throbber

`
`
`
`iduosnuewJOLlanva-HINiduosnuewJouinvva-HIN.
`
`
`
`
`
`
`
`
`
`idiJasnuewJOllanva-HIN
`
`g i
`
`9%»?
`
`NIH Public Access
`
`Author Manuscript
`
`Published in final edited form as:
`
`Frog Rem? Etc Res. 2008 July ‘_. 27(4): 331—37]. doi:10.l0l6ij.pretcyercs.2008.05.00l.
`
`Vascular Endothelial Growth Factor in Eye Disease
`
`J.S. Penna, A. Madanb. R.B. Caldwellc. M. Bartolic, R.W. Caldwell“, and ME. Hartnettd
`
`a\l'anderbilt University School of Medicine, Nashville, TN
`
`blStanford University School of Medicine, Palo Alto, CA
`
`l=Medical College of Georgia, Augusta, GA
`
`dUniversity of North Carolina School of Medicine, Chapel Hill, NC
`
`Abstract
`
`Collectively. angiogenic oeular conditions represent the leading cause of irreversible vision loss in
`developed countries. In the U.S.. for example. retinopathy of prematurity. diabetic retinopathy and
`age-related macular degeneration are the principal causes of blindness in lite infant. working age
`and elderly populations. respectively. Evidence suggests that vascular endothelial growth factor
`(VEGF). a 40 kDa dimcric glycoprotcin. promotes angiogenesis in each of these conditions,
`making it a highly significant therapeutic target. However. VEGF is plciotropic, affecting a broad
`spectrum of endothelial. neuronal and glial behaviors, and confounding the validity of anti-VEGF
`strategies, particularly under chronic disease conditions. In fact. among other functions VEGF can
`influence cell proliferation, cell migration. proteolysis. cell survival and vessel permeability in a
`wide variety of biological contexts. This article will describe the roles played by VEGF in the
`pathogenesis of retinopathy of prematurity. diabetic rctinopathy and a ge-relatcd macular
`degeneration. The potential disadvantages of inhibiting VEGF will be discussed, as will the
`rationales for targeting other VEGF-related modulators of angiogenesis.
`
`Keywords
`
`retina: angiogenesis: vascular endothelial growth factor; age-related macular degeneration:
`diabetic rctinopathy; rctinopathy of prematurity
`
`1. Introduction
`
`1.1 Vascular Endothelial Growth Factor (VEGF)
`
`Vascular endothelial growth factor (VEGF). a dimcric glycoprotein of approximately 40
`kDa. is a potent- endothelial cell mitogen that stimulates proliferation, migration and tube
`formation leading to angiogenic gr0w1h of new blood vessels. It is essential for angiogenesis
`during development; the deletion of a single allele arrests angiogenesis and causes
`embryonic lethality (Ferrara et al., 1996). In mammals. the VEGF family consists of seven
`members: VEGF-A (typically. and hereafter. referred to as VEGF). VEGF-B. VEGF-C.
`VEGF-D. VEGF-E. VEGF-F and PlGF (placental growth factor) (Fig. IA). Alternative
`
`
`it} 2008 Elsevier Ltd. .-'\ll rights reserved.
`Corresponding author: .iohn S. Penn. Pli.D,, Vanderbilt Eye Institute. 8016 Medical Center East, North Tower. Vanderbilt University
`School ol‘ Medicine. Nashville. TN 3T232—8808.
`
`Publisher's Disclaimer: This is a l-‘Dl-‘ file oi‘an unedited manuscript that has been accepted for publication. As a service to our
`customers we are providing this early version of the manuscript. 'llie manuscript will undergo copyediting, typesetting. and review of
`the resulting proof before it is published in its filial citable form. Please note that during the production process errors may be
`discovered which could affect the content, and all legal disclaimers that apply to tlte journal pertain.
`
`Regeneron Exhibit 1055.001
`
`

`

`
`
`1duosnuewJOLfll’lVVd'HlNiduosnuewJOLfll’lVVd‘HlN
`
`
`
`
`
`
`
`
`
`1duosnuewJouinvVd-HIN
`
`Penn et a].
`
`Page 2
`
`splicing results in several VEGF variants. In humans. these include the relatively abundant
`VEGFlgl. VEGF165. VEGF 189 and VEGF206. and several less abundant forms (Fig. 1B).
`The solubility of these splice variants (collectively referred to as VEGFXXX) is dependent on
`heparin binding affinity. VEGF206 and VEGF 189 bind very tightly to heparin and. thus.
`remain sequestered in the extracellular matrix. VEGF165 binds heparin with less affinity. but
`also can be associated with the matrix. and VEGF 121 lacks heparin-binding capacity.
`rendering it highly soluble. Investigations in mice genetically engineered to express less than
`the full complement of splice variants confirm that the relative solubility of VEGF splice
`variants strongly affects their specific bioactivities {Takahashi and Shibuya. 2005).
`Moreover. plasrnin and various rnetalloproteinases can cleave VEGF165. resulting in an N-
`terminal lI3-arnino acid peptide that is non-heparin—binding. but retains its bioactivity (Keyt
`et al.. 1996'. Ferrara et al.. 2003). Our understanding of the relative expression of the
`different VEGF isofonns under normal or pathological oorrditions and the molecular
`regulators of VEGF alternative splicing is relatively limited.
`
`Recently. discovery of the so called “VEGFXXXIJ isofonns” has sparked new interest in the
`molecular events that regulate VEGF expression (for review. see Ladomery et al.. 2007).
`The VEGFXXXb isofomrs share approximately 94—98% homology with the corresponding
`VEGFxxx isofonns and have the same length. However. due to alterations in the C-
`terminus they bind to VEGF receptors. but do not fully activate them and act as "dominant
`negative splice variants” {Bates et al.. 2002). Studies showng that VEGF165b is
`dowruegulated in angiogenic tissues (Ladomery et al.. 2007) suggest a primary role for this
`isoforrn in controlling VEGF activity in health and disease. Administration of VEGF 165b has
`been shown to inhibit retinal angiogenesis in the mouse model of oxygen-induced
`retinopathy (Konopatskaya et al.. 2006).
`
`1.2 VEGF Receptors
`
`VEGFR-lr'Flt-l (fms-like tyrosine kinase) and VEGFR-Zr’KDRIFIk-l (kinase insert domain-
`containing receptori’fetal liver kinase), along with structurally related receptors. Flt-3fFlk-2
`and VEGFR-3lFlt-4. belong to the receptor tyrosine kinase family (Fig. 1A) (Hanks and
`QuimL 1991: Blume-Jensen and Hunter. 2001). VEGFR-I and -2 are primarily involved in
`angiogenesis. (Yancopoulos et al.. 2000) whereas Flt-3 and Flt-4 are involved in
`hematopoiesis and lymphangiogenesis (Jussila and Alitalo. 2000). The VEGFRs contain an
`approximately 750-anuno-acid-residue extracellular domain. which is organized into seven
`irmnunoglobulin—like folds. Adjacent to the extracellular domain is a single trausrnernbrarre
`region. followed by a juxtamembrane domain, a split tyrosine-kinase domain that is
`interrupted by a TO-amino-acid kinase insert, and a C-tenninal tail.
`
`VEGF receptor activation requires dimerization. Guided by the binding properties of the
`ligands, VEGFRs forrrr both lrornodimers and lreterodirners (Rahimi. 2006). The signal
`transduction properties of the VEGFR heterodimers. compared with hornodirners. remain to
`be fully elucidated. Dimerization of VEGFR is accompanied by activation of receptor kinase
`activity. leading to autoplrosphorylation. Site-directed mutagenesis studies have
`demonstrated that the Tyrl2 l4 residue. located in the carboxy terminus of VEGFR-2. is
`required for the ligand-dependent autophosphorylation of the receptor and its ability to
`activate signaling proteins. Signal transduction is propagated when activated VEGF
`receptors plrosphorylate SH2 domain-containing protein substrates.
`
`In addition to VEGFRs. VEGF serves as a ligand to another family of receptors. the
`neuropilins. Neuropilins are 120- to I30-kDa non-tyrosine kinase receptors that mediate
`critical functions in tumor cells and the nervous and vascular systems. In errdotlrelial cells.
`neuropilins serve as receptors for the class 3 semaphorirrs and co-receptors for VEGF family
`members. The role of Neuropilin—I (NRP-I) in mediating VEGF activity is now being
`
`ng Rem: Eye Res. Author manuscript; available in PMC 2013 June 14.
`
`Regeneron Exhibit 1055.002
`
`

`

`
`
`1duosnuewJOLfll’lVVd'HlNtduosnuewJOLflnVVd‘HlN
`
`
`
`
`
`
`
`
`
`tduosnuew.100thVd-HIN
`
`Penn et a].
`
`Page 3
`
`elucidated. VEGF signaling through NRP-l stimulates endothelial cell migration and
`adhesion. The addition of an anti-NRP-l antibody suppressed the mitogenic effects of
`VEGF165 on bovine retinal endothelial cells (RECs) (011 et al.. 2002). In another in Vitro
`model. the VEGF-dependent differentiation of a subset of human bone marrow-derived cells
`into vascular precursors. and subsequent proliferation of these cells. required the activation
`of a VEGFR-2l'NRP-l -dependent signaling pathway (Fons et al.. 2004). Finally. VEGF
`promotion of the synthesis and release of prostacyclin (P612). an important mediator of
`angiogenesis. is thought to be mediated via NRP-l binding (Neagoe et al.. 2005). The
`angiogenic effects regulated tlu‘ough VEGF binding to NRP-2 are less well characterized
`and appear to be modulated differently from the effects controlled by NRP-l. For example.
`VEGF selectively up-regulates NRP-l, but not NRP-2, on endothelial cells (Oh et al.. 2002).
`
`BlAcore analysis has shown NRP-l to interact with VEGFR-l. greatly reducing its binding
`affinity for VEGF165 (Fuh et al.. 2000). Co-culture systems of endothelial cells and breast
`carcinoma cells indicate that NRP-l significantly enhances VEGF165 binding to VEGFR—2
`(Soker et al.. 2002). In aortic endothelial cells, NRP-2 interacted with VEGFR-l. but less is
`known at present about how this influences VEGF bioactivity (Gluzman-Poltorak et al..
`2001). Finally. using multiple in who systems. NRP-2 was shown to interact with
`VEGFR-3. leading to lymphartgiogenic activity, but no interaction was seen between NRP-2
`and VEGFR-2 (Karpanen et al.. 2006).
`
`1.3 VEGF Signaling
`
`Few SH2 domain-containing proteins have been shown to interact diIECtly with VEGFR-2.
`Phospholipase C -*y (PLC‘y) binds to phosphorylated Tyrl 175 (Tyrl 173 in the mouse). and
`mediates the activation of the mitogen—activated protein kinase (MAPK) cascade. leading to
`proliferation of endothelial cells (Takahashi et al.. 2001) (Fig. 2). PLCy activates protein
`kinase C via the production of diacylglycerol and increased concentrations of intracellular
`calcium. A Tyrl 173Phe mutation of VEGFR-2 causes embryonic lethality due to vascular
`defects. mimicking the defects of VEGFR-2 " mice (Sakurai et al.. 2005). These data
`demonstrate an essential function of the Tyr1173 residue during vascular development.
`
`In addition to PLCT. the adaptor molecule. Shb, also binds to phosphorylated Tyrl 175.
`VEGF-induced migration and PI3K activation is inhibited by small interfering RNA
`(siRNA)—mediated knockdown of Shb in endothelial cells (Holmqvist et al.. 2004). The
`Serinefthreonine kinase. Akt. is activated downstream of PI3K and mediates endothelial cell
`
`survival (Fuj io and Walsh. 1999). Akt also regulates nitric oxide (NO) production by direct
`phosphorylation and activation of endothelial NO synthase (eNOS). Finally. phosphorylated
`Tyr1175 is known to interact with Sck (Igarashi et al.. 1998; Sakai et al.. 2000). an adaptor
`molecule that binds Grb2, and participates in MAPK signaling in the epidermal growth
`factor pathway (Thelemann et al.. 2005).
`
`Another important phosphorylation site in VEGFR—2 is Tyr951 (Tyr‘949 in the mouse). a
`binding site for the signaling adaptor VRAP (VEGF receptor-associated protein)
`(Matsumoto et al.. 2005). The phosphorylated Tyr951-VRAP pathway has been shown to
`regulate endothelial cell migration (Matsumoto et al.. 2005; Zeng et al.. 2001). Reduced
`microvessel density and tumor growth in VRAPT— mice confirm an essential function for
`this residue in endothelial cells of the angiogenic phenotype (Matsumoto et al.. 2005).
`VEGF induces the formation of a complex between VRAP and Src (Matsumoto et al..
`2005). indicating that VRAP might regulate Src activation and its signaling downstream of
`VEGFR-2.
`
`Mice that express 2] Tyr] 2 I 2Phe (corresponding to the human Tyr1214) VEGFR-Z mutant
`are viable and fertile (Sakurai et al.. 2005). However. phosphorylation of Tyr1212s’1214 has
`
`ng Rem: Eye Res. Author manuscript; available in PMC 2013 June 14.
`
`Regeneron Exhibit 1055.003
`
`

`

`
`
`tduosnuewJOLfll’lVVd'HlNrduosnuewJOLflI’lVVd‘HlN
`
`
`
`
`
`
`
`
`
`tduosnuewJoutnvVd-HIN
`
`Penn et a].
`
`Page 4
`
`been implicated in VEGF-induced actin remodeling through the sequential activation of
`CDC42 and p38 MAPK (Lamalice et al.. 2004). Inhibition of p38 MAPK augments VEGF—
`induced angiogenesis in the chicken chorioallantoic membrane (CAM) assay (Issbmcker et
`al.. 2003; Matsurnoto et al.. 2002). without an accompanying increase in vascular
`permeability (lssbrucker et al.. 2003). Moreover. p38 MAPK induces phosphorylation of
`heat-shock protein-27 (HSP27). a molecular chaperone that positively regulates VEGF-
`induced actin reorganization and migration (Mthrllen etal.. 2005‘. Rousseau et al.. 1997).
`
`The existence of multiple ligands and receptors provides initial diversity to VEGF
`bioactivity. Groupings of receptor homo- and heterodimers. activated by both common and
`specific ligands, further augment the diversity of VEGF signaling. A final level of diversity
`is provided by activation of distinct signaling intermediates downstream of each VEGF
`receptor. The combination of these features yields an elaborate signaling network capable of
`regulating the extremely complex angiogenic cascade.
`
`1.4 Regulation of VEGF Expression
`
`During hypoxia. VEGF gene expression increases via several different mechanisms (Dor et
`al.. 2001). These mechanisms include increased transcription. mRNA stability. and protein
`translation using an internal ribosornal entry site. as well as increased expression of oxygen
`regulated protein 150. a chaperone required for intracellular transport of proteins from the
`endoplasmic reticulum to the Golgi apparatus prior to secretion (Chen and Slryu. 1995;
`Forsythe etal., 1996; Levy et al.. 1996‘, Levy et al.. 1998; Ozawa et al.. 2001).
`
`The increase in VEGF transcription is largely mediated via hypoxia inducible factor-l
`(HLF-l) (Fig. 3). HLF-l is a heterodimeric transcription factor composed of two subunits —
`the constitutively produced HlF-lfi subunit and the inducible component. HIF-lo. (Wang
`and Semenza. 1995). Under normoxic conditions. HlF- la. is inactivated and targeted for
`proteasoma] degradation by hydroxylation. whereas under hypoxic conditions the specific
`hydroxylases are inhibited. resulting in the rescue of HIP-la. from degradation (Schofield
`and Ratcliffe. 2004). When this occurs. HlF-lo. complexes with HLF- l|3_. translocates into
`the nucleus and binds to a specific sequence in the 5’ flanking region of the VEGF gene. the
`hypoxia responsive element (HRE) (lkeda et al.. 1995. Laughner et al.. 2001: Shirna et al..
`1996; Wenger. 2002; Forsythe et al.. 1996). The importance of interaction between HIF-la.
`and the VEGF promoter has been confirmed in studies of HIP-107"" mouse embryonic stem
`cells. in which basal expression of VEGF mRNA remains low in response to hypoxia
`(Canneliet et al.. 1998; Iyer et al.. 1998).
`
`Two additional isoforms of HIF, known as HIP-2o. and HIP-3a.. have been identified by
`screening for proteins that associate with HIP-[[3 (Ratcliffe. 2007). HIP-2o. appears to be
`closely related to HlF- lo. and can promote HRE-dependent gene transcription. While
`structurally and functionally similar. HIP-lo. and HLF-2o. appear to exert different biological
`functions. as demonstrated in studies using knockout mice (Hu et al.. 2003). For example.
`while HlF- lo. antagonizes c—Myc function. inhibiting renal cell carcinoma {RCC) growth.
`HlF-2o. promotes cell cycle progression in hypoxic RC C and many other cell lines (Gordan
`et al.. 2007). Interestingly. the most distantly related isoform, HIP-3a. appears to antagonize
`HRE-dependerrt gene expression. suggesting a possible negative influence on hypoxia-
`induced gene expression. Additional study is needed to determine if HIP-2o. or HLF-3u. is
`involved in the regulation of retinal VEGF expression.
`
`Clearly. post-transcriptional events are also important in the regulation of VEGF production
`in the diseased retina, as underscored by the correlation of polymorphisms within the 5'-
`untranslated region (UTR) of the VEGF gene with the occurrence of age-related Inacular
`degeneration (AMD). The 3’ UTR and the 5'UTR of the VEGF gene are important sites of
`
`ng Refit: Eye Res. Author manuscript; available in PMC 2013 June 14.
`
`Regeneron Exhibit 1055.004
`
`

`

`
`
`idliosnuewJoutnvVd'HlNtduosnuewJOLfll’lVVd‘HlN
`
`
`
`
`
`
`
`
`
`tduosnuewJoutnvVd-HIN
`
`Penn et a].
`
`Page 5
`
`regulation controlling lnRN A stability and the rate of translation through the internal
`ribosomal entry site (IRES) (for review, see Yoo et al._. 2006).
`
`Evidence for the increase in VEGF lnRNA stability in response to hypoxia comes from in
`who mRNA degradation assays that have led to the identification of adenylateturidylate-rich
`elements (ARES) in the 3 ’ UTR of VEGF lnRNA. VEGF mRNA is extremely labile in
`normoxic conditions. with a half-life of less than 1 hour. as compared with the average half-
`life of 10 to 12 hours for eukaryotic mRNA. During hypoxia the half-life of VEGF mRNA
`increases by two to tluee-fold (levy et al.. 1996) due to a stabilizing effect of HuR a 36
`kDa RNA-binding protein which binds with high affinity to AREs in the 3 ’ -UTR of VEGF
`mRNA. protecting it from degradation by endonucleases (Brennan and Steitz. 2001;
`Robinow et al.. 1988).
`
`Post—transcriptional regulation can also occur in the in 5’-UTR of VEGF mRNA. This
`region contains multiple fRES. These are specific sites of attachment to the ribosornal
`machinery, which provide sites for initiation of translation alternative to the classical 5' cap-
`and elF-dependent translational system (for review. see van der Velden and Thomas, I999).
`Several IRES have been identified in the 5' -UTR of VEGF mRNA, and these provide
`alternative sites of translational control of VEGF expression (Bornes et al.. 2004). Notably.
`evidence suggests that IRES sites in the VEGF 5’-UTR can potentially control the
`generation of alternatively spliced VEGF (Bornes et al.. 2004: Huez et al.. 2001).
`
`Another regulatory mechanism consists of increased production of oxygen regulated protein
`150 (ORP150) in response to hypoxia. Studies using human macrophages transfected with
`adenovirus coding for ORP150 showed that overexpression of ORP150 resulted in increased
`VEGF secretion in hypoxia. Evidence suggests that under hypoxic conditions, ORPI 50
`functions as a molecular chaperone to facilitate VEGF protein transpon and secretion
`(Oyawa et al., 2001). VEGF is not only regulated by hypoxia.
`
`VEGF function is also affected by insulin like growth factor 1 (IGF- l) which plays an
`important role in retinal vaSCulariyation. Several lines of evidence, including in i-‘frm studies.
`suppon the notion that lGF-l is critical for vessel development (King et al._. 1985; Grant et
`al., 1993). Preterm infants with reduced serum levels of IGF-I have a higher incidence of
`development of retinopathy (Hellstrom et al., 2003). Mice null for the lGF-l gene have
`retarded retinal vaswlar growth. compared to wild type controls (Hellstrorn et al., 2001 ).
`However. the action of lGF-l is not mediated by decreasing VEGF expression. as the
`amount of VEGF mRNA is similar in knock out and wild type control mice: instead lGF-l
`acts by decreasing VEGF activation of the Akt signaling pathway. Both MAPK and Akt
`pathways have been shown to be necessary for endothelial cell survival (Smith et al._. 1999).
`
`1.5 Retinal Expression of VEGF and VEGFR
`
`The influence of VEGF in retinal diseases is profound. It has been implicated in a large
`number of retinal diseases and conditions including, but not limited to, highly prevalent
`conditions like AMD and diabetic retinopathy: less common disorders such as retinopathy of
`prematurity. sickle cell retinopathy and retinal vascular occlusion; and as a non-causal. but
`important. secondary influence in neovaSCular glaucoma (Bock et al.. 2007) and inherited
`retinal dy strophies (Penn et al._. 2000). Collectively, these conditions, all of which have
`critical angiogenic components. account for the vast majority of irreversible vision loss in
`developed countries.
`
`At least five retinal cell types have the capacity to produce and secrete VEGF. These include
`the retinal pigmented epithelium (RPE) (Miller et al., 1997), astrocytes (Stone et al._. 1995),
`Muller cells (Robbins et al., 1997), vaSCular endotltelium (Aiello et al._. 1995) and ganglion
`
`ng Refit: Eye Res. Author manuscript; available in PMC 2013 June I4.
`
`Regeneron Exhibit 1055.005
`
`

`

`
`
`tdtrosnuewJoutnvVd'HlNtdtrosnuewrouinvVd‘HlN
`
`
`
`
`
`
`
`
`
`tduosnuewJouinvVd-HIN
`
`Penn el al.
`
`Page 6
`
`cells (Ida et 31., 2003). These cells differ widely in their responses to hypoxia; r}: rr'tro
`studies show that Mtiller cells and astrocytes generally produce the greatest amounts of
`VEGF under hypoxic conditions (Morrison et al.. 2007; Aiello et al, 1995; Hata et al..
`1995). To date. the relative capacity of these cells to produce specific splice variants remains
`unclear. as do the patterns of splice variant production tluoughout retinal development and
`aging.
`
`The distinct roles of the different VEGF splice variants in retinal vascular development is
`being explored. however. in mice expressing only a single variant (Stalmans et al., 2002).
`Vascular development was normal in the retinas of mice expressing only VEGFIM
`( VEGFJM’IM), indicating that this variant is sufficient for directing normal vascular growth
`and remodeling. In contrast, retinas of VEGFJM'?” mice exhibited severe vascular defects.
`displaying retarded venous and severely flawed arterial development. VEGF15’&"1881nice had
`normal development of retinal veins but little or no arterial growth.
`
`Evidence for the expression patterns and roles of VEGFR in retinal tissues comes from a
`variety of species and experimental venues. In the human retina VEGFR-l and -2 can be
`expressed by neural, glial and vascular elements. In adults, expression is generally restricted
`to the inner nuclear laycr (Muller cells and amacrinc cells), the ganglion cell layer, and the
`retinal vasculaturc (Stilt et al.. 1998). However. during retinal neurogcnesis VEGFR-Z is
`also expressed by neural progenitor cells (I—Iashimoto et at. 2006). Notably, neural cell
`VEGFR-2 can be activated by VEGF in Vitro (Yang et al., 1996). In cultured retinal
`pericytcs VEGFR-l, but not -2, is expressed (Takagi et al., 1996), whereas in cultured RPE
`cells, both receptors are expressed and are induced by oxidativc stress (Sreekumar at al.,
`2006). In the mouse, ganglion cells express both receptors, but only VEGFR-2 is increased
`by intraocular inoculation with hcrpesvinis {Vinores et al,, 2001). Studies in newborn mice
`using the VEGFR-spccitic kinasc inhibitor, SU5416, indicate that Muller cell survival or
`proliferation during retinal development is VEGFR- and MAPK-dependent (Robinson et al..
`200 l ). In a study of patients with diabetic retinopathy. VEGFR-I expression dominated in
`normal retina, but was not increased in the diabetic retina, while VEGFR-2 levels were
`increased, pattiCularly in the vaSCular elements (Smith ct at, 1999). Finally, VEGFR-I and
`-2 are found on uterine smooth muscle cells in viva. When these cells are cultured in wire,
`VEGFR-I can be phosphorylated and is capable of inducing smooth muscle cell
`proliferation (Brown et al,, 1997). To date, neither VEGFR-l nor -2 has been identified in
`retinal smooth muscle cells.
`
`This article will review the role of VEGF in angiogencsis related to three blinding
`conditions: retinopathy of prematurity, diabetic retinopathy and age-related macular
`degeneration. These conditions constitute the leading causes of irreversible vision loss in
`infants. and working age and elderly Americans. respectively. That VEGF is believed to
`play a causal role in all three of these disorders underscores its profound impact in eye
`disease. VEGF antagonists have already proven their value in tumor angiogencsis and
`choroidal neovascularization, and new VEGF antagonists are being tested pro-clinically and
`clinically for other ocular indications. Only with a more complete understanding of VEGF
`and its retinal and choroidal activities can we hope to develop better strategies to prevent,
`retard or repair the damage caused by ocular neovasculariyation.
`
`2. Retinopathy of Prematurity
`
`Retinopathy of prematurity (ROP). a neovascularizing disease affecting preterm infants. is
`one of the most conunon causes of childhood blindness in the world. Recent estimates
`
`indicate that each year in the United States, 68% of the approximately 10.000 babies born
`with a birth weight of less than 1250 grants will develop ROP. Thirty six percent of these
`
`ng Refit: Eye Res. Author manuscript; available in PMC 2013 June 14.
`
`Regeneron Exhibit 1055.006
`
`

`

`
`
`tduosnuewJoutnvVd'HlNtduosnuewJOLfll’lVVd‘HlN
`
`
`
`
`
`
`
`
`
`tduosnuewJouinvVd-HIN
`
`Penn et a].
`
`Page 7
`
`infants will progress to severe ROP, a condition that can lead to retinal detachment and
`blindness. The incidence of the disease is highly correlated with the gestational age of the
`infant at birth. That is, the more immature the infant, the higher the likelihood of developing
`severe ROP (CRYO-ROP Cooperative Group, 1988: ETROP Cooperative Group, 2003).
`ROP is caused by perturbation of the process of normal vascular development of the retina.
`Its correlation with gestational age stems from the fact that the retina is one of the last
`organs to be vascularized in the human fetus. in very premature infants. the retina is nearly
`avascular at birth. The process of retinal vascular development normally occurs in the
`hypoxic uterine environment, but it must occur in a relatively hyperoxic extra-uterine
`environment in these infants. For reasons defined in the following paragraphs, this leads to
`arrested growth of retinal blood vessels. followed by their unregulated growth into the
`vitreous cavity, with potentially catastrophic consequences.
`
`2.1 Pathogenesis of ROP
`
`The pathogenesis of ROP is hypothesired to consist of two distinct phases (Madan and
`Penn, 2003). In the initial phase, normal retinal vascular growth is retarded. This occurs as a
`consequence, primarily, of exposure to extra-uterine hyperOxia aggravated by therapeutic
`oxygen delivery- but it may also be due to other noxious stimuli, andfor to the premature
`withdrawal of certain maternally derived factors at the time of birth. Relative retinal hypoxia
`results from the increasing metabolic demands of the developing neural retina that are unmet
`secondary to the attenuation of blood vessels. This leads to the second phase of ROP,
`consisting of the release of VEGF and other angiogenic factors, producing excessive growth
`of abnormal leaky blood vessels into the vitreous, followed by vitreous hemorrhage and
`tractional retinal detachment. This second phase is probably encouraged by the weaning of
`infants from oxygen therapy, but removal from therapy is not required for the neovascular
`response to occm. Understanding the process of normal retinal vasculariration can provide
`important clues regarding the molecular mechanisms underlying the pathogenesis of the two
`phases of ROP.
`
`2.2 Development of the Retinal Vasculature
`
`The process of normal vascularization has been extensively examined in the kitten, mouse
`and rat retinas {Ashton et al., 1953’; Ashton, 1961; Ashton, 1970; Blanks and Johnson. 1983 ;
`Connolly et al., 1988‘, Chan-Ling et al., 1990. Smith et al., I994; Stone et al., I995; Derrell
`and Fricdlander, 2006). Several studies have also examined the process in the human fetal
`retina (Michaelson. 1948; Nilauscn. I958; Cogan. 1963; Ashton, 1970; Nishimura and
`Taniguchi. 19821Penfold et al.. 1990: Gariano et al.. 1994: Hughes et al., 2000).
`Development of the retinal vasculature follows a common pattern in all species (Donell and
`Friedlander, 2006), but there are some dissimilarities as well (Ashton, I968; Gariano et al.,
`1994). For example, in humans the process occurs primarily during the latter half of
`gestation, whereas in rodents the process is completed in the first two weeks after birth
`(Michaelson et al., 1954; Ashton, I970; Stone et al., 19951Engennan and Meyer, I965).
`Generally, the completion of retinal vaswlar development is coincident with eye opening in
`mammalian species.
`
`Retinal tissue is provided with oxygen and nutrients by the adjacent choriocapillary plexus,
`which supplies the avasCular photoreceptor layers. and by the superficial and deep capillary
`plexuses within the retina. supplying its inner layers (Yu and Cringle, 200 l ). The superficial
`plexus lies immediately beneath the inner limiting membrane. while the deep plexus
`permeates the inner nuclear layer. The endothelial cells lining the vessels in the retinal
`capillary plexuses form tight junctions and previde an important part of the blood-retinal
`barrier, while the choroidal vessels are fenestrated, lacking this barrier (Raviola, 197?;
`Campochiaro, 2000). At the choroidfretina interface, barrier function is provided by tight
`
`ng Refill Eye Res. Author manuscript; available in PMC 2013 June 14.
`
`Regeneron Exhibit 1055.007
`
`

`

`
`
`1duosnuewJouinvVd'HlNiduosnuewJOLfll’lVVd‘HlN
`
`
`
`
`
`
`
`
`
`idliosnuewJouinvVd-HIN
`
`Penn et a].
`
`Page 8
`
`junctions between adjacent retinal pigment epithelial cells. Additionally, prior to the
`formation of the retinal vasculature. a transient network of vessels called the hyaloid
`vascular system forms to nourish the immature lens. The vessels of the hyaloid extend from
`their source at the optic nerve head to the posterior surface of the lens, where they bifurcate
`to form the dense capillary arbor known as the tunica vasculosa lentis. This network
`regresses during mid-gestation in the human and two to three weeks post-natally in the
`mouse and rat, coinciding with the period of retinal vascular development (Gogat et al..
`2004; Fruttiger. 2007).
`
`2.2.1 The Superficial Plexus—Retinal astrocytes guide the formation of the retinal
`vasculature in manmlals. Inuuediately prior to the development of the vascular plexus, a
`network of astrocytes radiates from the optic nerve head across the surface of the immature
`retina in a central to peripheral pattern. This network forms the scaffold upon which the
`primary vascular plexus is formed (Ling and Stone, 1988: Fnrttiger et al..1996). Astrocytcs
`express VEGF; which promotes endothelial cell proliferation and migration within this
`superficial plane (Stone et al., 1995). The formation of this primary vascular plexus begins
`in the region around the optic disc at the base of the hyaloid artery. A network of capillaries
`spreads across the developing neural layer along the inner surface of the retina towards the
`periphery. Because of its location. this vessel network is often called the superficial plexus.
`1n rodents. the development of the superficial plexus begins at birth. while in humans the
`process begins at about 16 weeks gestation (Provis, 2001; Saint-Geniez and D’Amore, 2004;
`Dorrell and Friedlander. 2006).
`
`VEGF expression in the retina is closely linked to retina] vascular development. On post-
`natal day zero (P0), intense VEGF expression is noted in the innennost nerve fiber layer in
`the mouse retina. From P0 - P7. expression increases and is greater at the leading edge of
`blood vessel growth (Gariano et al.. 2006). As blood vessel growth proceeds. there is a
`gradual decrease in VEGF expression (Fig. 4). The pattern suggests a strong correlation
`between the presence or absence of physiologically patent blood vessels and the level of
`local VEGF expression.
`
`Studies indicate that development of the retinal vasculature may occur by both
`vasculogenesis, dc mm formation of blood vessels from mesodermal precursor cells
`(angioblasts). and angiogenesis, sprouting of new vessels from existing blood vessels
`(Ashton. 1966: Ashton, 1970'. Flower et al._. 1985'. McLeod et al., 1987; Kretzer and Hittner.
`1988; Chan-Ling etal.. 1990; Jiang et al., 1995‘. Risau and Flamme. 1995; Risau. 1997'.
`Hughes et al., 2000). "Spindle-cells”, so called for their shape. have been identified within
`the inner plexifonn layer of the immature mammalian retina (Flower et al._. 1985', McLeod et
`al., 1987; Stone and Dreher, 1987', Watanabe and Raff, 1988', Hughes et al., 2000; Taomoto
`et al., 2000). These cells are purported to coalesce to fonn vascular cords, which
`subsequently form the patent vessels that constitute the superficial vascular plexus. By a
`process that is not yet completely understood. the vessels then undergo remodeling, leading
`to the development of mature arteries. veins and capillaries. The identification of vascular
`precursor cells in the retinas of various species supports the notion that vasculogenesis
`participates in the formation of the primary plexus (Chan Ling et al., 2004). However. some
`evidence argues in favor of angiogenesis as being the mechanism d

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket