of neuronal cell death. Increased levels of intracellular calcium can be very detrimental to the health of the cell. Most recently, inhibitors of the L-type voltage-gated calcium channel, clinidipine [94] and lomerizine [95] as well as the alpha-2 adrenergic agonist brimonidine [96] have also been shown to limit glutamate-induced excitotoxicity. Although the exact mechanism of action of brimonidine remains unknown, intraperitoneal pre-treatment with brimonidine has been shown by several groups to increase survival of retinal ganglion cells after optic nerve or retinal injury in animal models of NDMA excitotoxicity, optic nerve crush and ischemia. [97-100]

Antiapoptotic strategies utilize neurotrophins [101] or aim at the activation of Bcl-2 antiapoptotic pathways. [102] Neurotrophins are factors that signal the survival and growth of neurons. The first neurotrophin to be discovered was Nerve Growth Factor (NGF) in the 1960s. In 1986 Levi-Montalcini and Cohen shared the Nobel prize “for their discovery of growth factors for neurons.” Neurotrophins are peptides that bind to cell surface receptors and activate survival signals, thus suppressing the apoptotic process (Fig. 4). [80, 103, 104] Exogenous administration of brain-derived neurotrophic factor (BDNF) or nerve growth factor (NGF) has delayed but not prevented retinal ganglion cell death. [105-108] Injection of BDNF, ciliary neurotrophic factor (CNTF), neurotrophin-4 (NT-4), fibroblast growth factor-2 (FGF-2), and neurturin (a ligand for a glial cell line-derived neurotrophic factor family related receptor A2 - GFRA2) into the vitreous has also increased survival of retinal ganglion cells. [105, 109, 110] Neurotrophin delivery and overexpression via viral vector delivery seems to promote survival of retinal ganglion cells even further. [106] Finally, agents that interact with the two major neurotrophin cell surface receptor systems, tropomyosin-related kinase (Trk) receptor and p75 neurotrophin receptor (p75NTR), can also prolong survival of retinal neuronal tissue (Fig. 4). [111-113] Neurotrophins are difficult to be used in clinics because of their polypeptidic nature: they are destroyed in the acidic milieu of the stomach, while their size hinders their ability of crossing the blood-brain barrier. Thus, special techniques have to be invented, such as intravitreal implants of cells producing these molecules locally, such as the CNTF producing cells by Neurotech (Cumberland, RI, USA).

Figure 4.

Neurotrophin Signaling Summary. (courtesy of Dr A. Gravanis). Neurotrophins include the first to be discovered: Nerve Growth Factor (NGF) as well as Brain Derived Neurotrophic Factor (BDNF) and Neurotrophin 3 and 4 (NT-3 and NT-4). There are two classes of receptors for neurotrophins: p75 and the "Trk" family of Tyrosine kinase receptors. P75 can bind all factors, whereas Trk receptors are specific for their ligands. Binding of neurotrophins to their receptors leads to activation of many pro-survival signals including PI3K, Akt, and MAPK among others.

Tumor necrosis factor-alpha (TNF-α) can lead to death of retinal ganglion cells by inducing mitochondrial damage and activation of caspases. [114] Inhibition of TNF-α via the use of etanercept [115] or an anti-TNF-α neutralizing antibody [116] can protect retinal ganglion cells in mouse models of glaucoma.

Providing free radical scavengers, such as coenzyme Q10 [117, 118] or thioredoxin [119], or inhibiting the formation of free radicals by blocking the action of nitric oxide synthase [120] has shown promise as an alternative neuroprotective strategy. Inflammatory and immune mechanisms also play a role in glaucomatous damage of retinal ganglion cells. [121] Immunomodulation [122], inhibition of calcineurin by tacrolimus [123], and subcutaneous injection of granulocyte-colony stimulating factor [124] also have neuroprotective potential in glaucoma.

Other mechanisms that have been investigated in the lab with encouraging results include the use of mesenchymal stem cells, which are thought to exert their effect through production of neurotrophins or stimulation of inflammation [125, 126]. A recent study has shown that amyloid β (Aβ) is found to be elevated in an animal model of ocular hypertension induced glaucoma and that inhibition of the generation of amyloid β led to preservation of RGCs. [67] Other studies have shown that treatment with minocycline reduces RGC death in experimental glaucoma. [127] The antiapoptotic effects of minocycline are not very clear and seem to be pleiotropic. They are exerted, at least in part, by modulating inflammation and metalloproteinases and by reducing mitochondrial calcium overloading. Minocycline stabilizes the mitochondrial membrane and inhibits release of cytochrome c and other apoptotic factors into the cytoplasm, thus resulting in decreased caspase activation and nuclear damage. Minocycline also exerts caspase-independent neuroprotective effects including upregulation of anti-apoptotic factor Bcl-2. Another promising agent is Rasagiline, a monoamine oxidase inhibitor that has been found have neuroprotective and anti-apoptotic effects partially through increase in the mitochondrial family of Bcl-2 proteins, prevention in the fall in mitochondrial membrane potential, prevention of the activation of caspase 3, and of translocation of glyceraldehyde-3-phosphate dehydrogenase from the cytoplasm to the nucleus. It can also affect the secretion of amyloid precursor protein (APP) and it has been shown to delay RGC cell death in experimental glaucoma [128]. However, it has to be noted that it has not taken approval by the United States Food and Drug Administration in a Parkinson’s disease trial. Erythropoietin (EPO) activates the NF-κB pathway and results in pro-survival and anti-oxidant enzyme upregulation and has been shown to be protective of RGC in the DBA/2J mouse of pigmentary glaucoma [129]. Citicholine (cytidine 5’-diphosphocholine) which exhibits neuroprotective effects by preserving cardiolipin and sphingomyelin among other actions, has been tested in patients with anterior ischemic optic neuropathy and showed preliminary benefits. [130]

Advertisement

5. Difficulties in designing a clinical trial

Over the last 30 years, numerous pharmacologic agents and gene therapeutic approaches have been shown to be neuroprotective in animal models of retinal and optic nerve injury. To date, none of the trials on neuroprotection of the visual system have shown efficacy and none of the agents developed in the laboratory have translated into a definitive clinical treatment. [131] There are several reasons for the universal failure of clinical trials to confirm pre-clinical results; they stem from the nature of glaucoma itself and from the poor design of previous neuroprotective trials. [51, 131] First, the long, slow course of glaucomatous optic neuropathy hinders our efforts to measure whether an improvement in progressive worsening has been achieved and asks for a design of therapeutic trials that last several years. Second, the rate of worsening varies among patients and thus a larger sample size is required to account for this inherent variability in disease progression. Third, the current standard of outcome measurement remains visual field testing and this carries a significant test-retest variance even among patients who are adept in taking the test. Fourth, any neuroprotection clinical trial would have to include patients that are already on topical medications that lower IOP. IOP-lowering agents have proven effective in slowing glaucoma progression in several controlled clinical trials [8-10] and it would be unethical to preclude neuroprotection study patients from using IOP-lowering medications.

Advertisement

6. Failed and ongoing trials

Success in the lab has not paralleled success in neuroprotection clinical trials. A Cochrane review in 2010 failed to identify any neuroprotection studies with significant results. [132] The largest neuroprotection study to date consisted of two industry-supported, parallel, randomized, phase III clinical trials on oral memantine in patients with chronic progressive open angle glaucoma. A total of 2,200 patients were enrolled into the trials at 89 sites and they were followed for at least 4 years. Despite the success of memantine in animal models of glaucoma, both clinical trials failed to show efficacy with respect to their primary outcome measures. The results of these two trials are as of yet unpublished and only two press releases hinted on their results. [133] The first release stated, “Two measures of visual function were selected in the statistical analysis plan to assess the efficacy of memantine in glaucoma. The functional measure chosen as the endpoint (glaucomatous field progression) did not show a benefit of memantine in preserving visual function. In a number of analyses using the secondary functional measure, memantine demonstrated a statistically significant benefit of the high-dose compared to placebo.” The second release stated, “Allergan unmasked the second Phase III clinical trial examining the safety and efficacy of oral memantine as a treatment for glaucoma. Although the study showed that the progression of disease was significantly lower in patients receiving the higher dose of memantine compared to patients receiving the low dose of memantine, there was no significant benefit compared to patients receiving placebo. Therefore, the study failed to meet its primary endpoint and to sufficiently replicate the results of the first Phase III trial.” In going forward, knowing the specifics of these trials and the reasons for failure would facilitate a more well-thought design of future trials; yet, the results remain unpublished.

The second most studied agent in neuroprotection trials is the highly selective alpha-2 adrenergic agonist, brimonidine, which had also shown great promise in animal studies. The first trial included 9 patients with Leber’s hereditary optic neuropathy in whom use of brimonidine after loss of vision in one eye failed to prevent loss of vision in the second eye, which naturally occurs within weeks to months after first eye involvement. [134] The second trial also failed to show efficacy of brimonidine in aiding recovery of vision loss in patients with anterior ischemic optic neuropathy, though the trial itself may have been underpowered. [135]

The last trial assessing the neuroprotective effects of brimonidine was the Low-Pressure Glaucoma Treatment Study, which recruited patients with normal-tension glaucoma and randomized them to either treatment with brimonidine or with timolol. [136] Timolol has no neuroprotective properties and it thus served as a control for the pressure-lowering effects of brimonidine. However, the IOP was only minimally lowered in both groups, which raises concerns about patient adherence to the treatment regimen. [51] Results of the trial were published in 2011 and showed that low-pressure glaucoma patients treated with brimonidine were less likely to have visual field progression than patients treated with timolol. [137]

A review of ongoing trials at clinicaltrials.gov revealed one phase I trial that is still recruiting patients and aims to investigate the safety and efficacy of the NT-501 clinical neurotrophic factor (CNTF) implant in patients with chronic progressive open angle glaucoma (trial identifier: NCT01408472).

Advertisement

7. Hope for success

Designing neuroprotection trials in glaucoma is challenging. Modifications in study design, patient selection, and outcome measures can aid in the clinical testing of neuroprotective agents with positive pre-clinical results. [51] Instead of the standard randomized controlled clinical trial prototype, neuroprotection trials in glaucoma may be served better by using a so-called futility design strategy. Detection of beneficial agents with robust treatment effects in a short period of time in a single treatment group (i.e. there is no need for a control group) are advantages of futility design. Clearly, a major disadvantage of this approach is the inability to adequately assess for side effects and time-dependent treatment effects in a trial that has fewer patients and a shorter testing time frame. [138, 139] In addition, selection of patients whose disease is rapidly can maximize the opportunity to detect differences after the use of neuroprotective agents. Older age, higher baseline intraocular pressure, bilateral disease, low perfusion pressure, presence of exfoliation, disc hemorrhages, and thinner central corneal thickness are all risk factors for rapid progression and should be used in the selection of the study population in neuroprotection trials. [51, 140] The agent in question also should reach its target tissue(s), the retina and optic nerve head. Steps should be taken to ensure patient adherence with medication administration or the results of any neuroprotective trial that is performed on a background of co-administered IOP-lowering therapy are deemed to be confounded. [141]. If the IOP is reduced to an identical degree, while one agent (like brimonidine or other neuroporotectant) shows fewer injuries to the visual fields, this would indirectly support the additional neuroprotective effect of the agent on top of its IOP lowering effects.

In terms of endpoints for such trials, visual field testing is likely more suitable than structural measures since it has been employed extensively to measure progression of disease. Its disadvantages of high variability in some test point areas, the patient effort it requires, and the insensitivity to show the earliest stages of damage are well known. Nevertheless, it has been well established as a method to assess glaucoma progression. Given the lack of reliable software to measure progression using structural tests, such as the Heidelberg Retinal Tomograph or Optical Coherence Tomography, visual field testing remains the most reliable endpoint to use. If structural measures are to be used in the future, one should keep in mind that the more optic nerve damage present at the outset, the less sensitive structural change will be. [51]

Advertisement

8. Conclusion

Novel neuroprotective agents and mechanisms show promise in pre-clinical studies and animal models. However, translating these findings into effective treatments still remains a challenge. This challenge can be met by a careful study design, appropriate selection of the study population, and use of better outcome measures and clinical end points. In addition, the various laboratory investigations suggest that there are multiple pathways that play a role in the loss of retinal ganglion cells. It is thus necessary to espouse combinatorial treatment approaches, if we want to successfully provide neuroprotection in the clinical setting.

Advertisement

Acknowledgments

We would like to thank Aristomenis Thanos for his assistance with the figures included in this chapter.

References

  1. 1. Gupta N, Ang LC, Noel de Tilly L, Bidaisee L, Yucel YH. Human glaucoma and neural degeneration in intracranial optic nerve, lateral geniculate nucleus, and visual cortex. British J Ophthalmol. 2006;90(6):674-8.
  2. 2. Quigley HA, Nickells RW, Kerrigan LA, Pease ME, Thibault DJ, Zack DJ. Retinal ganglion cell death in experimental glaucoma and after axotomy occurs by apoptosis. Invest Ophthalmol Vis Sci 1995;36(5):774-86.
  3. 3. Quigley HA, Broman AT. The number of people with glaucoma worldwide in 2010 and 2020. British J Ophthalmol. 2006;90(3):262-7.
  4. 4. Fingert JH, Heon E, Liebmann JM, Yamamoto T, Craig JE, Rait J, et al. Analysis of myocilin mutations in 1703 glaucoma patients from five different populations. Human Molecul Genetics. 1999;8(5):899-905.
  5. 5. Libby RT, Gould DB, Anderson MG, John SW. Complex genetics of glaucoma susceptibility. Ann Rev Genomics Human Genetics. 2005;6:15-44.
  6. 6. Mabuchi F, Tang S, Kashiwagi K, Yamagata Z, Iijima H, Tsukahara S. The OPA1 gene polymorphism is associated with normal tension and high tension glaucoma. Am J Ophthalmol. 2007;143(1):125-30.
  7. 7. Ray K, Mukhopadhyay A, Acharya M. Recent advances in molecular genetics of glaucoma. Mol Cel Biochem. 2003;253(1-2):223-31.
  8. 8. Group CN-TGS. Comparison of glaucomatous progression between untreated patients with normal-tension glaucoma and patients with therapeutically reduced intraocular pressures. Collaborative Normal-Tension Glaucoma Study Group. Am J Ophthalmol. 1998;126(4):487-97.
  9. 9. Heijl A, Leske MC, Bengtsson B, Hyman L, Bengtsson B, Hussein M. Reduction of intraocular pressure and glaucoma progression: results from the Early Manifest Glaucoma Trial. Arch Ophthalmol. 2002;120(10):1268-79.
  10. 10. Kass MA, Heuer DK, Higginbotham EJ, Johnson CA, Keltner JL, Miller JP, et al. The Ocular Hypertension Treatment Study: a randomized trial determines that topical ocular hypotensive medication delays or prevents the onset of primary open-angle glaucoma. Arch Ophthalmol. 2002;120(6):701-13.
  11. 11. Baltmr A, Duggan J, Nizari S, Salt TE, Cordeiro MF. Neuroprotection in glaucoma - Is there a future role? Exp Eye Res. 2010;91(5):554-66.
  12. 12. Kroemer G, Galluzzi L, Vandenabeele P, Abrams J, Alnemri ES, Baehrecke EH, et al. Classification of cell death: recommendations of the Nomenclature Committee on Cell Death 2009. Cell Death Differ. 2009;16(1):3-11.
  13. 13. Kerr JF, Wyllie AH, Currie AR. Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. British J Cancer. 1972;26(4):239-57.
  14. 14. Riedl SJ, Shi Y. Molecular mechanisms of caspase regulation during apoptosis. Nature Rev Mol Cell Biol. 2004;5(11):897-907.
  15. 15. Vandenabeele P, Galluzzi L, Vanden Berghe T, Kroemer G. Molecular mechanisms of necroptosis: an ordered cellular explosion. Nature Rev Mol Cell Biol. 2010;11(10):700-14.
  16. 16. Degterev A, Huang Z, Boyce M, Li Y, Jagtap P, Mizushima N, et al. Chemical inhibitor of nonapoptotic cell death with therapeutic potential for ischemic brain injury. Nat Chem Biol. 2005;1(2):112-9.
  17. 17. Chan FK, Shisler J, Bixby JG, Felices M, Zheng L, Appel M, et al. A role for tumor necrosis factor receptor-2 and receptor-interacting protein in programmed necrosis and antiviral responses. J Biol Chem. 2003;278(51):51613-21.
  18. 18. Bredesen DE, Rao RV, Mehlen P. Cell death in the nervous system. Nature. 2006;443(7113):796-802.
  19. 19. Nagata S. Apoptosis by death factor. Cell. 1997;88(3):355-65.
  20. 20. Peter ME, Krammer PH. The CD95(APO-1/Fas) DISC and beyond. Cell Death Different. 2003;10(1):26-35.
  21. 21. Luo X, Budihardjo I, Zou H, Slaughter C, Wang X. Bid, a Bcl2 interacting protein, mediates cytochrome c release from mitochondria in response to activation of cell surface death receptors. Cell. 1998;94(4):481-90.
  22. 22. Li H, Zhu H, Xu CJ, Yuan J. Cleavage of BID by caspase 8 mediates the mitochondrial damage in the Fas pathway of apoptosis. Cell. 1998;94(4):491-501.
  23. 23. Wang X. The expanding role of mitochondria in apoptosis. Gen Develop. 2001;15(22):2922-33.
  24. 24. Li P, Nijhawan D, Budihardjo I, Srinivasula SM, Ahmad M, Alnemri ES, et al. Cytochrome c and dATP-dependent formation of Apaf-1/caspase-9 complex initiates an apoptotic protease cascade. Cell. 1997;91(4):479-89.
  25. 25. Du C, Fang M, Li Y, Li L, Wang X. Smac, a mitochondrial protein that promotes cytochrome c-dependent caspase activation by eliminating IAP inhibition. Cell. 2000;102(1):33-42.
  26. 26. Verhagen AM, Ekert PG, Pakusch M, Silke J, Connolly LM, Reid GE, et al. Identification of DIABLO, a mammalian protein that promotes apoptosis by binding to and antagonizing IAP proteins. Cell. 2000;102(1):43-53.
  27. 27. Shembade N, Ma A, Harhaj EW. Inhibition of NF-kappaB signaling by A20 through disruption of ubiquitin enzyme complexes. Science. 2010;327(5969):1135-9.
  28. 28. Wright A, Reiley WW, Chang M, Jin W, Lee AJ, Zhang M, et al. Regulation of early wave of germ cell apoptosis and spermatogenesis by deubiquitinating enzyme CYLD. Develop Cell. 2007;13(5):705-16.
  29. 29. Micheau O, Tschopp J. Induction of TNF receptor I-mediated apoptosis via two sequential signaling complexes. Cell. 2003;114(2):181-90.
  30. 30. Stanger BZ, Leder P, Lee TH, Kim E, Seed B. RIP: a novel protein containing a death domain that interacts with Fas/APO-1 (CD95) in yeast and causes cell death. Cell. 1995;81(4):513-23.
  31. 31. Lin Y, Devin A, Rodriguez Y, Liu ZG. Cleavage of the death domain kinase RIP by caspase-8 prompts TNF-induced apoptosis. Gen Develop. 1999;13(19):2514-26.
  32. 32. Vercammen D, Beyaert R, Denecker G, Goossens V, Van Loo G, Declercq W, et al. Inhibition of caspases increases the sensitivity of L929 cells to necrosis mediated by tumor necrosis factor. J Exp Med. 1998;187(9):1477-85.
  33. 33. Vercammen D, Brouckaert G, Denecker G, Van de Craen M, Declercq W, Fiers W, et al. Dual signaling of the Fas receptor: initiation of both apoptotic and necrotic cell death pathways. J Expe Med. 1998;188(5):919-30.
  34. 34. Holler N, Zaru R, Micheau O, Thome M, Attinger A, Valitutti S, et al. Fas triggers an alternative, caspase-8-independent cell death pathway using the kinase RIP as effector molecule. Nature Immunol. 2000;1(6):489-95.
  35. 35. Degterev A, Hitomi J, Germscheid M, Ch'en IL, Korkina O, Teng X, et al. Identification of RIP1 kinase as a specific cellular target of necrostatins. Nat Chemical Biol. 2008;4(5):313-21.
  36. 36. Cho YS, Challa S, Moquin D, Genga R, Ray TD, Guildford M, et al. Phosphorylation-driven assembly of the RIP1-RIP3 complex regulates programmed necrosis and virus-induced inflammation. Cell. 2009;137(6):1112-23.
  37. 37. He S, Wang L, Miao L, Wang T, Du F, Zhao L, et al. Receptor interacting protein kinase-3 determines cellular necrotic response to TNF-alpha. Cell. 2009;137(6):1100-11.
  38. 38. Zhang DW, Shao J, Lin J, Zhang N, Lu BJ, Lin SC, et al. RIP3, an energy metabolism regulator that switches TNF-induced cell death from apoptosis to necrosis. Science. 2009;325(5938):332-6.
  39. 39. Kim YS, Morgan MJ, Choksi S, Liu ZG. TNF-induced activation of the Nox1 NADPH oxidase and its role in the induction of necrotic cell death. Mol Cell. 2007;26(5):675-87.
  40. 40. Yazdanpanah B, Wiegmann K, Tchikov V, Krut O, Pongratz C, Schramm M, et al. Riboflavin kinase couples TNF receptor 1 to NADPH oxidase. Nature. 2009;460(7259):1159-63.
  41. 41. Sumimoto H. Structure, regulation and evolution of Nox-family NADPH oxidases that produce reactive oxygen species. FEBS J. 2008;275(13):3249-77.
  42. 42. Yu L, Wan F, Dutta S, Welsh S, Liu Z, Freundt E, et al. Autophagic programmed cell death by selective catalase degradation. Proc Nat Acad Sci USA. 2006;103(13):4952-7.
  43. 43. Sun L, Wang H, Wang Z, He S, Chen S, Liao D, et al. Mixed lineage kinase domain-like protein mediates necrosis signaling downstream of RIP3 kinase. Cell. 2012;148(1-2):213-27.
  44. 44. Wang Z, Jiang H, Chen S, Du F, Wang X. The mitochondrial phosphatase PGAM5 functions at the convergence point of multiple necrotic death pathways. Cell. 2012;148(1-2):228-43.
  45. 45. Kerrigan LA, Zack DJ, Quigley HA, Smith SD, Pease ME. TUNEL-positive ganglion cells in human primary open-angle glaucoma. Arch Ophthalmol. 1997;115(8):1031-5.
  46. 46. Quigley HA. Glaucoma: macrocosm to microcosm the Friedenwald lecture. Invest Ophthalmol Vis Sci. 2005;46(8):2662-70.
  47. 47. Guo L, Moss SE, Alexander RA, Ali RR, Fitzke FW, Cordeiro MF. Retinal ganglion cell apoptosis in glaucoma is related to intraocular pressure and IOP-induced effects on extracellular matrix. Invest Ophthalmol Vis Sci. 2005;46(1):175-82.
  48. 48. Kwon YH, Fingert JH, Kuehn MH, Alward WL. Primary open-angle glaucoma. N Engl J Med. 2009;360(11):1113-24.
  49. 49. Howell GR, Libby RT, Jakobs TC, Smith RS, Phalan FC, Barter JW, et al. Axons of retinal ganglion cells are insulted in the optic nerve early in DBA/2J glaucoma. J Cell Biol. 2007;179(7):1523-37.
  50. 50. Sommer A. Intraocular pressure and glaucoma. Am J Ophthalmol. 1989;107(2):186-8.
  51. 51. Quigley HA. Clinical trials for glaucoma neuroprotection are not impossible. Curr Opin Ophthalmol. 2012;23(2):144-54.
  52. 52. Burgoyne CF, Downs JC. Premise and prediction-how optic nerve head biomechanics underlies the susceptibility and clinical behavior of the aged optic nerve head. J Glaucoma. 2008;17(4):318-28.
  53. 53. Sigal IA, Ethier CR. Biomechanics of the optic nerve head. Exp Eye Res. 2009;88(4):799-807.
  54. 54. Dreyer EB, Zurakowski D, Schumer RA, Podos SM, Lipton SA. Elevated glutamate levels in the vitreous body of humans and monkeys with glaucoma. Arch Ophthalmol. 1996;114(3):299-305.
  55. 55. Guo L, Salt TE, Maass A, Luong V, Moss SE, Fitzke FW, et al. Assessment of neuroprotective effects of glutamate modulation on glaucoma-related retinal ganglion cell apoptosis in vivo. Invest Ophthalmol Vis Sci. 2006;47(2):626-33.
  56. 56. Osborne NN, Ugarte M, Chao M, Chidlow G, Bae JH, Wood JP, et al. Neuroprotection in relation to retinal ischemia and relevance to glaucoma. Surv Ophthalmol. 1999;43 Suppl 1:S102-28.
  57. 57. Salt TE, Cordeiro MF. Glutamate excitotoxicity in glaucoma: throwing the baby out with the bathwater? Eye (Lond). 2006;20(6):730-1.
  58. 58. Kaur C, Foulds WS, Ling EA. Hypoxia-ischemia and retinal ganglion cell damage. Clin Ophthalmol. 2008;2(4):879-89.
  59. 59. Tezel G, Yang X, Luo C, Cai J, Kain AD, Powell DW, et al. Hemoglobin expression and regulation in glaucoma: insights into retinal ganglion cell oxygenation. Invest Ophthalmol Vis Sci. 2010;51(2):907-19.
  60. 60. Costa VP, Arcieri ES, Harris A. Blood pressure and glaucoma. British J Ophthalmol. 2009;93(10):1276-82.
  61. 61. Leske MC. Ocular perfusion pressure and glaucoma: clinical trial and epidemiologic findings. Curr Opin Ophthalmol. 2009;20(2):73-8..
  62. 62. Mittag TW, Danias J, Pohorenec G, Yuan HM, Burakgazi E, Chalmers-Redman R, et al. Retinal damage after 3 to 4 months of elevated intraocular pressure in a rat glaucoma model. Invest Ophthalmol Vis Sci. 2000;41(11):3451-9.
  63. 63. Tatton WG, Chalmers-Redman RM, Sud A, Podos SM, Mittag TW. Maintaining mitochondrial membrane impermeability. an opportunity for new therapy in glaucoma? Surv Ophthalmol. 2001;45 Suppl 3:S277-83.
  64. 64. Tezel G, Yang X. Caspase-independent component of retinal ganglion cell death, in vitro. Invest Ophthalmol Vis Sci. 2004;45(11):4049-59.
  65. 65. Hisatomi T, Nakazawa T, Noda K, Almulki L, Miyahara S, Nakao S, et al. HIV protease inhibitors provide neuroprotection through inhibition of mitochondrial apoptosis in mice. J Clin Invest. 2008;118(6):2025-38.
  66. 66. Lebrun-Julien F, Duplan L, Pernet V, Osswald I, Sapieha P, Bourgeois P, et al. Excitotoxic death of retinal neurons in vivo occurs via a non-cell-autonomous mechanism. J Neurosc. 2009;29(17):5536-45.
  67. 67. Guo L, Salt TE, Luong V, Wood N, Cheung W, Maass A, et al. Targeting amyloid-beta in glaucoma treatment. Proc Nat Acad Sci USA. 2007;104(33):13444-9.
  68. 68. McKinnon SJ, Lehman DM, Kerrigan-Baumrind LA, Merges CA, Pease ME, Kerrigan DF, et al. Caspase activation and amyloid precursor protein cleavage in rat ocular hypertension. Invest Ophthalmol Vis Sci. 2002;43(4):1077-87.
  69. 69. Yoneda S, Hara H, Hirata A, Fukushima M, Inomata Y, Tanihara H. Vitreous fluid levels of beta-amyloid((1-42)) and tau in patients with retinal diseases. Japan J Ophthalmol. 2005;49(2):106-8.
  70. 70. Ko ML, Peng PH, Ma MC, Ritch R, Chen CF. Dynamic changes in reactive oxygen species and antioxidant levels in retinas in experimental glaucoma. Free Rad Biol Med. 2005;39(3):365-73.
  71. 71. Tezel G. Oxidative stress in glaucomatous neurodegeneration: mechanisms and consequences. Progr Ret Eye Res. 2006;25(5):490-513.
  72. 72. Wax MB. The case for autoimmunity in glaucoma. Exp Eye Res. 2011;93(2):187-90.
  73. 73. Cui Q, Harvey AR. At least two mechanisms are involved in the death of retinal ganglion cells following target ablation in neonatal rats. J Neurosci. 1995;15(12):8143-55.
  74. 74. Rudzinski M, Wong TP, Saragovi HU. Changes in retinal expression of neurotrophins and neurotrophin receptors induced by ocular hypertension. J Neurobiol. 2004;58(3):341-54.
  75. 75. Tezel G, Li LY, Patil RV, Wax MB. TNF-alpha and TNF-alpha receptor-1 in the retina of normal and glaucomatous eyes. Invest Ophthalmol Vis Sci. 2001;42(8):1787-94.
  76. 76. Tezel G, Yang X, Luo C, Peng Y, Sun SL, Sun D. Mechanisms of immune system activation in glaucoma: oxidative stress-stimulated antigen presentation by the retina and optic nerve head glia. Invest Ophthalmol Vis Sci. 2007;48(2):705-14.
  77. 77. Danesh-Meyer HV. Neuroprotection in glaucoma: recent and future directions. Curr Opin Ophthalmol. 2011;22(2):78-86.
  78. 78. Danias J, Shen F, Kavalarakis M, Chen B, Goldblum D, Lee K, et al. Characterization of retinal damage in the episcleral vein cauterization rat glaucoma model. Exp Eye Res. 2006;82(2):219-28.
  79. 79. Levkovitch-Verbin H, Quigley HA, Martin KR, Valenta D, Baumrind LA, Pease ME. Translimbal laser photocoagulation to the trabecular meshwork as a model of glaucoma in rats. Invest Ophthalmol Vis Sci. 2002;43(2):402-10.
  80. 80. Johnson EC, Guo Y, Cepurna WO, Morrison JC. Neurotrophin roles in retinal ganglion cell survival: lessons from rat glaucoma models. Exp Eye Res. 2009;88(4):808-15.
  81. 81. Sappington RM, Carlson BJ, Crish SD, Calkins DJ. The microbead occlusion model: a paradigm for induced ocular hypertension in rats and mice. Invest Ophthalmol Vis Sci. 2010;51(1):207-16.
  82. 82. Levkovitch-Verbin H, Quigley HA, Kerrigan-Baumrind LA, D'Anna SA, Kerrigan D, Pease ME. Optic nerve transection in monkeys may result in secondary degeneration of retinal ganglion cells. Invest Ophthalmol Vis Sci. 2001;42(5):975-82.
  83. 83. John SW, Smith RS, Savinova OV, Hawes NL, Chang B, Turnbull D, et al. Essential iris atrophy, pigment dispersion, and glaucoma in DBA/2J mice. Invest Ophthalmol Vis Sci. 1998;39(6):951-62.
  84. 84. Anderson MG, Smith RS, Hawes NL, Zabaleta A, Chang B, Wiggs JL, et al. Mutations in genes encoding melanosomal proteins cause pigmentary glaucoma in DBA/2J mice. Nature Gen. 2002;30(1):81-5.
  85. 85. Cone FE, Gelman SE, Son JL, Pease ME, Quigley HA. Differential susceptibility to experimental glaucoma among 3 mouse strains using bead and viscoelastic injection. Exp Eye Res. 2010;91(3):415-24.
  86. 86. Chrysostomou V, Trounce IA, Crowston JG. Mechanisms of retinal ganglion cell injury in aging and glaucoma. Ophthalm Res. 2010;44(3):173-8.
  87. 87. Faden AI. Neuroprotection and traumatic brain injury: theoretical option or realistic proposition. Curr Opin Neurol. 2002;15(6):707-12.
  88. 88. Tolias CM, Bullock MR. Critical appraisal of neuroprotection trials in head injury: what have we learned? NeuroRx. 2004;1(1):71-9.
  89. 89. Lebrun-Julien F, Di Polo A. Molecular and cell-based approaches for neuroprotection in glaucoma. Optom Vis Sci. 2008;85(6):417-24.
  90. 90. Levin LA. Retinal ganglion cells and neuroprotection for glaucoma. Surv Ophthalmol. 2003;48 Suppl 1:S21-4.
  91. 91. Lipton SA. Possible role for memantine in protecting retinal ganglion cells from glaucomatous damage. Surv Ophthalmol. 2003;48 Suppl 1:S38-46.
  92. 92. Hare WA, WoldeMussie E, Weinreb RN, Ton H, Ruiz G, Wijono M, et al. Efficacy and safety of memantine treatment for reduction of changes associated with experimental glaucoma in monkey, II: Structural measures. Invest Ophthalmol Vis Sci. 2004;45(8):2640-51.
  93. 93. Lagreze WA, Knorle R, Bach M, Feuerstein TJ. Memantine is neuroprotective in a rat model of pressure-induced retinal ischemia. Invest Ophthalmol Vis Sci. 1998;39(6):1063-6.
  94. 94. Sakamoto K, Kawakami T, Shimada M, Yamaguchi A, Kuwagata M, Saito M, et al. Histological protection by cilnidipine, a dual L/N-type Ca(2+) channel blocker, against neurotoxicity induced by ischemia-reperfusion in rat retina. Exp Eye Res. 2009;88(5):974-82.
  95. 95. Fitzgerald M, Payne SC, Bartlett CA, Evill L, Harvey AR, Dunlop SA. Secondary retinal ganglion cell death and the neuroprotective effects of the calcium channel blocker lomerizine. Invest Ophthalmol Vis Sci. 2009;50(11):5456-62.
  96. 96. Saylor M, McLoon LK, Harrison AR, Lee MS. Experimental and clinical evidence for brimonidine as an optic nerve and retinal neuroprotective agent: an evidence-based review. Arch Ophthalmol. 2009;127(4):402-6.
  97. 97. Dong CJ, Guo Y, Agey P, Wheeler L, Hare WA. Alpha2 adrenergic modulation of NMDA receptor function as a major mechanism of RGC protection in experimental glaucoma and retinal excitotoxicity. Invest Ophthalmol Vis Sci. 2008;49(10):4515-22.
  98. 98. Ma K, Xu L, Zhang H, Zhang S, Pu M, Jonas JB. Effect of brimonidine on retinal ganglion cell survival in an optic nerve crush model. Am J Ophthalmol. 2009;147(2):326-31.
  99. 99. Goldenberg-Cohen N, Dadon-Bar-El S, Hasanreisoglu M, Avraham-Lubin BC, Dratviman-Storobinsky O, Cohen Y, et al. Possible neuroprotective effect of brimonidine in a mouse model of ischaemic optic neuropathy. Clin Exp Ophthalmol. 2009;37(7):718-29.
  100. 100. Hernandez M, Urcola JH, Vecino E. Retinal ganglion cell neuroprotection in a rat model of glaucoma following brimonidine, latanoprost or combined treatments. Exp Eye Res. 2008;86(5):798-806.
  101. 101. Saragovi HU, Hamel E, Di Polo A. A neurotrophic rationale for the therapy of neurodegenerative disorders. Curr Alzh Res. 2009;6(5):419-23.
  102. 102. Koriyama Y, Tanii H, Ohno M, Kimura T, Kato S. A novel neuroprotective role of a small peptide from flesh fly, 5-S-GAD in the rat retina in vivo. Brain Res. 2008;1240:196-203.
  103. 103. Cooper NG, Laabich A, Fan W, Wang X. The relationship between neurotrophic factors and CaMKII in the death and survival of retinal ganglion cells. Progr Brain Res. 2008;173:521-40.
  104. 104. Weber AJ, Harman CD, Viswanathan S. Effects of optic nerve injury, glaucoma, and neuroprotection on the survival, structure, and function of ganglion cells in the mammalian retina. J Physiol. 2008;586:4393-400.
  105. 105. Parrilla-Reverter G, Agudo M, Sobrado-Calvo P, Salinas-Navarro M, Villegas-Perez MP, Vidal-Sanz M. Effects of different neurotrophic factors on the survival of retinal ganglion cells after a complete intraorbital nerve crush injury: a quantitative in vivo study. Exp Eye Res. 2009;89(1):32-41.
  106. 106. Pease ME, Zack DJ, Berlinicke C, Bloom K, Cone F, Wang Y, et al. Effect of CNTF on retinal ganglion cell survival in experimental glaucoma. Invest Ophthalmol Vis Sci. 2009;50(5):2194-200.
  107. 107. Fu QL, Hu B, Wu W, Pepinsky RB, Mi S, So KF. Blocking LINGO-1 function promotes retinal ganglion cell survival following ocular hypertension and optic nerve transection. Invest Ophthalmol Vis Sci. 2008;49(3):975-85.
  108. 108. Quigley HA, McKinnon SJ, Zack DJ, Pease ME, Kerrigan-Baumrind LA, Kerrigan DF, et al. Retrograde axonal transport of BDNF in retinal ganglion cells is blocked by acute IOP elevation in rats. Invest Ophthalmol Vis Sci. 2000;41(11):3460-6.
  109. 109. Grozdanic SD, Lazic T, Kuehn MH, Harper MM, Kardon RH, Kwon YH, et al. Exogenous modulation of intrinsic optic nerve neuroprotective activity. Graefe's Arch Clin Exp Ophthalmol. 2010;248(8):1105-16.
  110. 110. Weber AJ, Viswanathan S, Ramanathan C, Harman CD. Combined application of BDNF to the eye and brain enhances ganglion cell survival and function in the cat after optic nerve injury. Invest Ophthalmol Vis Sci. 2010;51(1):327-34.
  111. 111. Park KJ, Grosso CA, Aubert I, Kaplan DR, Miller FD. p75NTR-dependent, myelin-mediated axonal degeneration regulates neural connectivity in the adult brain. Nature Neurosci. 2010;13(5):559-66.
  112. 112. Lebrun-Julien F, Morquette B, Douillette A, Saragovi HU, Di Polo A. Inhibition of p75(NTR) in glia potentiates TrkA-mediated survival of injured retinal ganglion cells. Mol Cel Neurosci. 2009;40(4):410-20.
  113. 113. Bai Y, Xu J, Brahimi F, Zhuo Y, Sarunic MV, Saragovi HU. An agonistic TrkB mAb causes sustained TrkB activation, delays RGC death, and protects the retinal structure in optic nerve axotomy and in glaucoma. Invest Ophthalmol Vis Sci. 2010;51(9):4722-31.
  114. 114. Tezel G. TNF-alpha signaling in glaucomatous neurodegeneration. Progr Brain Res. 2008;173:409-21.
  115. 115. Roh M, Zhang Y, Murakami Y, Thanos A, Lee SC, Vavvas DG, et al. Etanercept, a Widely Used Inhibitor of Tumor Necrosis Factor-alpha (TNF- alpha), Prevents Retinal Ganglion Cell Loss in a Rat Model of Glaucoma. PloS One. 2012;7(7):e40065.
  116. 116. Nakazawa T, Nakazawa C, Matsubara A, Noda K, Hisatomi T, She H, et al. Tumor necrosis factor-alpha mediates oligodendrocyte death and delayed retinal ganglion cell loss in a mouse model of glaucoma. J Neurosci. 2006;26(49):12633-41.
  117. 117. Nucci C, Tartaglione R, Cerulli A, Mancino R, Spano A, Cavaliere F, et al. Retinal damage caused by high intraocular pressure-induced transient ischemia is prevented by coenzyme Q10 in rat. Internat Rev Neurobiol. 2007;82:397-406.
  118. 118. Russo R, Cavaliere F, Rombola L, Gliozzi M, Cerulli A, Nucci C, et al. Rational basis for the development of coenzyme Q10 as a neurotherapeutic agent for retinal protection. Progr Brain Res. 2008;173:575-82.
  119. 119. Munemasa Y, Ahn JH, Kwong JM, Caprioli J, Piri N. Redox proteins thioredoxin 1 and thioredoxin 2 support retinal ganglion cell survival in experimental glaucoma. Gene Ther. 2009;16(1):17-25.
  120. 120. Neufeld AH. Pharmacologic neuroprotection with an inhibitor of nitric oxide synthase for the treatment of glaucoma. Brain Res Bul. 2004;62(6):455-9.
  121. 121. Wax MB, Tezel G. Immunoregulation of retinal ganglion cell fate in glaucoma. Exp Eye Res. 2009;88(4):825-30.
  122. 122. Bakalash S, Kessler A, Mizrahi T, Nussenblatt R, Schwartz M. Antigenic specificity of immunoprotective therapeutic vaccination for glaucoma. Invest Ophthalmol Vis Sci. 2003;44(8):3374-81.
  123. 123. Huang W, Fileta JB, Dobberfuhl A, Filippopolous T, Guo Y, Kwon G, et al. Calcineurin cleavage is triggered by elevated intraocular pressure, and calcineurin inhibition blocks retinal ganglion cell death in experimental glaucoma. Proc Nat Acad Sci USA. 2005;102(34):12242-7.
  124. 124. Frank T, Schlachetzki JC, Goricke B, Meuer K, Rohde G, Dietz GP, et al. Both systemic and local application of granulocyte-colony stimulating factor (G-CSF) is neuroprotective after retinal ganglion cell axotomy. BMC Neurosci. 2009;10:49.
  125. 125. Bull ND, Irvine KA, Franklin RJ, Martin KR. Transplanted oligodendrocyte precursor cells reduce neurodegeneration in a model of glaucoma. Invest Ophthalmol Vis Sci. 2009;50(9):4244-53.
  126. 126. Johnson TV, Bull ND, Hunt DP, Marina N, Tomarev SI, Martin KR. Neuroprotective effects of intravitreal mesenchymal stem cell transplantation in experimental glaucoma. Invest Ophthalmol Vis Sci. 2010;51(4):2051-9.
  127. 127. Levkovitch-Verbin H, Kalev-Landoy M, Habot-Wilner Z, Melamed S. Minocycline delays death of retinal ganglion cells in experimental glaucoma and after optic nerve transection. Arch Ophthalmol. 2006;124(4):520-6.
  128. 128. Levkovitch-Verbin H, Vander S, Melamed S. Rasagiline-induced delay of retinal ganglion cell death in experimental glaucoma in rats. J Glaucoma. 2011;20(5):273-7.
  129. 129. Zhong L, Bradley J, Schubert W, Ahmed E, Adamis AP, Shima DT, et al. Erythropoietin promotes survival of retinal ganglion cells in DBA/2J glaucoma mice. Invest Ophthalmol Vis Sci. 2007;48(3):1212-8.
  130. 130. Parisi V, Coppola G, Ziccardi L, Gallinaro G, Falsini B. Cytidine-5'-diphosphocholine (Citicoline): a pilot study in patients with non-arteritic ischaemic optic neuropathy. Eur J Neurol. 2008;15(5):465-74.
  131. 131. Danesh-Meyer HV, Levin LA. Neuroprotection: extrapolating from neurologic diseases to the eye. Am J Ophthalmol. 2009;148(2):186-91 e2.
  132. 132. Sena DF, Ramchand K, Lindsley K. Neuroprotection for treatment of glaucoma in adults. Cochrane Database Syst Rev. 2010(2):CD006539.
  133. 133. Inc. A. Press releases on Memantine Trials. http://wwwallergancom/newsroom/indexhtm.
  134. 134. Newman NJ, Biousse V, David R, Bhatti MT, Hamilton SR, Farris BK, et al. Prophylaxis for second eye involvement in leber hereditary optic neuropathy: an open-labeled, nonrandomized multicenter trial of topical brimonidine purite. Am J Ophthalmol. 2005;140(3):407-15.
  135. 135. Wilhelm B, Ludtke H, Wilhelm H. Efficacy and tolerability of 0.2% brimonidine tartrate for the treatment of acute non-arteritic anterior ischemic optic neuropathy (NAION): a 3-month, double-masked, randomised, placebo-controlled trial. Graefe's Archive Clin Exp Ophthalmol. 2006;244(5):551-8.
  136. 136. Krupin T, Liebmann JM, Greenfield DS, Rosenberg LF, Ritch R, Yang JW. The Low-pressure Glaucoma Treatment Study (LoGTS) study design and baseline characteristics of enrolled patients. Ophthalmology. 2005;112(3):376-85.
  137. 137. Krupin T, Liebmann JM, Greenfield DS, Ritch R, Gardiner S. A randomized trial of brimonidine versus timolol in preserving visual function: results from the Low-Pressure Glaucoma Treatment Study. Am J Ophthalmol. 2011;151(4):671-81.
  138. 138. Schwid SR, Cutter GR. Futility studies: spending a little to save a lot. Neurology. 2006;66(5):626-7.
  139. 139. Tilley BC, Palesch YY, Kieburtz K, Ravina B, Huang P, Elm JJ, et al. Optimizing the ongoing search for new treatments for Parkinson disease: using futility designs. Neurology. 2006;66(5):628-33.
  140. 140. Leske MC, Heijl A, Hyman L, Bengtsson B, Dong L, Yang Z. Predictors of long-term progression in the early manifest glaucoma trial. Ophthalmology. 2007;114(11):1965-72.
  141. 141. Nordstrom BL, Friedman DS, Mozaffari E, Quigley HA, Walker AM. Persistence and adherence with topical glaucoma therapy. Am J Ophthalmol. 2005;140(4):598-606.

Written By

Sotiria Palioura and Demetrios G. Vavvas

Submitted: 11 April 2012 Published: 17 April 2013