Sunteți pe pagina 1din 11

Recent Patents on Inflammation & Allergy Drug Discovery 2009, 3, 221-231 221

Corneal Neovascularization: Molecular Events and Therapeutic Options


Yadollah Shakiba*,1,2, Kamran Mansouri2, Delnia Arshadi2 and Nima Rezaei3

1
Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran, 2Medical
Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran, 3Growth and Development
Research Center, Pediatrics Center of Excellent, Children's Medical Center, Tehran University of Medical Sciences,
Tehran, Iran

Received: April 2, 2009; Accepted: June 3, 2009; Revised: August 20, 2009
Abstract: Corneal neovascularization (NV) is a significant complication of numerous infectious and non-infectious ocular
surface disorders. Presence of newly formed blood vessels in the cornea can compromise clarity and therefore vision.
Various growth factors and proteinases seem to be involved in the corneal NV. During corneal injury, angiogenic factors
are released from corneal epithelial and stromal cells as well as infiltrating immune cells like macrophages. In fact, the
balance between angiogenic and anti-angiogenic factors is shifted towards angiogenic molecules in the corneal NV.
Numerous investigations support this idea that vascular endothelial growth factor (VEGF) plays a pivotal role in corneal
NV by inducing endothelial cells proliferation, migration, and tubulogenesis. There is a growing body of evidence that
corneal NV can be reduced by using anti-VEGF agents. This article reviews the most known molecular events in corneal
NV and also some of the recent patents relevant to the field. Understanding the role of growth factors, proteinases and
inflammatory cytokines in corneal NV can help the investigators to design therapeutic options for controlling this
debilitating condition.
Keywords: Cornea, neovascularization, VEGF, MMP, inflammation.

INTRODUCTION occurs when the balance between angiogenic and anti-


angiogenic factors is tilted towards angiogenic molecules.
Cornea
This phenomenon occurs in a variety of pathological con-
The cornea is the transparent avascular tissue of the eye, ditions, such as hypoxia, chemical burns, ischemia, inflam-
which its transparency is essential for optimal clarity and mation, infection and trauma [3]. Presence of new blood
vision. The cornea is a dynamic structure that interacts with vessels in previously avascular cornea disturbs light passing
its surrounding tissues and plasma through tear fluid, and influences visual acuity and clarity negatively. Newly
aqueous humor and circulating immune cells. These inter- formed vessels are not completely functional. These vessels
actions play an important role in the maintaining of its have no basement membrane and pericytes; therefore, they
normal structure and functions. Moreover, the cornea is an are very fragile and susceptible to hemorrhage. The leakage
immune-privileged site; immune privilege is a dynamic of plasma components leads to the corneal edema. In
phenomenon in which the immune response to antigens is addition, these compounds can be deposited in the cornea
absent in order to protect this highly organized structure of and impair its transparency permanently [4]. To date, many
the eye. The lack of blood and lymphatic vessels in a normal studies have been conducted for clarifying mechanisms
cornea prevents entering innate and adaptive immune system underlying corneal NV and finding new therapeutic options
cells and behaves as a mechanical barrier. The corneal for controlling the formation of new blood vessels and
avascularity is considered as corneal angiogenic privilege maintaining corneal avascularity. In this article, we review
[1]. Recent investigations indicate a close relationship in the the recent findings on the molecular mechanisms of corneal
molecular mechanisms maintaining corneal angiogenic and NV, focusing on the pivotal role of inflammation and
immune privilege. The cornea is supplied with oxygen and therapeutic potential of anti-angiogenic drugs.
nutrients by pericorneal plexus (limbal plexus), tear and
anterior chamber aqueous humor. Limbal plexus of the EPIDEMIOLOGY AND ETIOLOGY OF CORNEAL
cornea arises from the ciliary arteries, which are branches of NEOVASCULARIZATION
the ophthalmic artery that divide and end in the pericorneal
plexus in the limbus area [2]. Corneal NV is a main cause of loss of vision and
In physiological conditions, the corneal avascularity blindness in a wide range of corneal diseases worldwide. It is
requires low levels of angiogenic factors and high levels of estimated that 4% of the US population has corneal NV and
anti-angiogenic factors. Corneal neovascularization (NV) every year 1.4 million patients in the US may develop
corneal NV [5]. Although, the etiology of corneal NV is not
completely identified, numerous pathological conditions
*Address correspondence to this author at the Department of Immunology, such as inflammatory, infectious, degenerative, and
School of Medicine, Tehran University of Medical Sciences, Tehran, Postal traumatic disorders may induce NV. Among these different
Zip Code: 1417613151, Iran; Tel/Fax: +98-21-66419536;
E-mail: yshakiba@razi.tums.ac.ir; y_shakiba@yahoo.com etiologies, infectious disease of the cornea seems to be the

1872-213X/09 $100.00+.00 © 2009 Bentham Science Publishers Ltd.


222 Recent Patents on Inflammation & Allergy Drug Discovery 2009, Vol. 3, No. 3 Shakiba et al.

most important cause of corneal NV leading to vision loss ANGIOGENIC MOLECULES INVOLVED IN
[6]. A typical example is trachoma, a chronic infection of the CORNEAL NEOVASCULARIZATION
conjunctiva caused by the Chlamydia trachomatis, which is Vascular Endothelial Growth Factor
the leading cause of preventable blindness in the world.
Untreated active trachoma is accompanied by corneal NV as Vascular endothelial growth factor (VEGF), also called
pannus (fibrovascular tissue) formation. The pannus may VEGF-A, belongs to a family of closely related growth
extend to central cornea and cause severe disturbance in factors that are characterized by the presence of eight
corneal transparency. It has been estimated that about 5.9 conserved cysteine residues. Other members of this family
million people are blind or have severe vision-loss due to include VEGF-B, VEGF-C, VEGF-D, VEGF-E and placen-
trachoma infection and 10 million are at high risk worldwide tal growth factor. All of the VEGFs are expressed in
[6,7]. Corneal NV is also a common manifestation of herpes mammalian cells, except VEGF-E that is encoded by the orf
simplex keratitis (HSK), which is the most common virus; a parapoxvirus that affects sheep, goats and sometimes
infectious cause of corneal blindness in the Western humans [16, 17]. VEGF is a homodimeric glycoprotein that
countries [8]. Infection of corneal stroma by herpes simplex is encodeds by a gene resides on chromosome 6 [18]. The
virus (HSV) is usually associated with NV. In fact, it has human VEGF gene spans ~14 kb and contains eight exons
been demonstrated that formation of NV is the essential step separated by seven introns [19, 20]. As a result of alternative
for the pathogenesis of HSK and anti-angiogenic agents can splicing, at least 4 types of monomeric VEGF containing
be used for controlling herpes simplex induced corneal 121, 165, 189, and 206 amino acid residues (VEGF121,
angiogenesis [9]. Corneal NV is also a common manifesta- VEGF165, VEGF189, and VEGF206) have been detected.
tion of severe infections of the cornea by other micro- Exons 1 to 5 are common in all VEGF isoforms, while exons
organisms such as staphylococcus, streptococcus and pseu- 6 to 8 can be added to this backbone and form various types
domonas [8]. Although, all infectious keratitis may induce of VEGF [21].
corneal NV, Acanthamoeba keratitis has been reported to VEGF exerts its pivotal role in vasculogenesis (formation
lack corneal NV even in relatively severe and long-standing of blood vessels from endothelial cells) and angiogenesis
cases [10]. (formation of new capillaries from pre-existing vessels)
Corneal NV can affect the prognosis of corneal trans- through binding to specific receptors. The receptor binding
plantation. It has been reported that 20% of corneal site of VEGF is located in exons 3 and 4; thereby all VEGF
specimens obtained during corneal transplantation show isoforms can bind to their receptors [22]. VEGF is thought to
histopathological evidences of NV [11]. In fact, corneal NV signals through at least two transmembrane high-affinity
can significantly worsen the prognosis of penetrating receptor tyrosine kinases: VEGFR-1 (FLT-1; fms-like
keratoplasty (PK). Previous studies showed that 41% of eyes tyrosine kinase) and VEGFR-2 (KDR; kinase domain
in low risk patients developed corneal NV 6 to 9 months region). Binding of VEGF induces dimerization and
after PK. Active blepharitis and stimulation of corneal autophosphory-lation of the receptor, which leads to multiple
stroma by sutures are considered as risk factors for evolution cellular out-comes, including: endothelial cell migration,
of NV after PK [11,12]. Today, there is a growing body of proliferation, survival, and enhanced vascular permeability
interests to prevent NV after PK using anti-angiogenic that occur during vasculogenesis and angiogenesis. In fact,
agents [13]. VEGF indu-ces vascular permeability 50,000 times more
than histamine [23-25].
Contact lenses have become one of the most common There are several evidences that support a causal role for
etiologies of corneal NV in developed countries during past VEGF in corneal NV [26-33]. It has been shown that VEGF
two decades. The prevalence of soft hydrogel lenses induced expression is significantly higher in vascularized corneas
corneal NV is very higher than hard gas-permeable lenses than in normal corneas. Macrophages, infiltrating leuko-
[14]. Although, the precise mechanism of contact lens cytes, corneal epithelial and stromal cells are the main
induced NV is not clear, there are numerous evidences that sources of VEGF production during corneal NV [34]. In fact,
support the role of hypoxia in its pathology. As noted above, it has been reported that application of recombinant VEGF
the cornea supplies oxygen from surface layer by diffusion; induces corneal NV in animal models. VEGF overexpression
thus contact lens, especially soft lenses, can induce hypoxia in the cornea promotes several steps of angiogenesis,
by reducing corneal oxygen uptake. including proteolytic activities, proliferation, migration, and
Contact lens induced hypoxia causes significant changes tube formation of endothelial cells [35]. The role of VEGF
to the corneal cells such as decreased metabolic rate, stromal and VEGFR-2 in the regulation of corneal NV and wound
edema, and decreased endothelial cell density and function. healing was investigated in an animal model of alkali burn
Moreover, long term hypoxia upregulates angiogenic factors [27]. Moreover the overexpression of VEGF and flt-1 has
that induce corneal NV [15]. In overall, corneal NV caused been reported in miscellaneous morbid corneas that may play
by contact lens wear is generally mild and does not affect an important role in the repair of corneal injuries [33].
vision in the aggressive manner that infections do. However, The critical role of VEGF in the pathogenesis of NV
it is a sign of corneal distress, which needs medical attention confirmed by studies that showed anti-VEGF agents can
[8]. inhibit corneal NV. It has been reported that VEGF
Corneal Neovascularization Recent Patents on Inflammation & Allergy Drug Discovery 2009, Vol. 3, No. 3 223

inhibition with mFlt(1-3)-immunoglobulin G diminishes brane during angiogenesis. Their substrates include most of
angiogenesis and the severity of lesions after HSV infection the extracellular matrix components such as collagen, fibro-
[36]. Another study showed that stromal implantation of an nectin, laminin and cell surface molecules. The MMP family
anti-VEGF blocking antibody can inhibit corneal NV in a rat composed of at least 25 different proteinases that are divided
model [37]. In addition, it has been reported that VEGF into five groups, including: the gelatinases, collagenases,
antagonists, like soluble form of the VEGF receptor and anti- stromelysins, membrane-type MMPs, and others. Several
VEGF siRNA, inhibit herpes simplex induced corneal NV in MMPs are believed to be important in angiogenesis, but
mice [20, 32]. Recently, Bevacizumab (Avastin), an anti- particular interest has been focused on the MMP-2 and
VEGF monoclonal antibody, is considered as a safe and MMP-9 (gelatinase A and B, respectively), because they
effective therapeutic option for corneal NV. Avastin has preferentially degrade basement membrane components such
been investigated in both human and animal corneal NV and as type IV collagen [67,68]. In the cornea, both MMP-2 and
the results are very promising. Many studies showed that MMP-9 are produced by the endothelial cells and stromal
topical and subconjunctival application of Avastin inhibits cells around the invading vessels.
chemical and viral induced corneal NV [38-49]. In addition
MMP-2 is expressed in tissues where NV occurs,
to VEGF neutralizing agents, it has been reported that
including solid tumors, wound healing, and choroidal NV in
tyrosine kinase inhibitors that blocks VEGF signaling
age-related macular degeneration [69]. There is a general
through its receptors can inhibit corneal NV and improve consensus that MMP-2 promotes angiogenesis by breaking
corneal graft outcome in animal models [50,51]. To date,
down either the vascular endothelial cell basal membrane or
there are numerous patents that have been focused on the
the surrounding ECM [68]. The over expression of MMP-2
inhibitors of VEGF and its receptor for controlling ocular
during corneal NV has been confirmed by numerous
NV conditions [52-54].
investigations [70-73]. Kvanta et al. reported that MMP-2
expression increases during inflammation-associated corneal
FIBROBLAST GROWTH FACTOR NV in a rat model [74]. A recent study investigated corneal
Basic fibroblast growth factor (bFGF) is another NV in MMP-2-deficient mice compared with wild-type mice
angiogenic factor that its role in induction of corneal NV has and showed statistically significant delay of NV in MMP-2-
been investigated in several studies. bFGF belongs to the deficient mice [75]. Another study reported that MMP-2 is
fibroblast growth factor family, with more than 20 members required for optimal experimental choroidal NV in a mouse
[11, 55]. It has been localized in the corneal epithelial and model [76].
endothelial cells in low levels [56, 57]. Injured corneal MMP-9 (gelatinase B) is one of the major degrading
epithelial cells produce bFGF in response to external stimuli enzymes in ocular surface inflammation. MMP-9 is
[58]. To date, bFGF has been used extensively in corneal overexpressed in vascularized cornea and plays a role in
angiogenesis models and is thought to be one of the major wound healing [77]. There is high level of MMP-9 in tear of
factors involved in the induction of corneal angiogenesis [11, patients with recurrent corneal ulcers [78]. In addition, it has
59]. The angiogenic properties of bFGF are mediated by been reported that in MMP-9-deficient mice, healing of
tyrosine kinase receptors, denoted as FGFR-1, -2, -3, and -4. corneal wounds is accelerated as a result of their increased
Signal transduction through these receptors is essential for keratinocyte proliferation rate [79]. Taken together, these
development and angiogenesis, because FGFR-1 or FGFR-2 data indicate that both MMP-2 and MMP-9 have a role in the
gene disruption leads to embryonic death [60-62]. Numerous disruption of the basement membrane, integrity of the
evidences showed that FGF can stimulate proliferation of the corneal epithelium and corneal NV; thus targeting them may
corneal epithelial cells, stromal fibroblasts, and endothelial be a useful therapeutic option for inhibition of angiogenesis
cells. bFGF is expressed in the normal corneal epithelium, in the cornea. In fact, promising studies showed that MMPs
but is not detectable in corneal epithelial cell basement inhibitors can inhibit corneal NV and help to maintain
membrane (Bowman's layer). Injury to the corneal corneal clarity [80-82].
epithelium results in the releasing of bFGF from epithelial
cells and subsequent binding to the Bowman's layer [63]. It In addition to these mediators, several hormones and
has been shown that FGF-2 is up-regulated in a corneal biologically active molecules may be involved in corneal
stromal cell-vascular endothelial cell coculture [60]. bFGF NV. Numerous studies showed that angiopoietin 1 and 2 [83-
induced micropocket assay is one of the most reliable 85], insulin like growth factor (IGF) [86], placenta growth
methods for induction of NV in the cornea. Placement of low factor (PGF) [87], platelet derived growth factor (PDGF)
levels of bFGF in the cornea in animal models induces new [88], leptin [89] and hepatocyte growth factor (HGF) [90]
blood vessels formation from limbal plexus. Anti-bFGF are contributed to the pathogenesis of corneal NV. Further
agents can inhibit in vitro endothelial cell tube formation and investigations are needed to clarify the exact role of these
in vivo corneal NV, which can indicate the important role of molecules in corneal NV.
bFGF in angiogenesis [64-66].
ANTI-ANGIOGENIC MOLECULES AND CORNEAL
MATRIX METALLOPROTEINASES NEOVASCULARIZATION

Matrix metalloproteinases (MMPs) are considered as Thrombospondin


other key mediators of angiogenesis. They are a family of Thrombospondins (TSP) are glycoproteins encoding by a
zinc-dependent enzymes, essential for degradation of family of five genes that behave as multifunctional proteins.
extracellular matrix (ECM) and vascular basement mem- Among this family, TSP-1 and -2 show potent anti-angio-
224 Recent Patents on Inflammation & Allergy Drug Discovery 2009, Vol. 3, No. 3 Shakiba et al.

genic properties. TSP-1 inhibits critical events in angio- ENDOSTATIN


genesis, including endothelial cell proliferation, survival and
Endostatin is a 20 - kDa fragment of collagen XVIII,
migration [91]. TSP-1 binds to CD36 on endothelial cell
which shows anti-angiogenic properties. Endostatin was first
surface and induces apoptosis [92-94]. Moreover it has been
discovered in the Dr. Judah Folkman laboratory from
reported that TSP-1 inhibits VEGF secretion and down
regulates MMP expression in endothelial cells [95, 96]. In conditioned medium of hemangioendothelioma cells [120].
Anti-angiogenic mechanism of endostatin is mediated by
normal cornea, TSP-1 expresses in the Bowman’s layer,
several mechanisms: 1) Endostatin blocks the activity of
endothelial cells, and Descemet’s membrane [97]. Interes-
gelatinases (MMP-2 and MMP-9) and MMP-13 [121]. 2)
tingly, it has been shown that corneas of TSP1,2-/- mice show
Endostatin inhibits the binding of VEGF to its receptors as
greater NV than normal mice in response to injuries.
well as its signal transduction. It blocks VEGF receptor
Therefore TSP could be a major natural angiogenic inhibitor
in the cornea which plays an active role in preserving tyrosine kinase phosphorylation and downstream signaling
events like activation of ERK, p38 MAPK, and p125FAK
angiogenic privilege [91]. In animal models of corneal NV,
[122]. 3) Binding of endostatin to endothelial cells arrests
TSP-1 derived peptide inhibits bFGF induced angiogenesis
their cell cycle in G1 [123]. 4) Endostatin induces apoptosis
[98]. Moreover, a recent study reported that topical appli-
in endothelial cell by activation of caspase 3 [124]. 5)
cation of mesenchymal stem cells to injured corneas inhibits
Endostatin inhibits endothelial cell tubule formation [125].
NV through production of TSP-1 [99].
The results of microarrays studies covering over 90% of the
human genome showed that endostatin regulated about 12%
PIGMENT EPITHELIUM DERIVED FACTOR of all genes in human endothelial cells [126]. In normal
Pigment epithelium derived factor (PEDF) is a 50 - kDa cornea, collagen XVIII can be found naturally. It seems that
natural anti-angiogenic and neurotrophic protein that was during corneal NV and wound healing, this type of collagen
firstly localized in fetal human retinal pigment epithelial can be digested by MMPs, leading to production of
cells [100]. In the eye, PEDF can be detected in retinal cells, Endostatin like fragments [11, 127,128]. Previous studies
iris, and the cornea [11]. PEDF is a member of serine confirmed the efficacy of endostatin application in control-
protease inhibitors family but in practice it does not inhibit ling corneal NV. In animal models, endostatin inhibits bFGF
serine proteases [101]. In contrast to VEGF that is induced induced corneal NV [129-131]. Altogether, the results of
by low oxygen levels; the expression of PEDF is suppressed numerous studies showed that endostatin and its fragments
by hypoxia [102]. The anti-angiogenic properties of PEDF can be considered as potent and safe anti-angiogenic agents
have been confirmed by numerous studies [103-106]. in ocular NV disorders, including corneal NV [132-134].
Neutralization of PEDF in normal cornea by specific
antibodies induces NV, which shows its important role in THE ROLE OF INFLAMMATORY CYTOKINES IN
corneal angiogenic privilege [11, 102, 107]. It has been CORNEAL NV
reported that application of PEDF can inhibit bFGF induced
As noted above, inflammatory and infectious diseases are
corneal NV in animal models [108]. Further studies are
major etiologies of corneal NV. In fact, corneal NV is almost
needed to clarify the exact role of PEDF in corneal NV.
always accompanied by inflammation. Previous investi-
gations showed that the corneal cells secret various cytokines
ANGIOSTATIN that are involved in ocular surface diseases. In in vitro
Angiostatin is a 38 - kDa protein derived from enzymatic conditions, the corneal epithelial cells express IL-1 [135-
digestion of plasminogen, which is a potent anti-angiogenic 137]; the corneal epithelial, stromal and endothelial cells
factor. It was first found in urine of mice carrying Lewis produce IL-6 [137-139]; and the corneal epithelial and
lung carcinomas [109]. Angiostatin inhibits endothelial cells stromal cells produce IL-8 [140]. It has been reported that
proliferation, migration and tubule formation [11, 110, 111]. IL-1 is the main inflammatory cytokine produced by the
Moreover, angiostatin induces vascular endothelial cell corneal epithelial cells. In the inflamed cornea, IL-1 is
apoptosis, and stops cell growth at the G2/M transition [112]. associated with inflammatory and degradative functions.
Plasminogen and its mRNA were found in all three layers of This cytokine is chemotactic for fibroblasts and inflam-
the cornea. Interestingly, it has been reported that the corneal matory cells and stimulates fibroblast proliferation and
cells have ability to digest plasminogen to angiostatin. In collagenase expression. In addition to the corneal epithelial
fact, all of the essential MMPs for converting plasminogen to cells, IL-1 is produced by infiltrating monocytes and
angiostatin are found in the corneal cells [108]. It seems that macrophages and it’s over expression (along with TNF-) is
angiostatin has a role in maintaining corneal avascularity. one of the early hallmarks of corneal inflammation [141].
Angiostatin can be detected in the tear film of contact lens Previous studies confirmed that the IL-1 receptor, which
bearing patients [113]. Depletion of intra-corneal angiostatin both IL-1 and IL-1 bind, is expressed in the corneal
molecules using specific antibodies increases the risk of NV fibroblasts. Binding of IL-1 to the corneal fibroblast induces
in animal models [114]. In addition, application of angio- the expression of various angiogenic factors [142]. It has
statin inhibits bFGF and angiogenin induced NV [110]. been demonstrated that IL-1 induces NF-B signaling in the
Altogether, these data indicate the importance of angiostatin mouse cornea during NV. In the corneal alkali burns model,
and its fragments in normal and injured cornea and bring it NF-B is activated in the corneal epithelial and stromal cells
to the mind as a potent therapeutic option for clinical uses [143]. Inhibitor of NF-B (IB-) is mainly phosphorylated
[115-119]. in the corneal stromal cells, suggesting that NF-B-activated
corneal stromal fibroblasts play an important role in corneal
inflammatory conditions, such as angiogenesis and wound
Corneal Neovascularization Recent Patents on Inflammation & Allergy Drug Discovery 2009, Vol. 3, No. 3 225

healing [144]. Recently, an interesting study showed that subsets of corneal NV, but it requires high amounts of
topical administration of 2% IL-1 receptor antagonist (IL- thermal laser energy to produce adequate destruction of
1Ra) inhibits inflammation induced corneal NV [141]. vessels, and there is a very high recurrence rate. Moreover,
this high energy can lead to some complications, including
TNF- is another important proinflammatory cytokine
involved in corneal NV. It is a pleiotropic proinflammatory increased inflammation and exacerbation of the NV
[162,163]. Laser photocoagulation is used by some investi-
cytokine that mediates various immunoregulatory functions,
gators for diminishing NV before and after penetrating
such as up-regulation of adhesion molecules, activation of
keratoplasty. Results from laser photocoagulation showed
neutrophils, and activation of the NF-B signaling pathway
that it is more useful for controlling low to moderate forms
[145]. TNF- is produced by various cells in the inflam-
of corneal NV [11].
matory site and induces angiogenic cytokines, including IL-
8, VEGF, and bFGF [146]. TNF- exerts inflammatory Photodynamic therapy is another method that involves
properties through its receptors, TNF-R1 and TNF-R2. the administration of a photosensitizing compound, which is
Binding of TNF- to its receptors on the endothelial cells selectively absorbed by neovascular tissues. In this method,
increases the expression of many angiogenic molecules the photosensitizer may be administered systemic or topical
[147]. Previous studies showed that TNF- is a two-edged on the neovascular cornea. Subsequent activation of this
sword in controlling angiogenesis. It has been demonstrated compound by low energy laser light generates cytotoxic
that angiogenic properties of TNF- is dose-dependent. In mediators, which cause selective newly formed vessel
low doses, TNF- induces angiogenesis, whereas a high dose thrombosis and destruction [164]. This method can be
inhibits it [148]. In the inflamed cornea, TNF- is secreted considered as a safe and effective procedure for controlling
by the corneal resident cells [148,149]. In addition TNF- corneal NV, because total used energy is 20 times less than
significantly increases IL-8 expression in the corneal conventional thermal laser [165]. Moreover, thermal damage
endothelial and stromal cells. IL-8 is a potent chemo- to normal pericorneal tissues is less common. In recent
attractant factor that promotes angiogenesis in vitro as well years, many investigations showed the efficacy of photo-
as in vivo [150]. It has been reported that intracorneal injec- dynamic therapy in both human and animal models of
tion of TNF- increases the number of Langerhans cells in corneal NV [166,167].
limbus and induces migration of them to the central cornea
[151]. More investigations are needed for clarifying the role CORTICOSTEROIDS
of TNF- in corneal NV.
Corticosteroids inhibit inflammation and angiogenesis.
Transforming growth factor  (TGF-) is another main To date, corticosteroid therapy has been considered as the
cytokine that has an important role in corneal NV. TGF- standard anti-inflammatory and anti-angiogenic treatment in
can act as pro- or anti-angiogenic factor depending on the cornea. Previous studies supported this idea that anti-
context and concentration. In low doses, TGF- stimulates angiogenic effect of corticosteroids is due to their anti-
endothelial cells proliferation, whereas excessively high inflammatory properties [168,169]. It has been clarified that
doses can inhibit it [152,153]. During corneal injuries, TGF- various signaling pathways are common in inflammation and
 is produced by inflammatory cells and resident corneal angiogenesis. In the injured cornea, infiltrating inflammatory
cells [154]. TGF- also stimulates angiogenesis by recruiting cells are the major sources of angiogenic molecules like
inflammatory cells which, in turn, release other pro- VEGF. In addition, these cells produce many cytokines like
angiogenic cytokines [155]. It has been reported that three IL-1 and TNF- that are categorized as potent angiogenic
isoforms of TGF- and their receptors are present in the molecules [170]. The steroids exert their anti-inflammatory
cornea [156-159]. In fact, TGF- is present on the edge of effects, mainly by inhibiting the enzyme phospholipase and
wound healing in the cornea [160]. In animal models, thereby the arachidonic acid pathway. Inhibition of phos-
neutralization of TGF- inhibited corneal NV [156]. Further- pholipase decreases production of prostaglandins from cell
more, it seems that TGF- directly stimulates the production membrane lipids. In addition to inflammatory properties,
of VEGF in various cells, including fibroblastic epithelial some prostaglandins act as angiogenic factors. In fact,
cells [161]. Altogether, the exact role of TGF- in corneal steroids behave as anti-angiogenic agents by inhibiting
NV is not completely understood and there are controversial prostaglandin synthesis [171]. Systemically administered
reports regard its role in angiogenesis. Further investigations steroids inhibit the transcription of several inflammatory and
are needed for understanding its role in normal and angiogenic cytokines, including IL-1, IL-3, IL-6, and IL-8
vascularized cornea. [172]. Moreover, previous studies notified that reducing the
expression of VEGF may be another mechanism of steroids
THERAPEUTIC OPTIONS anti-angiogenic effects [173].
Surgical Treatments The first investigated steroids had both anti-inflammation
and anti-angiogenic activities and some of them appeared to
To date, numerous surgical methods have been recom-
require specific cofactors such as heparin or cyclodextran
mended for controlling corneal NV. The first investigations
[174]. Although, this type of steroids has anti-angiogenic
considered diathermy for destruction of newly formed blood
effects, they may cause significant ocular side effects like
vessels in the cornea. In recent years, other techniques like
development of posterior subcapsular cataracts and elevated
laser photocoagulation have been evaluated in the treatment
intra ocular pressure [175-177]. In recent years, new class of
of corneal NV. The effect of laser coagulation is due to steroids discovered, which has broad angiostatic and low
energy absorption by hemoglobin in blood vessels. It seems
glucocorticoid activities. This class of compounds is consi-
that this technique is effective in treatment of numerous
226 Recent Patents on Inflammation & Allergy Drug Discovery 2009, Vol. 3, No. 3 Shakiba et al.

dered as potential therapy for ocular neovascular disease, approved by the US Food and Drug Administration (FDA)
since undesirable side effects, which are commonly asso- for use in the treatment of metastatic colorectal cancer [189].
ciated with glucocorticoids, would be avoided [178,179]. Previous studies confirmed that topical or subconjuctival
application of Bevacizumab controls new blood vessel
Anecortave acetate is a known member of this class of
steroids. This anti-angiogenic drug is derived from the formation in the cornea [190-193]. It is reported that single
subconjunctival dose of Bevacizumab inhibits NV in alkali
glucocorticoid cortisol acetate and has no glucocorticoid
burned rabbit corneas [194]. Moreover, several case reports
activity. Anecortave acetate has broad based angiostatic
notified the therapeutic properties of Bevacizumab in
activity and inhibits critical steps of angiogenesis. It inhibits
patients with corneal NV [49, 195]. Interestingly, the recent
the angiogenic proteolytic cascade at several points by
studies showed the efficacy of Bevacizumab in inhibiting
suppressing production of uPA and proMMPs from
endothelial cells and increasing the expression of PAI-1, an herpetic corneal NV. A recent investigation reported that
subconjunctival application of Bevacizumab decreases
endogenous inhibitor of uPA. Moreover anecortave acetate
corneal NV dramatically 1 week after the injection in a
decreases the expression of gelatinase A and B and inhibits
woman with herpetic stromal keratitis [196]. In addition to
VEGF-induced bovine and human endothelial cell
Bevacizumab, recent evidences showed that inhibitors of
proliferation and tube formation [180]. In animal models, it
VEGF receptors can be efficient in controlling corneal NV.
has been reported that topical administration of anecortave
acetate suppresses LPS and FGF2 induced corneal NV. In In animal models, tyrosine kinase inhibitors decrease corneal
NV and prolong graft survival [51].
that study, it has been reported that application of 0.1% or
1% suspensions of anecortave acetate four times in day Taken together, these data indicate that Bevacizumab can
almost totally inhibits corneal NV in rabbits [168]. After be a promising drug in treatment of cornea NV and graft
confirming safety and efficacy by more investigations, ane- survival [197,198]. Anti-VEGF substances may increasingly
cortave acetate can be considered for controlling corneal NV. play a role in treatment of anterior segment NV in the future.
In addition to these well-known therapeutic strategies
NONSTEROIDAL ANTI-INFLAMMATORY DRUGS described above, there are numerous agents that showed
Nonsteroidal anti-inflammatory drugs (NSAIDs) are inhibitory effects in corneal NV. It has been reported that
widely used in ophthalmology for controlling pain and ‘suleparoide [199], thalidomide [200], suramin [201],
inflammation in ocular surface disorders. NSAIDs inhibit the genistein [202], somatostatin [203], octreotide [204],
activity of the cyclooxygenases (COX-1 and COX-2) that cyclosporine [205], methotrexate [206], rapamycin [207] and
convert arachidonic acid to prostaglandins (PGs). Previous tacrolimus [208] can abolish corneal NV in animal models.
studies showed that COX-2 is an inducible isoform of However, it seems that these medications exert their
cyclooxygenases, which induces multiple events of therapeutic properties through reducing inflammatory and
angiogenesis like proliferation and migration of endothelial angiogenic factors, mainly VEGF. Further investigations in
cells and inhibits apoptosis [181]. It has been clarified that animal models and human cases are needed to place these
some of PGs, like PGE2, induces angiogenesis by increasing agents alongside corneal NV therapeutics.
VEGF and bFGF gene expression [182,183]. In fact, it has
been showed that inhibitors of COX-2 inhibit angiogenesis CURRENT & FUTURE DEVELOPMENTS
in vitro and in vivo [184-186]. Both COX-1 and COX-2 are
During past two decades, paralle with our understanding
found in the cornea and their role in the pathogenesis of NV
of molecular events underlying ocular NV, there was a
has been investigated [187].
general interesting for using anti-VEGF angents as a potent
Previous studies showed that NSAIDs can control and safe theraputic option for controlling corneal
corneal NV. Indomethacin and ketoprofen, nonselective angiogenesis. It is expected that VEGF blocking agents or
COX-1 and COX-2 inhibitors, significantly suppress bFGF- tyrosin kinase inhibitors will play an imporant role in
and VEGF-induced angiogenesis in mouse cornea [184]. increasing the graft survival and conserving vision after
Interestingly, this study notified that selective inhibitor of keratoplasty in near future. Moreover, VEGF is known as a
COX-2 (celecoxib and rofecoxib) suppresses bFGF-induced main mediator involves in the pathogenesis of herpetic
NV more effectively than VEGF induced NV. Thus it was keratitis and pterygium; therefore anti-VEGF drugs can
concluded that selective inhibitors of COX-2 should be used revolutionize their treatment strategies. Today, at least two
in the early course of corneal NV and before the upregu- VEGF inhibitors are approved for treatment of angiogenesis-
lation of large amounts of VEGF. Once VEGF is present, related ophthalmic diseases; Lucentis (rhuFab V2;
selective COX-2 inhibitors would be less active in inhibiting Genentech, Inc.) and Macugen (pegaptanib sodium; Eyetech
VEGF-induced angiogenesis [184]. Pharmaceuticals) and more are poised to enter into clinical
practice. Focus on determination of safe and effective
ANTI-VEGF AGENTS concentration, exposure time, and administration routes of
anti-angiogenic drugs can accelerate their entrance to
Over the past decade, there were progressive attempts for ophthalmology in near future.
using anti VEGF agents for controlling corneal NV.
Numerous studies showed that Bevacizumab can inhibit
ACKNOWLEDGEMENT
ocular NV in both human and animal models. Bevacizumab
is a recombinant humanized monoclonal antibody against the The authors would like to thank Dr. Amir Kiani and
VEGF molecule that binds and neutralizes it effectively Dr. Behrouz Nikbin for their valuable assistance.
[188]. Today, Bevacizumab is commercially available and is
Corneal Neovascularization Recent Patents on Inflammation & Allergy Drug Discovery 2009, Vol. 3, No. 3 227

CONFLICT OF INTEREST [26] Singh N, Amin S, Richter E, et al. Flt- 1 intraceptors inhibit
hypoxia-induced VEGF expression in vitro and corneal
No financial contribution to the work has been declared. neovascularization in vivo. Invest Ophthalmol Vis Sci 2005; 46(5):
No patents are reported in any stage of legal litigations. 1647-1652.
[27] Gan L, Fagerholm P, Palmblad J. Vascular endothelial growth
factor VEGF and its receptor VEGFR-2 in the regulation of corneal
REFERENCES neovascularization and wound healing. Acta Ophthalmol Scand
2004; 82(5): 557-563.
[1] Niederkorn JY. Immune privilege in the anterior chamber of the [28] Joussen AM, Poulaki V, Mitsiades N, et al. VEGF-dependent
eye. Crit Rev Immunol 2002; 22(1): 13-46.
conjunctivalization of the corneal surface. Invest Ophthalmol Vis
[2] Gipson IK, Inatomi T. Cellular origin of mucins of the ocular Sci 2003; 44(1): 117-123.
surface tear film. Adv Exp Med Biol 1998; 438: 221-227.
[29] Zhou LH, Xing YQ, Chen CL, et al. Antisense vascular endothelial
[3] Hosseini H, Nejabat M. A potential therapeutic strategy for growth factor suppressed corneal neovascularization in rats.
inhibition of corneal neovascularization with new anti-VEGF
Zhonghua Yan Ke Za Zhi 2006; 42(5): 426-430.
agents. Med Hypotheses 2007; 68(4): 799-801. [30] Murata M, Takanami T, Shimizu S, et al. Inhibition of ocular
[4] Jain RK. Molecular regulation of vessel maturation. Nat Med 2003;
angiogenesis by diced small interfering RNAs (siRNAs) specific to
9(6): 685-693. vascular endothelial growth factor (VEGF). Curr Eye Res 2006;
[5] Colby KA, Adamis AP. Prevalence of corneal neovascularization in
31(2): 171-180.
a general eye service population (Abstract). Invest Ophthalmol Vis [31] Lai CM, Spilsbury K, Brankov M, et al. Inhibition of corneal
Sci 1996; 37: S593.
neovascularization by recombinant adenovirus mediated antisense
[6] Zhang SX, Ma JX. Ocular neovascularization: Implication of VEGF RNA. Exp Eye Res 2002; 75(6): 625-634.
endogenous angiogenic inhibitors and potential therapy. Prog Retin
[32] Kim B, Tang Q, Biswas PS, et al. Inhibition of ocular angiogenesis
Eye Res 2007; 26(1): 1-37. by siRNA targeting vascular endothelial growth factor pathway
[7] West SK. Trachoma: New assault on an ancient disease. Prog Retin
genes: Therapeutic strategy for herpetic stromal keratitis. Am J
Eye Res 2004; 23(4): 381-401. Pathol 2004; 165(6): 2177-2185.
[8] Lee P, Wang CC, Adamis AP. Ocular neovascularization: An
[33] Chen J, Liu W, Liu Z, et al. Expression of vascular endothelial
epidemiologic review. Surv Ophthalmol 1998; 43(3): 245-269. growth factor and its receptors (FLT-1) in morbid human corneas
[9] Zheng M, Schwarz MA, Lee S, et al. Control of stromal keratitis by
and investigation of its clinic importance. Yan Ke Xue Bao 2002;
inhibition of neovascularization. Am J Pathol 2001; 159(3): 1021- 18(4): 203-207.
1029.
[34] Seta F, Patil K, Bellner L, et al. Inhibition of VEGF expression and
[10] Kremer I, Cohen EJ, Eagle RC Jr, et al. Histopathologic evaluation corneal neovascularization by siRNA targeting cytochrome P450
of stromal inflammation in Acanthamoeba keratitis. CLAO J 1994;
4B1. Prostaglandins Other Lipid Mediat 2007; 84(3-4): 116-127.
20(1): 45-48. [35] Phillips GD, Stone AM, Jones BD, et al. Vascular endothelial
[11] Chang JH, Gabison EE, Kato T, et al. Corneal neovascularization.
growth factor (rhVEGF165) stimulates direct angiogenesis in the
Curr Opin Ophthalmol 2001; 12(4): 242-249. rabbit cornea. In Vivo 1994; 8(6): 961-965.
[12] Dana MR, Schaumberg DA, Kowal VO, et al. Corneal
[36] Zheng M, Deshpande S, Lee S, et al. Contribution of vascular
neovascularization after penetrating keratoplasty. Cornea 1995; endothelial growth factor in the neovascularization process during
14(6): 604-609.
the pathogenesis of herpetic stromal keratitis. J Virol 2001; 75(20):
[13] Gerten G. Bevacizumab (avastin) and argon laser to treat 9828-9835.
neovascularization in corneal transplant surgery. Cornea 2008;
[37] Amano S, Rohan R, Kuroki M, et al. Requirement for vascular
27(10): 1195-1199. endothelial growth factor in wound- and inflammation related
[14] Rozenman Y, Donnenfeld ED, Cohen EJ, et al. Contact lens-related
corneal neovascularization. Invest Ophthalmol Vis Sci 1998; 39(1):
deep stromal neovascularization. Am J Ophthalmol 1989; 107(1): 18-22.
27-32.
[38] Uy HS, Chan PS, Ang RE. Topical bevacizumab and ocular surface
[15] Liesegang TJ. Physiologic changes of the cornea with contact lens neovascularization in patients with stevens-johnson syndrome.
wear. CLAO J 2002; 28(1): 12-27.
Cornea 2008; 27(1): 70-73.
[16] Li X, Eriksson U. Novel VEGF family members: VEGF-B, VEGF- [39] Mackenzie SE, Tucker WR, Poole TR. Bevacizumab (avastin) for
C and VEGF-D. Int J Biochem Cell Biol 2001; 33(4): 421-426.
corneal neovascularization-corneal light shield soaked application.
[17] Lyttle DJ, Fraser KM, Fleming SB, et al. Homologs of vascular Cornea 2009; 28(2): 246-247.
endothelial growth factor are encoded by the poxvirus orf virus. J
[40] Bahar I, Kaiserman I, McAllum P, et al. Subconjunctival
Virol 1994; 68(1): 84-92. bevacizumab injection for corneal neovascularization in recurrent
[18] Vincenti V, Cassano C, Rocchi M, et al. Assignment of the vascular
pterygium. Curr Eye Res 2008; 33(1): 23-28.
endothelial growth factor gene to human chromosome 6p21.3. [41] Gerten G. Bevacizumab (avastin) and argon laser to treat
Circulation 1996; 93(8): 1493-1495.
neovascularization in corneal transplant surgery. Cornea 2008;
[19] Houck KA, Leung DW, Rowland AM, et al. Dual regulation of 27(10): 1195-1199.
vascular endothelial growth factor bioavailability by genetic and
[42] Han YS, Lee JE, Jung JW, et al. Inhibitory effects of bevacizumab
proteolytic mechanisms. J Biol Chem 1992; 267(36): 26031-26037. on angiogenesis and corneal neovascularization. Graefes Arch Clin
[20] Tischer E, Mitchell R, Hartman T, et al. The human gene for
Exp Ophthalmol 2009; 247(4): 541-548.
vascular endothelial growth factor. Multiple protein forms are [43] Bock F, Onderka J, Rummelt C, et al. Safety profile of topical
encoded through alternative exon splicing. J Biol Chem 1991;
VEGF-neutralization at the cornea. Invest Ophthalmol Vis Sci
266(18): 11947-11954. 2009; 50(5): 2095-2102.
[21] Robinson CJ, Stringer SE. The splice variants of vascular
[44] Hurmeric V, Mumcuoglu T, Erdurman C, et al. Effect of
endothelial growth factor (VEGF) and their receptors. J Cell Sci subconjunctival bevacizumab (Avastin) on experimental corneal
2001; 114: 853-865.
neovascularization in guinea pigs. Cornea 2008; 27(3): 357-362.
[22] Ng YS, Krilleke D, Shima DT. VEGF function in vascular [45] Chen WL, Lin CT, Lin NT, et al. Subconjunctival injection of
pathogenesis. Exp Cell Res 2006; 312(5): 527-537.
bevacizumab (avastin) on corneal neovascularization in different
[23] Senger DR, Connolly DT, Van de Water L, et al. Purification and rabbit models of corneal angiogenesis. Invest Ophthalmol Vis Sci
NH2-terminal amino acid sequence of guinea pig tumor-secreted
2009; 50(4): 1659-1665.
vascular permeability factor. Cancer Res 1990; 50(6): 1774-1778. [46] Kim TI, Kim SW, Kim S, et al. Inhibition of experimental corneal
[24] Meadows KN, Bryant P, Pumiglia K. Vascular endothelial growth
neovascularization by using subconjunctival injection of
factor induction of the angiogenic phenotype requires Ras bevacizumab (Avastin). Cornea 2008; 27(3): 349-352.
activation. J Biol Chem 2001; 276(52): 49289-49298.
[47] Bahar I, Kaiserman I, McAllum P, et al. Subconjunctival
[25] Takahashi T, Yamaguchi S, Chida K, et al. A single bevacizumab injection for corneal neovascularization. Cornea 2008;
autophosphorylation site on KDR/Flk-1 is essential for VEGF-A-
27(2): 142-147.
dependent activation of PLC-gamma and DNA synthesis in [48] Doctor PP, Bhat PV, Foster CS. Subconjunctival bevacizumab for
vascular endothelial cells. EMBO J 2001; 20(11): 2768-2778.
corneal neovascularization. Cornea 2008; 27(9): 992-995.
228 Recent Patents on Inflammation & Allergy Drug Discovery 2009, Vol. 3, No. 3 Shakiba et al.

[49] Kim SW, Ha BJ, Kim EK, et al. The effect of topical bevacizumab factor (VEGF) in inflammation-associated corneal neovas-
on corneal neovascularization. Ophthalmology 2008; 115(6): e33- cularization. Exp Eye Res 2000; 704: 419-428.
38. [75] Samolov B, Steen B, Seregard S, et al. Delayed inflammation-
[50] Shakiba Y, Arshadi D. Inhibition of corneal neovascularization with associated corneal neovascularization in MMP-2-deficient mice.
new tyrosine kinase inhibitors targeting vascular endothelial growth Exp Eye Res 2005; 802: 159-166.
factor receptors: Sunitinib malate and Sorafenib. Irn J Med [76] Berglin L, Sarman S, van der Ploeg I, et al. Reduced choroidal
Hypotheses Ideas 2007; 1:1 neovascular membrane formation in matrix metalloproteinase
[51] Hos D, Bock F, Dietrich T, et al. Inflammatory corneal (lymph) metalloproteinase-2-deficient mice. Invest Ophthalmol Vis Sci
angiogenesis is blocked by VEGFR-tyrosine kinase inhibitor ZK 2003; 441: 403-408.
261991, resulting in improved graft survival after corneal [77] Ye HQ, Azar DT. Expression of gelatinases A and B, and TIMPs 1
transplantation. Invest Ophthalmol Vis Sci 2008; 49(5): 1836-1842. and 2 during corneal wound healing. Invest Ophthalmol Vis Sci
[52] Kimihiko, F., Tatsuro, I., Tadahisa, K., Yukio, S.: EP0811711A1 1998; 39: 913-921.
(2006). [78] Sakimoto T, Shoji J, Yamada A, et al. Upregulation of matrix
[53] Rosenblum, M.G., Campochiaro, P. A.: US2007025957A1 (2007). metalloproteinase in tear fluid of patients with recurrent corneal
[54] Adamis, A. P., Guyer, D. R., Modi, M. W.: US2006293270A1 erosion. Jpn J Ophthalmol 2007; 51(5): 343-346.
(2006). [79] Swarnakar S, Ganguly K, Kundu P, et al. Curcumin regulates
[55] Hayashi N, Nakayasu K, Okisaka S. Immunohistochemical expression and activity of matrix metalloproteinases 9 and 2 during
localization of acidic and basic fibroblast growth factor through prevention and healing of indomethacin-induced gastric ulcer. J
corneal neovascularization in vivo and in vitro. Nippon Ganka Biol Chem 2005; 280(10): 9409-9415.
Gakkai Zasshi 1996; 100: 587-591. [80] Gonzales RP, D-Soares FS, Farias RF, et al. Demonstration of
[56] Gan L, Fagerholm P, Palmblad J. Expression of basic fibroblast inhibitory effect of oral shark cartilage on basic fibroblast growth
growth factor in rabbit corneal alkali wounds in the presence and factor-induced angiogenesis in the rabbit cornea. Biol Pharm Bull
absence of granulocytes. Acta Ophthalmol Scand 2005; 83(3): 374- 2001; 24: 151-154.
378. [81] Tamargo RJ, Bok RA, Brem H. Angiogenesis inhibition by
[57] Saghizadeh M, Chwa M, Aoki A, et al. Altered expression of minocycline. Cancer Res 1991; 51(2): 672-675.
growth factors and cytokines in keratoconus, bullous keratopathy [82] Sucholeiki, I., Gege, C., Gallagher, B, Powers, T., Deng, H., Wu,
and diabetic human corneas. Exp Eye Res 2001; 73(2): 179-189. X., Steeneck, C., Kiely, A., Taveras, A.: US20080021024A1
[58] Adamis AP, Meklir B, Joyce NC. in situ Injury-induced release of (2008).
basic-fibroblast growth factor from corneal epithelial cells. Am J [83] Singh N, MacNamara E, Rashid S, et al. Systemic soluble Tie-2
Pathol 1991; 139: 961-967. expression inhibits and regresses corneal neovascularization.
[59] Rogers MS, Birsner AE, D'Amato RJ. The mouse cornea Biochem Biophys Res Commun 2005; 332: 194-199.
micropocket angiogenesis assay. Nat Protoc 2007; 2(10): 2545- [84] White RR, Shan S, Rusconi CP, et al. Inhibition of rat corneal
2550. angiogenesis by a nuclease-resistant RNA aptamer specific for
[60] Kojima T, Chang JH, Azar DT. Proangiogenic role of angiopoietin-2. Proc Natl Acad Sci USA 2003; 100: 5028-5033.
ephrinB1/EphB1 in basic fibroblast growth factor-induced corneal [85] Oike Y, Ito Y, Maekawa H, et al. Angiopoietin-related growth
angiogenesis. Am J Pathol 2007; 170(2): 764-773. factor (AGF) promotes angiogenesis. Blood 2004; 103(10): 3760-
[61] Xu X, Weinstein M, Li C, et al. Fibroblast growth factor receptor 2 3765.
(FGFR2)-mediated reciprocal regulation loop between FGF8 and [86] Grant MB, Mames RN, Fitzgerald C, et al. Insulin-like growth
FGF10 is essential for limb induction. Development 1998; 125(4): factor I acts as an angiogenic agent in rabbit cornea and retina:
753-765. Comparative studies with basic fibroblast growth factor.
[62] Deng CX, Wynshaw-Boris A, Shen MM, et al. Murine FGFR-1 is Diabetologia 1993; 36: 282-291.
required for early postimplantation growth and axial organization. [87] Cao Y, Linden P, Shima D, Browne F, Folkman J. in vivo
Genes Dev 1994; 8: 3045-3057. Angiogenic activity and hypoxia induction of heterodimers of
[63] Soubrane G, Jerdan J, Karpouzas I, et al. Binding of basic fibroblast placenta growth factor/vascular endothelial growth factor. J Clin
growth factor to abnormal and neovascular rabbit cornea. Invest Invest 1996; 98(11): 2507-2511.
Opthalmol Vis Sci 1990; 31: 323-333. [88] Cohen RA, Gebhardt BM, Bazan NG. A platelet-activating factor
[64] Kurokawa M, Doctrow SR, Klagsbrun M. Neutralizing antibodies antagonist reduces corneal allograft inflammation and
inhibit the binding of basic fibroblast growth factor to its receptor neovascularization. Curr Eye Res 1994; 13(2): 139-144.
but not to heparin. J Biol Chem 1989; 264(13): 7686-7691. [89] Park HY, Kwon HM, Lim HJ, et al. Potential role of leptin in
[65] Xiang, J., Deng, N., Li, H.: CN1560082A (2004). angiogenesis: Leptin induces endothelial cell proliferation and
[66] Yoshitake, Y., Nishikawa, K., Osawa, M, Asano, M., Koda, A., expression of matrix metalloproteinases in vivo and in vitro. Exp
Suzuki, H.: JP11049701A (1997) Mol Med 2001; 33(2): 95-102.
[67] Visse R, Nagase H. Matrix metalloproteinases and tissue inhibitors [90] Xin X, Yang S, Ingle G, et al. Hepatocyte growth factor enhances
of metalloproteinases: Structure, function, and biochemistry. Circ vascular endothelial growth factor-induced angiogenesis in vitro
Res 2003; 928: 827-839. and in vivo. Am J Pathol 2001; 158(3): 1111-1120.
[68] Pepper MS. Role of the matrix metalloproteinase and plasminogen [91] Cursiefen C, Masli S, Ng TF, et al. Roles of thrombospondin-1 and
activator-plasmin systems in angiogenesis. Arterioscler Thromb -2 in regulating corneal and iris angiogenesis. Invest Ophthalmol
Vasc Biol 2001; 217: 1104-1117. Vis Sci 2004; 45: 1117-1124.
[69] Mignatti P, Rifkin DB. Plasminogen activators and matrix [92] Nor JE, Mitra RS, Sutorik M, et al. Thrombospondin-1 induces
metalloproteinases in angiogenesis. Enzyme Protein 1996; 49(1-3): endothelial cell apoptosis and inhibits angiogenesis by activating
117-137. the caspase death pathway. J Vasc Res 2000; 37: 209-218.
[70] Kato T, Kure T, Chang JH, et al. Diminished corneal angiogenesis [93] Jimenez B, Volpert OV, Crawford SE, et al. Signals leading to
in gelatinase A-deficient mice. FEBS Lett 2001; 5082: 187-190. apoptosis-dependent inhibition of neovascularization by
[71] Zhang H, Li C, Baciu PC. Expression of integrins and MMPs thrombospondin-1. Nat Med 2000; 6: 41-48.
during alkaline-burn-induced corneal angiogenesis. Invest [94] Vande Woude, G. F., Zhang, Y.W.: US20070020234A1 (2007).
Ophthalmol Vis Sci 2002; 434: 955-962. [95] Zak S, Treven J, Nash N, et al. Lack of thrombospondin-1 increases
[72] Rigal-Sastourne JC, Tixier JM, Renard JP, et al. Corneal burns and angiogenesis in a model of chronic inflammatory bowel disease. Int
matrix metalloproteinases MMP-2 and -9: The effects of human J Colorectal Dis 2008; 23(3): 297-304.
amniotic membrane transplantation. J Fr Ophtalmol 2002; 257: [96] Zhou L, Isenberg JS, Cao Z, et al. Type I collagen is a molecular
685-693. target for inhibition of angiogenesis by endogenous
[73] Ma DH, Chen JK, Kim WS, et al. Expression of matrix thrombospondin-1. Oncogene 2006; 25: 536-545.
metalloproteinases 2 and 9 and tissue inhibitors of metallo- [97] Hiscott P, Seitz B, Schlotzer-Schrehardt U, et al. Immunolo-
proteinase 1 and 2 in inflammation-induced corneal neovas- calisation of thrombospondin 1 in human, bovine and rabbit cornea.
cularization. Ophthalmic Res 2001; 336: 353-362. Cell Tissue Res 1997; 289(2): 307-310.
[74] Kvanta A, Sarman S, Fagerholm P, et al. Expression of matrix
metalloproteinase-2 (MMP-2) and vascular endothelial growth
Corneal Neovascularization Recent Patents on Inflammation & Allergy Drug Discovery 2009, Vol. 3, No. 3 229

[98] Kanda S, Shono T, Tomasini-Johansson B, et al. Role of [123] Hanai J, Dhanabal M, Karumanchi SA, et al. Endostatin causes G1
thrombospondin-1-derived peptide, 4N1K, in FGF-2-induced arrest of endothelial cells through inhibition of cyclin D1. J Biol
angiogenesis. Exp Cell Res 1999; 252(2): 262-272. Chem 2002; 277: 16464-16469.
[99] Oh JY, Kim MK, Shin MS, et al. The anti-inflammatory and anti- [124] Dhanabal M, Ramchandran R, Waterman MJ, et al. Endostatin
angiogenic role of mesenchymal stem cells in corneal wound induces endothelial cell apoptosis. J Biol Chem 1999; 274: 11721-
healing following chemical injury. Stem Cells 2008; 26(4): 1047- 11726.
1055. [125] Urbich C, Reissner A, Chavakis E, et al. Dephosphorylation of
[100] Tombran-Tink J, Johnson LV. Neuronal differentiation of endothelial nitric oxide synthase contributes to the antiangiogenic
retinoblastoma cells induced by medium conditioned by human effects of endostatin. FASEB J 2002; 16: 706-708.
RPE cells. Invest Ophthalmol Vis Sci 1989; 30: 1700-1707. [126] Abdollahi A, Hahnfeldt P, Maercker C, et al. Endostatin’s
[101] Becerra SP, Sagasti A, Spinella P, et al. Pigment epithelium- antiangiogenic signaling network. Mol Cell 2004; 13: 649-663.
derived factor behaves like a noninhibitory serpin. Neurotrophic [127] Ferreras M, Felbor U, Lenhard T, et al. Generation and degradation
activity does not require the serpin reactive loop. J Biol Chem 1995; of human endostatin proteins by various proteinases. FEBS Lett
270(43): 25992-25999. 2000; 486: 247-251.
[102] Dawson DW, Volpert OV, Gillis P, et al. Pigment epithelium- [128] Maatta M, Heljasvaara R, Pihlajaniemi T, et al. Collagen
derived factor: A potent inhibitor of angiogenesis. Science 1999; XVIII/endostatin shows a ubiquitous distribution in human ocular
285(5425): 245-248. tissues and endostatin-containing fragments accumulate in ocular
[103] Notari L, Miller A, Martinez A, et al. Pigment epithelium-derived fluid samples. Graefes Arch Clin Exp Ophthalmol 2007; 245(1):
factor is a substrate for matrix metalloproteinase type 2 and type 9: 74-81.
Implications for downregulation in hypoxia. Invest Ophthalmol Vis [129] Lai LJ, Xiao X, Wu JH. Inhibition of corneal neovascularization
Sci 2005; 46(8): 2736-2747. with endostatin delivered by adeno-associated viral (AAV) vector
[104] Abe R, Fujita Y, Yamagishi S, et al. Pigment epithelium-derived in a mouse corneal injury model. J Biomed Sci 2007; 14(3): 313-
factor prevents melanoma growth via angiogenesis inhibition. Curr 322.
Pharm Des 2008; 14(36): 3802-3809. [130] Niu XG, Wang W, Shi WY, et al. Inhibition of corneal
[105] Iizuka H, Awata T, Osaki M, et al. Promoter polymorphisms of the neovascularization by liposomes mediated plasmid encoding human
pigment epithelium-derived factor gene are associated with diabetic endostatin. Zhonghua Yan Ke Za Zhi 2005; 41(3): 260-264.
retinopathy. Biochem Biophys Res Commun 2007; 361(2): 421- [131] Zhang P, Wu D, Ge J, et al. Experimental inhibition of corneal
426. neovascularization by endostatin gene transfection in vivo. Chin
[106] Bouck N. PEDF: Anti-angiogenic guardian of ocular function. Med J 2003; 116(12): 1869-1874.
Trends Mol Med 2002; 8(7): 330-334. [132] Folkman, J., Javaherian, K., Robert, T.T.: US20060251699A1
[107] Meyer C, Notari L, Becerra SP. Mapping the type I collagen- (2006).
binding site on pigment epithelium-derived factor. Implications for [133] Folkman, M.J., O'Reilly, M.: US20050282253A1 (2005).
its antiangiogenic activity. J Biol Chem 2002; 277: 45400-45407. [134] O'Reilly, M., Folkman, M. J.: US20020123458A1 (2002).
[108] Azar DT. Corneal angiogenic privilege: Angiogenic and [135] Wilson SE, He YG, Lloyd SA. EGF, EGF receptor, basic FGF,
antiangiogenic factors in corneal avascularity, vasculogenesis, and TGF beta-1, and IL-1 alpha mRNA in human corneal epithelial
wound healing (an American Ophthalmological Society thesis). cells and stromal fibroblasts. Invest Ophthalmol Vis Sci 1992;
Trans Am Ophthalmol Soc 2006; 104: 264-302. 33(5): 1756-1765.
[109] O’Reilly MS, Holmgren L, Shing Y, et al. Angiostatin: A novel [136] Shams NB, Sigel MM, Davis RM. Interferon-gamma,
angiogenesis inhibitor that mediates the suppression of metastases Staphylococcus aureus, and lipopolysaccharides/ silica enhance
by a Lewis lung carcinoma. Cell 1994; 79: 315-328. interleukin-1 beta production by human corneal cells. Reg Immunol
[110] Cao R, Wu HL, Veitonmaki N, et al. Suppression of angiogenesis 1989; 2: 136-148.
and tumor growth by the inhibitor K1-5 generated by plasmin- [137] Sakamoto S, Inada K, Chiba K, et al. Production of IL-6 and IL-1a
mediated proteolysis. Proc Natl Acad Sci USA 1999, 96: 5728- by human corneal epithelial cells. Acta Soc Ophthalmol Jpn 1991;
5733. 95: 728-732.
[111] Shin SH, Kim JC, Chang SI, et al. Recombinant kringle 1-3 of [138] Sakamoto S, Inada K. Human corneal epithelial, stromal and
plasminogen inhibits rabbit corneal angiogenesis induced by endothelial cells produce interleukin-6. Acta Soc Ophthalmol
angiogenin. Cornea 2000; 19: 212-217. Jpn1992; 96: 702-709.
[112] Griscelli F, Li H, Bennaceur-Griscelli A, et al. Angiostatin gene [139] Cubitt CL, Lausch RN, Oakes JE. Differences in interleukin-6 gene
transfer: Inhibition of tumor growth in vivo by blockage of expression between cultured human corneal epithelial cells and
endothelial cell proliferation associated with a mitosis arrest. Proc keratocytes. Invest Ophthalmol Vis Sci 1995; 36: 330-336.
Natl Acad Sci USA 1998; 95: 6367-6372. [140] Cubitt CL, Tang Q, Monteiro CA, et al. IL-8 gene expression in
[113] Sack RA, Beaton AR, Sathe S. Diurnal variations in angiostatin in cultures of human corneal epithelial cells and keratocytes. Invest
human tear fluid: A possible role in prevention of corneal Ophthalmol Vis Sci 1993; 34: 3199-3206.
neovascularization. Curr Eye Res 1999; 18: 186-193. [141] Dana R. Comparison of topical interleukin-1 vs tumor necrosis
[114] Gabison E, Chang JH, Hernandez-Quintela E, et al. Anti- factor-alpha blockade with corticosteroid therapy on murine corneal
angiogenic role of angiostatin during corneal wound healing. Exp inflammation, neovascularization, and transplant survival (an
Eye Res 2004; 78: 579-558. American Ophthalmological Society thesis). Trans Am Ophthalmol
[115] Folkman, J., O'Reilly, M., Cao, Y., Sim, K.: US20090075369A1 Soc 2007; 105: 330-343.
(2009). [142] Hong JW, Liu JJ, Lee JS, et al. Proinflammatory chemokine
[116] Soff, G., Gately, S.T., Twardowski, P.: US20060099671A1 (2006). induction in keratocytes and inflammatory cell infiltration into the
[117] O'Reilly, M.S., Folkman, M. J., Cao, Y.: US20040023877A1 cornea. Invest Ophthalmol Vis Sci 2001; 42: 2795-2803.
(2004). [143] Saika S, Miyamoto T, Yamanaka O, et al. Therapeutic effect of
[118] Folkman, M. J., O'Reilly, M. S., Cao, Y., Sim, K.L.: topical administration of SN50, an inhibitor of nuclear factor-B, in
US20030064926A1 (2003). treatment of corneal alkali burns in mice. Am J Pathol 2005; 166:
[119] O'Reilly, M.S., Folkman, M.J., Cao, Y.: US20010029246A1 1393-1403.
(2001). [144] Nakao S, Hata Y, Miura M, et al. Dexamethasone inhibits
[120] O’Reilly MS, Boehm T, Shing Y, et al. Endostatin: An endogenous interleukin-1-induced corneal neovascularization role of nuclear
inhibitor of angiogenesis and tumor growth. Cell 1997; 88: 277- factor-B-activated stromal cells in inflammatory angiogenesis. Am
285. J Pathol 2007; 171(3): 1058-1065.
[121] Nyberg P, Heikkila P, Sorsa T, et al. Endostatin inhibits human [145] Eigler A, Sinha B, Hartmann G, et al. strategies to restrain this
tongue carcinoma cell invasion and intravasation and blocks the proinflammatory cytokine. Immunol Today 1997; 18: 487-492.
activation of matrix metalloprotease-2, - 9, and -13. J Biol Chem [146] Henricson BE, Benjamin WR, Vogel SN. Differential cytokine
2003; 278(25): 22404-22411. induction by doses of lipopolysaccharide and monophosphoryl lipid
[122] Kim YM, Hwang S, Pyun BJ, et al. Endostatin blocks vascular A that result in equivalent early endotoxin tolerance. Infect Immun
endothelial growth factor-mediated signaling via direct interaction 1990; 58: 2429-2437.
with KDR/Flk-1. J Biol Chem 2002; 277: 27872-27879.
230 Recent Patents on Inflammation & Allergy Drug Discovery 2009, Vol. 3, No. 3 Shakiba et al.

[147] Yoshida S, Ono M, Shono T, et al. Involvement of interleukin-8, [171] Haynes WL, Proia AD, Klintworth GK. Effect of inhibitors of
vascular endothelial growth factor, and basic fibroblast growth arachidonic acid metabolism on corneal neovascularization in the
factor in tumor necrosis factor alpha-dependent angiogenesis. Mol rat. Invest Ophthalmol Vis Sci 1989; 30(7): 1588-1593.
Cell Biol 1997; 17: 4015-4023. [172] Pleyer U, Dannowski H, Volk HD, et al. Corneal allograft rejection:
[148] Niederkorn JY, Peeler JS, Mellon J. Phagocytosis of particulate Current understanding. I. Immunobiology and basic mechanisms.
antigens by corneal epithelial cells stimulates interleukin-1 Ophthalmologica 2001; 215(4): 254-262.
secretion and migration of Langerhans cells into the central cornea. [173] Nauck M, Karakiulakis G, Perruchoud AP, et al. Corticosteroids
Reg Immunol 1989; 2: 83-90. inhibit the expression of the vascular endothelial growth factor gene
[149] Staats HF, Lausch RN. Cytokine expression in vivo during murine in human vascular smooth muscle cells. Eur J Pharmacol 1998;
herpetic stromal keratitis. Effect of protective antibody therapy. J 341(2-3): 309-315.
Immunol 1993; 151: 277-283. [174] Crum R, Szabo S, Folkman J. A new class of steroids inhibits
[150] Elner VM, Strieter RM, Pavilack MA, et al. Human corneal angiogenesis in the presence of heparin or a heparin fragment.
interleukin-8 IL-1 and TNF-induced gene expression and secretion. Science 1985; 230(4732): 1375-1378.
Am J Pathol 1991; 139: 977-988. [175] Gillies MC, Kuzniarz M, Craig J, et al. Intravitreal triamcinolone-
[151] Dekaris I, Zhu SN, Dana MR. TNF-alpha regulates corneal induced elevated intraocular pressure is associated with the
Langerhans cell migration. J Immunol1999; 162: 4235-4239. development of posterior subcapsular cataract. Ophthalmology
[152] Dorrell M, Uusitalo-Jarvinen H, Aguilar E, et al. Ocular 2005; 112(1): 139-143.
neovascularization: Basic mechanisms and therapeutic advances. [176] Jonas J, Heatley G, Spaide R, et al. Intravitreal triamcinolone
Surv Ophthalmol 2007; 52: 3-19. acetonide and secondary ocular hypertension. J Glaucoma 2005;
[153] Schreiber AB, Winkler ME, Derynck R. Transforming growth 14(2): 168-171.
factor-alpha: A more potent angiogenic mediator than epidermal [177] Jonas JB, Kreissig I, Hugger P, et al. Intravitreal triamcinolone
growth factor. Science 1986; 232(4755): 1250-1253. acetonide for exudative age related macular degeneration. Br J
[154] Cursiefen C, Rummelt C, Kuchle M. Immunohistochemical Ophthalmol 2003; 87(4): 462-468.
localization of vascular endothelial growth factor, transforming [178] Clark, A. F.: US5770592A (1998).
growth factor alpha, and transforming growth factor beta1 in human [179] Clark, A. F., Conrow R. E.: EP1236471A2 (2002).
corneas with neovascularization. Cornea 2000; 19(4): 526-533. [180] Clark AF. Mechanism of action of the angiostatic cortisene
[155] Pepper MS. Transforming growth factor-beta: Vasculogenesis, anecortave acetate. Surv Ophthalmol 2007; 52: S26-34.
angiogenesis, and vessel wall integrity. Cytokine Growth Factor [181] Gately S. The contributions of cyclooxygenase-2 to tumor
Rev 1997; 8(1): 21-43. angiogenesis. Cancer Metastasis Rev 2000; 19: 19 -27.
[156] Sakamoto T, Ueno H, Sonoda K, et al. Blockade of TGF-beta by [182] Form DM, Auerbach R. PGE2 and angiogenesis. Proc Soc Exp Biol
in vivo gene transfer of a soluble TGF-beta type II receptor in the Med 1983; 172: 214-218.
muscle inhibits corneal opacification, edema and angiogenesis. [183] Harada S, Nagy JA, Sullivan KA, et al. Induction of vascular
Gene Ther 2000; 7(22): 1915-1924. endothelial growth factor expression by prostaglandin E2 and E1 in
[157] Pasquale LR, Dorman-Pease ME, Lutty GA, et al. osteoblasts. J Clin Invest 1994; 93: 2490-2496.
Immunolocalization of TGF-beta 1, TGF-beta 2, and TGF-beta 3 in [184] Pakneshan P, Birsner AE, Adini I, et al. Differential suppression of
the anterior segment of the human eye. Invest Ophthalmol Vis Sci vascular permeability and corneal angiogenesis by nonsteroidal
1993; 34(1): 23-30. anti-inflammatory drugs. Invest Ophthalmol Vis Sci 2008; 49(9):
[158] Nishida K, Kinoshita S, Yokoi N, et al. Immunohistochemical 3909-3913.
localization of transforming growth factor-beta 1, -beta 2, and -beta [185] Jones MK, Wang H, Peskar BM, et al. Inhibition of angiogenesis
3 latency-associated peptide in human cornea. Invest Ophthalmol by nonsteroidal anti-inflammatory drugs: insight into mechanisms
Vis Sci 1994; 35(8): 3289-3294. and implications for cancer growth and ulcer healing. Nat Med
[159] Joyce NC, Zieske JD. Transforming growth factor-beta receptor 1999; 5: 1418-1423.
expression in human cornea. Invest Ophthalmol Vis Sci 1997; [186] Tsujii M, Kawano S, Tsuji S, et al. Cyclooxygenase regulates
38(10): 1922-1928. angiogenesis induced by colon cancer cells. Cell 1998; 93: 705-716.
[160] Hayashi K, Frangieh G, Wolf G, et al. Expression of transforming [187] Amico C, Yakimov M, Catania MV, et al. Differential expression
growth factor-beta in wound healing of vitamin A-deficient rat of cyclooxygenase-1 and cyclooxygenase-2 in the cornea during
corneas. Invest Ophthalmol Vis Sci 1989; 30: 239-247. wound healing. Tissue Cell 2004; 36(1): 1-12.
[161] Pertovaara L, Kaipainen A, Mustonen T, et al. Vascular endothelial [188] Shibuya M. Structure and function of VEGF/VEGF-receptor system
growth factor is induced in response to transforming growth factor- involved in angiogenesis. Cell Struct Funct 2001; 26(1): 25-35.
beta in fibroblastic and epithelial cells. J Biol Chem 1994; 269(9): [189] Hurwitz H, Fehrenbacher L, Novotny W, et al. Bevacizumab plus
6271-6274. irinotecan, fluorouracil, and leucovorin for metastatic colorectal
[162] Baer JC, Foster CS. Corneal laser photocoagulation for treatment of cancer. N Engl J Med 2004; 350: 2335-2342.
neovascularization. Efficacy of 577 nm yellow dye laser. [190] Bock F, König Y, Kruse F, et al. Bevacizumab (Avastin) eye drops
Ophthalmology 1992; 99(2): 173-179. inhibit corneal neovascularization. Graefes Arch Clin Exp
[163] Nirankari VS, Baer JC. Corneal argon laser photocoagulation for Ophthalmol 2008; 246(2): 281-284.
neovascularization in penetrating keratoplasty. Ophthalmology [191] Mackenzie SE, Tucker WR, Poole TR. Bevacizumab (avastin) for
1986; 93: 1304-1309. corneal neovascularization--corneal light shield soaked application.
[164] Ahmad N, Mukhtar H. Mechanism of photodynamic Cornea 2009; 28(2): 246-247.
therapyinduced cell death. Methods Enzymol 2000; 319: 342-358. [192] You IC, Kang IS, Lee SH, et al. Therapeutic effect of
[165] Sheppard Jr, Epstein RJ, Lattanzio Jr, et al. Argon laser subconjunctival injection of bevacizumab in the treatment of
photodynamic therapy of human corneal neovascularization after corneal neovascularization. Acta Ophthalmol 2008; 87(6): 653-658.
intravenous administration of dihematoporphyrin ether. Am J [193] Han YS, Lee JE, Jung JW, et al. Inhibitory effects of bevacizumab
Ophthalmol 2006; 141(3): 524-529. on angiogenesis and corneal neovascularization. Graefes Arch Clin
[166] Yoon KC, You IC, Kang IS, et al. Photodynamic therapy with Exp Ophthalmol 2009; 247(4): 541-548.
verteporfin for corneal neovascularization. Am J Ophthalmol 2007; [194] Hosseini H, Nejabat M, Mehryar M, et al. Bevacizumab inhibits
144(3): 390-395. corneal neovascularization in an alkali burn induced model of
[167] Clark, A.F.: US6297228B1 (1999). corneal angiogenesis. Clin Exp Ophthalmol 2007; 35(8): 745-748.
[168] BenEzra D, Griffin BW, Maftzir G, et al. Topical formulations of [195] Qian CX, Bahar I, Levinger E, et al. Combined use of superficial
novel angiostatic steroids inhibit rabbit corneal neovascularization. keratectomy and subconjunctival bevacizumab injection for corneal
Invest Ophthalmol Vis Sci 1997; 38: 1954-1962. neovascularization. Cornea 2008; 27(9): 1090-1092.
[169] Phillips K, Arffa R, Cintron C, et al. Effects of prednisolone and [196] Carrasco MA. Subconjunctival bevacizumab for corneal neovas-
medroxyprogesterone on corneal wound healing, ulceration, and cularization in herpetic stromal keratitis. Cornea 2008; 27(6): 743-
neovascularization. Arch Ophthalmol 1983; 101: 640-643. 745.
[170] Planck SR, Rich LF, Ansel JC, et al. Trauma and alkali burns [197] D'amato, R.J., Green, S.J., Swartz Jr., G. M., Shah, J.H., Madsen,
induce distinct patterns of cytokine gene expression in the rat J.: US20050004087A1 (2005).
cornea. Ocul Immunol Inflamm 1997; 5(2): 95-100. [198] D'amato, R.: US6071948 (2000).
Corneal Neovascularization Recent Patents on Inflammation & Allergy Drug Discovery 2009, Vol. 3, No. 3 231

[199] Benelli U, Bocci G, Danesi R, et al. The heparan sulfate [204] Demir T, Celiker UO, Kukner A, et al. Effect of Octreotide on
suleparoide inhibits rat corneal angiogenesis and in vitro experimental corneal neovascularization. Acta Ophthalmol Scand
neovascularization. Exp Eye Res 1998; 67: 133-142. 1999; 77: 386-390.
[200] Joussen AM, Germann T, Kirchhof B. Effect of thalidomide and [205] Reis A, Reinhard T, Sundmacher R, Braunstein S, Godehardt E. A
structurally related compounds on corneal angiogenesis is comparative investigation of FK506 and cyclosporin A in murine
comparable to their teratological potency. Graefes Arch Clin Exp corneal transplantation. Graefes Arch Clin Exp Ophthalmol 1998;
Ophthalmol 1999; 237(12): 952-961. 236: 785-789.
[201] Bocci G, Danesi R, Benelli U, et al. Inhibitory effect of suramin in [206] Joussen AM, Kruse FE: Topical application of methotrexate for
rat models of angiogenesis in vitro and in vivo. Cancer Chemother inhibition of corneal angiogenesis. Graefe’s Arch Clin Exp
Pharmacol 1999; 43: 205-212. Ophthalmol 1999; 237: 920-927.
[202] Fotsis T, Pepper M, Adlercreutz H, et al. Genistein, a dietary- [207] Kwon YS, Hong HS, Kim JC, Shin JS, Son Y. Inhibitory effect of
derived inhibitor of in vitro angiogenesis. Proc Natl Acad Sci USA rapamycin on corneal neovascularization in vitro and in vivo. Invest
1993; 90: 2690-2694. Ophthalmol Vis Sci 2005; 46: 454-460.
[203] Wu PC, Liu CC, Chen C, et al. Inhibition of experimental [208] Sloper CM, Powell RJ, Dua HS. Tacrolimus in the management of
angiogenesis of cornea by somatostatin. Graefe’s Arch Clin Exp high risk corneal and limbal grafts. Ophthalmology 2001; 108:
Ophthalmol 2003; 241: 63-69. 1838-1844.

S-ar putea să vă placă și