Вы находитесь на странице: 1из 5

REVIEW ARTICLE

Canadian Journal of Ophthalmology Lecture: Translational


research advances in glaucoma neuroprotection
James C. Tsai, MD
ABSTRACT RE
SUME

Given that glaucoma is a disease of the central nervous system, there must be greater emphasis and focus on developing
intraocular pressureindependent therapies. In addition, scientific research investigating the role of sustained drug delivery is critical
for advancing the field. Exciting innovation will require close collaboration and full integration of clinical and basic research efforts.
Thus, translational research advances in neuroprotection may lead to future development of truly revolutionary therapies for
glaucoma (e.g., neuroregeneration).

Comme le glaucome est une maladie du systeme  nerveux central, il faut mettre plus demphase et de concentration sur le
developpement de therapies independantes de la PIO. En outre, la recherche scientifique dinvestigations sur le role
de la livraison
soutenue des medicaments est critique pout faire progresser ce secteur. Les innovations passionnantes demanderont une etroite
 des efforts cliniques et fondamentaux. Ainsi, les progres
collaboration et une integration entiere  de la recherche translationnelle de la
neuro-protection peut mener a un developpement de therapies vraiment revolutionnaires du glaucome (e,g, neuro-regeneration).

According to the World Health Organization, glaucoma visual field testing. Recent advances in vision function
is the second leading cause of global blindness; by 2010, assessment include the multifocal and isolated check
nearly 80 million persons worldwide will be afflicted with visual evoked potential technologies that may provide
the disease.1 Because of its often asymptomatic nature and objective visual function parameters and complement
gradual loss of vision, primary open-angle glaucoma standard automated perimetry.6,7
(POAG) has been described as a thief in the night.2 Currently accepted therapeutic strategies for glaucoma,
Treatment does not always slow disease progression in whether by medication, laser procedure, or incisional
patients with open-angle glaucoma. In the Early Manifest surgery, aim to lower the IOP below a presumed thresh-
Glaucoma Treatment (EMGT) study, 45% of patients old level.810 Given the nontissue-specific targeted nature
randomized to initial treatment still had progression of of these therapies (i.e., they only lower the IOP risk
their visual fields at 5 years of follow-up.3 factor), the marked variability in patient outcomes is not
Intraocular pressure (IOP) is currently recognized as at all surprising (Fig. 1). Furthermore, multiple additional
the most significant contributing factor in the initial disease risk factors must be taken into account; these
development, as well as progression, of open-angle include history of previous IOP levels (with or without
glaucoma. Glaucoma can be best viewed as an IOP- treatment), central corneal thickness values, patient age
associated optic neuropathy, with glaucoma clinical trials and life expectancy, family history, vascular factors (e.g.,
and observational studies strongly supporting the need to low perfusion pressure), and other comorbidities (e.g.,
reduce IOP substantially and to maintain these pressures cardiovascular disease).3,811
in patients with advanced disease.4 Because the lateral As a chronic neurodegenerative disease, POAG is
geniculate nucleus (LGN) of the midbrain is the major characterized by primary optic nerve head injury and
synaptic target for retinal ganglion cells (RGCs), glau- subsequent loss of RGCs.12 Although initial experimental
coma must be viewed as a central nervous system (CNS) primate studies showed a selective loss of magnocellular
disease.5 RGC bodies and axons, recent contrast sensitivity testing of
A clinicians decision to prescribe IOP-lowering ther- patients with glaucoma suggests nonselective impairment
apy results often from estimation of IOP risk in the of the low-spatial-frequency components of both magno-
context of the individual patients existing optic nerve cellular and parvocellular pathways, presumably mediated
damage and/or visual field deficits, taking into account by cells with larger receptive fields.13 In addition to RGC
the slowly progressive course of the disease. In addition, death, there is atrophy and loss of target neurons in the
follow-up of patients with glaucoma may be challenging LGN of the brain. Studies using experimental primate
given the subjective and fluctuating nature of current models of glaucoma have shown reduced dendrite

From the Department of Ophthalmology and Visual Science, Yale Can J Ophthalmol 2013;48:141145
University School of Medicine, New Haven, Conn. 0008-4182/13/$-see front matter & 2013 Canadian Ophthalmological
Society. Published by Elsevier Inc. All rights reserved.
Originally received Oct. 16, 2012. Final revision Feb. 11, 2013. http://dx.doi.org/10.1016/j.jcjo.2013.02.003
Accepted Feb. 12, 2013

Correspondence to James C. Tsai, Yale Eye Center, 40 Temple Street,


Suite 1B, New Haven, CT 06510; james.tsai@yale.edu

CAN J OPHTHALMOL VOL. 48, NO. 3, JUNE 2013 141


Neuroprotection in glaucomaTsai

involving in vivo imaging and identification of dying RGCs


with fluorescent biomarkers are facilitating the clinical
evaluation of potential neuroprotection-focused strategies.17

Investigational agents
Medication: Surgery:
-adrenergic agonists Laser trabeculoplasty
Drugs/Biotechnology. It is important to recognize that
Beta-blockers IOP Laser iridotomy lowering IOP itself is neuroprotective. However, the devel-
CAIs (topical & oral) Trabeculectomy
Prostaglandins Control Glaucoma implants
opment of non-IOPdependent neuroprotective therapies
Miotic agents Cyclodestruction would be an attractive alternative (Table 1). A recent
Combination agents Trabecular-based
Schlemms canal prospective clinical trial randomizing initial topical therapy
Suprachoroidal with either brimonidine or timolol in patients with normal
tension glaucoma demonstrated a neuroprotection benefit
of brimonidine.18 Although memantine, a low-affinity,
open-channel blocker of the N-methyl-D-aspartatetype
glutamate receptor, was shown to be neuroprotective in
Fig. 1 Current glaucoma treatment. CAI: Carbonic Anhy- an experimental glaucoma model in primates,19 a recently
drase Inhibitors (Topical & Oral).
completed Phase III trial showed no beneficial effect in
patients with glaucoma treated with oral memantine as well
complexity by 47% and 41% in magnocellular layer 1 and as conventional IOP-lowering therapies.20
parvocellular layer 6, respectively, in experimental animals Neurotrophic factors show promise in retarding pro-
compared with control animals.14 The first clinicopatho- gression of neurodegenerative diseases. Current preclinical
logic case of human glaucoma also reported neural and clinical studies are ongoing involving factors such as
degeneration of the CNS involving the intracranial optic ciliary neurotrophic factor (CNTF), brain-derived neuro-
nerve, LGN, and visual cortex.15 trophic factor (BDNF), glial-derived neurotropic factor
(GDNF), and pigment epithelium-derived factor (PEDF).21
Moreover, inhibition of the process of reactive gliosis, a
NEUROPROTECTION hallmark of many neurodegenerative conditions including
glaucoma, has been shown to prevent apoptotic death of
Although current glaucoma treatment is focused on retinal neurons and to provide substantial neuroprotection
lowering IOP, future therapeutic strategies may involve in mice.22 In humans, a Phase I clinical safety trial described
disease modification/lifestyle intervention and develop- the successful delivery of human CNTF via cells transfected
ment of non-IOPdependent therapies to prevent against with the human CNTF gene and sequestered within
optic nerve head damage, RGC death, or both.4,10,12 The surgically implanted capsules, thereby suggesting its safety
desired aim would be to intervene at an early stage of the and possible future applicability in patients with glaucoma.23
disease to halt or slow down the neurodegenerative In addition to their known ocular hypotensive effects,
process. Thus, any treatment that eliminates or attenuates, inhibitors of both Rho kinase (ROCK) and Rho GTPase
or both, Z1 of these disease risk factors, thus delaying or have been shown to enhance ocular blood flow, RGC
halting RGC degeneration and reducing the rate of survival, and axon regeneration.24 Application of a cell-
disease progression, can be legitimately classified as a permeable Rho antagonist (C3-07) to either neuronal cell
neuroprotective therapeutic modality.12 Implicated causes bodies or distal axons promotes axonal growth on myelin-
of RGC death include ischemia, reactive oxygen species, associated glycoprotein substrates.24
excitotoxicity, defective axonal transport, trophic factor with- In vivo injection of the C3-07 Rho antagonist also
drawal, and loss of RGC electrical activity (involving specific promoted regeneration of RGC axons in the optic nerve
molecular pathways).12,16 In addition, technologic advances of rats after microcrush lesion.25 Therefore, these animal

Table 1Selective list of investigational agents for neuroprotection in glaucoman


Drug/Therapy Mechanism Approval indication Outcome in humans
Brimonidine a2 receptor Approved for IOP Positive Phase IV trial18
Memantine NMDA receptor None Failed Phase III trials20
CNTF Neurotrophic factor None Completed Phase I trial23
ROCK inhibitors Rho kinase antagonism None Ongoing Phase II/III trials
Erythropoietin HIF-mediated None No human trials
TNF-a blockers TNF-a inhibition None No human trials
Cop-1 T-cellbased vaccination None No human trials

IOP, intraocular pressure; NMDA, N-methyl-D-aspartate; CNTF, ciliary neurotrophic factor; ROCK, Rho kinase; HIF, hypoxia-inducible factor; TNF, tumor necrosis factor; Cop-1, copolymer
glatiramer acetate.
n
Currently, none is approved by U.S. Food and Drug Administration for neuroprotection indication.

142 CAN J OPHTHALMOL VOL. 48, NO. 3, JUNE 2013


Neuroprotection in glaucomaTsai

studies have stimulated interest in the results of upcoming and inhibition of downstream microglial activation.35
Phase II/III clinical trials of ROCK inhibitors in humans. Possibilities include blocking antibodies that interfere
Erythropoietin (EPO), a naturally occurring cytokine with TNF-a, soluble receptor(s), and a TNF-aconvert-
used to treat anemia by inhibiting apoptosis in erythro- ing enzyme inhibitor(s). Infliximab, a TNF-a receptor
cyte progenitors, has a neuroprotective effect in animal blocker, has been used to successfully control ocular
models of glaucoma.26 The EPO and EPO-R systems are inflammatory disease refractory to traditional immuno-
thought to convey endogenous feedback loop through modulatory therapy with a low rate of adverse effects.36
activation via hypoxia-inducible factor. Intravitreal injec- In the near future, T-cellbased vaccination for mor-
tions of EPO have been shown to rescue RGCs and phologic and functional neuroprotection will likely become
prevent caspase-3 activation in axotomized rats, as well as a reality.37 This therapeutic option has been shown to be
retard against RGC loss in a rat model of ocular hyper- effective in retarding RGC cell loss in a rat model of
tension.27,28 Dose toxicity studies of intravitreal injections glaucoma. In experimental animals with chronically ele-
of recombinant EPO in rats were shown to be safe based vated IOP, vaccination with the synthetic copolymer
on electroretinography and histopathology studies.29 glatiramer acetate (Cop-1) was shown to be protective
Additional safety studies of intravitreal recombinant against IOP-induced loss of RGCs by eliciting a systemic
human EPO were conducted in rabbits using electro- T-cellmediated response capable of cross-reacting with
retinography and fluorescein angiography.30 self-antigens in the eye.38 The antigen-specific autoimmune
Gene therapy/stem cell therapy. Another approach in the T cells and/or their cytokines may tailor the microglial
prevention of RGC loss is gene therapy to reprogram cells to phenotype to facilitate glutamate clearance.39 However, it
produce antiapoptotic neurotrophic proteins. As noted ear- is important to note that any benefits of the T-cell
lier, a recent human Phase I safety trial demonstrated the mediated immune response should be balanced against
successful delivery of CNTF by cells transfected with the the real risks of inducing autoimmune disease.
human CNTF gene.23 Gene therapy has shown promise in
promoting RGC survival in experimental models of optic
nerve injury using adeno-associated virus (AAV), adenoviral, NEUROREGENERATION
and lentiviral methods.31 For example, rat RGCs have been
successfully transfected with the gene for the prosurvival Growth of injured axons in the adult mammalian CNS
protein Bcl-XL using both the AAV and HIV-Tatderived is quite limited after injury. Thus, one of the ultimate
fusion proteins.32 An AAV vector capable of efficient trans- goals for glaucoma therapy would likely be neuroregen-
fection of rat RGCs has been developed to deliver BDNF.32 eration, the process of restoring damaged axons and/or
In this experimental study, animals transfected with the reversing RGC death in hopes of functional recovery of
AAV-BDNF gene were more resistant to RGC death in the lost vision. Methods are currently being developed to
hypertensive model of glaucoma. A recent study reported the deliver stem cells to replace RGCs and their axons,
effectiveness of stem cellbased delivery of the BDNF gene thereby reestablishing functional vision.40 In addition,
to rat retina using rat bone marrow mesenchymal stem cells; novel approaches for neuroenhancement of surviving
at 4 weeks after transplantation, BDNF mRNA and protein RGCs (e.g., electrical stimulation) have been studied.12
expression were shown to be increased.33 Advances in intravitreal drug delivery of neuroprotective
Immune system modulation. Involvement of the immune agents (e.g., embedded poly(lactic-co-glycolic acid) nano-
system in modulating RGC survival may occur via oxida- spheres) in a rat optic nerve crush model may also yield
tive stressstimulated antigen presentation by both retina novel treatment strategies.41
and optic nerve head glial cells.34 Tumor necrosis factor-a One therapeutic strategy for neuroregeneration may
(TNF-a) has been shown to cause a cascade of events that involve modulation of the Nogo-66 receptor (NgR)
leads to loss of RGCs. In a laser irradiation-induced murine pathway. Three myelin proteins, Nogo, myelin-
model of ocular hypertension, increased levels of TNF-a associated glycoprotein (MAG), and oligodendrocyte
led to microglial activation, loss of oligodendrocytes in the myelin glycoprotein (OMgp), bind to NgR and inhibit
optic nerve, and loss of RGCs.35 This same sequence of CNS axonal growth.42 Administration of the soluble
biological events was also observed after injection of TNF-a function-blocking NgR ectodomain has been shown to
in a control group of mice with normal IOP levels. cause axonal sprouting in spinal-injured rats with corre-
Conversely, no appreciable loss of RGCs was observed lation to improved spinal cord electrical conduction and
(above baseline control levels) when animals with elevated improved locomotion.42 In addition, soluble LINGO-1
IOP were treated with TNF-a inhibitors.35 Similar preser- (LINGO-1-Fc), which acts as an antagonist of these
vation of RGCs was observed in knockout mice with axonal outgrowth inhibition pathways by blocking
elevated IOP that were unable to produce TNF-a.35 LINGO-1 binding to NgR1, significantly improves func-
Based on the earlier noted TNF-a research, future tional recovery and promotes axonal sprouting after spinal
treatments for glaucoma may involve modulation of this cord injury in rats.43 Preliminary research in our labo-
pathway including direct blockage of TNF-a function ratory suggests that intravitreal injections of an anti-Nogo

CAN J OPHTHALMOL VOL. 48, NO. 3, JUNE 2013 143


Neuroprotection in glaucomaTsai

receptor blocking protein in rats with episcleral venous 4. Lichter PR. Glaucoma clinical trials and what they mean for our
patients. Am J Ophthalmol. 2003;136:136-45.
cautery-induced ocular hypertension may confer neuro- 5. Gupta N, Greenberg G, de Tilly LN, et al. Atrophy of the lateral
protective and neuroregenerative effects (Association for geniculate nucleus in human glaucoma detected by magnetic
Research in Vision and Ophthalmology (ARVO) 2010 resonance imaging. Br J Ophthalmol. 2009;93:56-60.
6. Fortune B, Demirel S, Zhang X, et al. Comparing multifocal VEP
Annual Meeting, Ft. Lauderdale, FL). and standard automated perimetry in high-risk ocular hypertension
and early glaucoma. Invest Ophthalmol Vis Sci. 2007;48:1173-80.
7. Zemon V, Tsai JC, Forbes M, et al. Novel electrophysiological
CONCLUSION instrument for rapid and objective assessment of magnocellular
deficits associated with glaucoma. Doc Ophthalmol. 2008;117:233-43.
8. American Academy of Ophthalmology Glaucoma Preferred Practice
Recent discoveries of the molecular mechanisms under- Pattern Panel: Primary Open Angle Glaucoma Preferred Practice
lying POAG have provided novel insights into its com- Pattern. San Francisco, 2005.
plex pathogenesis. This enhanced understanding offers 9. Tsai JC, Forbes M. Medical management of glaucoma.3rd ed.
Caddo, Okla: Professional Communications; 2009.
tremendous opportunities for the development of inno- 10. Tsai JC. Influencing ocular blood flow in glaucoma patients: the
vative therapeutic strategies. Future approaches including cardiovascular system and healthy lifestyle choices. Can J Ophthal-
anti-Nogo receptor agents, small interfering RNA-based mol. 2008;43:347-50.
11. Simmons ST, Cioffi GA, Gross RL, et al. Glaucoma. American
caspase inhibitors, and neurotrophic factor replacement Academy of Ophthalmology: Basic and Clinical Science Course. Section
therapy should be explored.12 10. American Academy of Ophthalmology: San Francisco, Calif;
The challenges of translating advances from the basic 2007.
12. Chang EE, Goldberg JL. Glaucoma 2.0: neuroprotection, neuro-
science laboratory into real and proven benefits in the regeneration, neuroenhancement. Ophthalmology. 2012;119:979-86.
clinical care of patients with glaucoma cannot be under- 13. McKendrick AM, Sampson GP, Walland MJ, Badcock DR.
stated. These challenges include the lengthy and slow Contrast sensitivity changes due to glaucoma and normal aging:
low-spatial-frequency losses in both magnocellular and parvocellular
course of the disease with its variable rate of progression; pathways. Invest Ophthalmol Vis Sci. 2007;48:2115-22.
the substantial variability and reliability of its main 14. Gupta N, Ly T, Zhang Q, et al. Chronic ocular hypertension
outcome measure, the visual field test; and the need for induces dendrite pathology in the lateral geniculate pathology in the
lateral geniculate nucleus of the brain. Exp Eye Res. 2007;84:176-84.
minimal treatment side-effects/complications given the 15. Gupta N, Ang LC, Noel de Tilly L, et al. Human glaucoma and
diseases often asymptomatic nature. Furthermore, inher- neural degeneration in intracranial optic nerve, lateral geniculate
ent challenges in formulating suitable pharmacologic/ nucleus, and visual cortex. Br J Ophthalmol. 2006;90:674-8.
16. Goldberg JL. Role of electrical activity in promoting neural repair.
biotherapeutic dosages and delivery regimens in humans Neurosci Lett. 2012;519:134-7.
include the following: differences in anatomy and phys- 17. Cordeiro MF, Guo L, Luong V, et al. Real-time imaging of single
iology between animals and humans, marked differences cell apoptosis in retinal neurodegeneration. Proc Natl Acad Sci U S A.
2004;101:13352-6.
in duration between animal studies and human glaucoma 18. Krupin T, Liebmann JM, Greenfield DS, et al. Low-Pressure
(e.g., weeks vs years), and inadequate in vivo laboratory Glaucoma Study Group. A randomized trial of brimonidine versus
models of glaucoma (i.e., majority of models use elevated timolol in preserving visual function: results from the Low-Pressure
Glaucoma Treatment Study. Am J Ophthalmol. 2011;151:671-81.
IOP in normal, young animals). Although there are 19. Yucel YH, Gupta N, Zhang Q, Mizisin AP, Kalichman MW,
substantive barriers, clinical trials for glaucoma neuro- Weinreb RN. Memantine protects neurons from shrinkage in the
protection are not impossible44; I also believe that some lateral geniculate nucleus in experimental glaucoma. Arch Ophthal-
mol. 2006;124:217-25.
will prove successful in the near future. 20. Allergan, Inc. Report on Fourth Quarter Operating Results:
Memantine Update, January 30, 2008.
21. Thanos C, Emerich D. Delivery of neurotrophic factors and
therapeutic proteins for retinal diseases. Expert Opin Biol Ther.
Disclosure: The author has no proprietary or commercial interest 2005;5:1443-52.
in any materials discussed in this article. Over the past 2 years, 22. Ganesh BS, Chintala SK. Inhibition of reactive gliosis attenuates
J.C.T. has received research funding, travel funds, and lecture and excitotoxicity-mediated death of retinal ganglion cells. PLoS One.
consulting honoraria from the National Eye Institute, Research to 2011;6:e18305.
Prevent Blindness, Alcon, Allergan, Bausch and Lomb, Inspire, 23. Sieving PA, Caruso RC, Tao W, et al. Ciliary neurotrophic factor
Merck, Pfizer, QLT, Quark, Rhodes, and Sucampo. (CNTF) for human retinal degeneration: phase I trial of CNTF
delivered by encapsulated cell intraocular implants. Proc Natl Acad
Sci U S A. 2006;103:3896-901.
Support: Supported in part by endowment funds from the Robert 24. Rao VP, Epstein DL. Rho GTPase/Rho kinase inhibition as a novel
R. Young Professorship at Yale University School of Medicine. target for the treatment of glaucoma. BioDrugs. 2007;21:167-77.
25. Bertrand J, Winton MJ, Rodriguez-Hernandez N, et al. Application
of Rho antagonist to neuronal cell bodies promotes neurite growth
REFERENCES in compartmented cultures and regeneration of retinal ganglion cell
axons in the optic nerve of adult rats. J Neurosci. 2005;25:1113-21.
1. Quigley HA, Broman AT. The number of people with glaucoma 26. Tsai JC, Song BJ, Wu L, Forbes M. Erythropoietin: a candidate
worldwide in 2010 and 2020. Br J Ophthalmol. 2006;90:262-7. neuroprotective agent in the treatment of glaucoma. J Glaucoma.
2. Bellows JG, Bellows RT. Glaucoma: a thief in the night. Compr 2007;16:567-71.
Ther. 1977;3:3. 27. Weishaupt J, Rohde G, Polking E, et al. Effect of erythropoietin
3. Leske MC, Heijl A, Bengtsson B, et al. Early Manifest Glaucoma axotomy-induced apoptosis in rat retinal ganglion cells. Invest
Trial Group. Factors for glaucoma progression and the effect of Ophthalmol Vis Sci. 2004;45:1514-22.
treatment: the early manifest glaucoma trial. Arch Ophthalmol. 28. Tsai JC, Wu L, Worgul B, Forbes M, Cai J. Intravitreal admin-
2003;121:48-56. istration of erythropoietin and preservation of retinal ganglion cells

144 CAN J OPHTHALMOL VOL. 48, NO. 3, JUNE 2013


Neuroprotection in glaucomaTsai

in an experimental rat model of glaucoma. Curr Eye Res. 37. Schwartz M. Neurodegeneration and neuroprotection in glaucoma:
2005;30:1025-31. development of a therapeutic neuroprotective vaccine: the Frieden-
29. Tsai JC. Safety of intravitreally administered recombinant eryth- wald lecture. Invest Ophthalmol Vis Sci. 2003;44:3374-81.
ropoietin (an AOS thesis). Trans Am Ophthalmol Soc. 2008;106: 38. Bakalash S, Shlomo GB, Aloni E, et al. T-cell-based vaccination for
459-72. morphological and functional neuroprotection in a rat model of
30. Song BJ, Cai H, Tsai JC, et al. Intravitreal recombinant human chronically elevated intraocular pressure. J Mol Med. 2005;83:
erythropoietin: a safety study in rabbits. Curr Eye Res. 2008;33: 904-16.
750-60. 39. Shaked I, Tchoresh D, Gersner R, et al. Protective autoimmunity:
31. Wilson AM, Di Polo A. Gene therapy for retinal ganglion cell interferon-gamma enables microglia to remove glutamate without
neuroprotection in glaucoma. Gene Ther. 2012;19:127-36. evoking inflammatory mediators. J Neurochem. 2005;92:997-1009.
32. Diem R, Taheri N, Dietz GP, et al. HIV-Tat-mediated Bcl-XL 40. Robinson R, Viviano SR, Criscione JM, et al. Nanospheres
delivery protects retinal ganglion cells during experimental auto- delivering the EGFR TKI AG1478 promote optic nerve regener-
immune optic neuritis. Neurobiol Dis. 2005;20:218-26. ation: the role of size for intraocular drug delivery. ACS Nano.
33. Park HY, Kim JH, Sun Kim H, Park CK. Stem cell-based delivery 2011;5:4392-400.
41. Li S, Liu BP, Budel S. Blockade of Nogo-66, myelin-associated
of brain-derived neurotrophic factor gene in the rat retina. Brain
glycoprotein, and oligodendrocyte myelin glycoprotein by soluble
Res. 2012;1469:10-23.
Nogo-66 receptor promotes axonal sprouting and recovery after
34. Tezel G, Yang X, Luo C, et al. Mechanisms of immune system
spinal injury. J Neurosci. 2004;24:10511-20.
activation in glaucoma: oxidative stress-stimulated antigen presen- 42. Ji B, Li M, Wu WT, et al. LINGO-1 antagonist promotes
tation by the retina and optic nerve head glia. Invest Ophthalmol Vis functional recovery and axonal sprouting after spinal cord injury.
Sci. 2007;48:705-14. Mol Cell Neurosci. 2006;33:311-20.
35. Nakazawa T, Nakazawa C, Matsubara A, et al. Tumor necrosis factor- 43. Steinhart MR, Cone FE, Nguyen C, et al. Mice with an induced
alpha mediates oligodendrocyte death and delayed ganglion cell loss in mutation in collagen 8A2 develop larger eyes and are resistant to
a mouse model of glaucoma. J Neurosci. 2006;26:12633-41. retinal ganglion cell damage in an experimental glaucoma model.
36. Sobrin L, Kim EC, Christen W, et al. Infliximab therapy for the Mol Vis. 2012;18:1093-106.
treatment of refractory ocular inflammatory disease. Arch Ophthal- 44. Quigley HA. Clinical trials for glaucoma neuroprotection are not
mol. 2007;125:895-900. impossible. Curr Opin Ophthalmol. 2012;23:144-54.

CAN J OPHTHALMOL VOL. 48, NO. 3, JUNE 2013 145

Вам также может понравиться