throbber
g#- 'Ns,?i NIH Public Access
`1<it?)eJ Author Manuscript
`~~#
`l -------------------------
`Pub Ii shed in final edited fonn as:
`ProgRetinEyeRes. 2008 July~ 27(4): 331-371. doi:10.1016/j.preteyeres.2008.05.001.
`
`Vascular Endothelial Growth Factor in Eye Disease
`
`J.S. Penna, A. Madanb, R.B. Caldwellc, M. Bartolic, R.W. Caldwellc, and M.E. Hartnettd
`avanderbilt University School of Medicine, Nashville, TN
`
`bStanford University School of Medicine, Palo Alto, CA
`
`cMedical College of Georgia, Augusta , GA
`
`dUniversity of North Carolina School of Medicine, Chapel Hill, NC
`
`Abstract
`Collectjvely, angiogenic ocular conditions represent the leading cause of irreversible vision loss in
`developed countries. In the U.S., for example, retinopathy of prematurity, diabetjc retinopathy and
`age-related macular degeneration are the principal causes of blindness in the infant, working age
`and elderly populations, respectively. Evidence suggests that vascular endothelial growth factor
`(VEGF), a 40 k:Da dimeric glycoprotein, promotes angiogenesis in each of these conditions,
`making it a highly significant therapeutic target. However, VEGF is pleiotropic, 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 mjgration, proteolysis, cell survival and vessel penneability in a
`wide variety of biological contexts. This article will describe the roles played by VEGF in the
`pathogenesis of retinopathy of prematurity, diabetic retinopathy and age-related macular
`degeneration. The potential disadvantages of inhibiting VEGF will be discussed, as will the
`rationales for targeting other VEGF-related modulators of angiogenesis.
`
`z
`:c
`I
`""'C
`)>
`)>
`C .....
`:::T
`0 -,
`
`z
`:c I
`""'C
`)>
`)>
`C .....
`:::T
`0 -,
`s:
`
`ll)
`::J
`C
`(/J
`
`(') -, -5·
`.....
`
`z
`:c
`I
`""'C
`)>
`)>
`C .....
`:::T
`0 -,
`s:
`
`ll)
`::J
`C
`(/J
`(') -,
`~
`
`Keywords
`retina; angiogenesis; vascular endothelial growth factor; age-related macular degeneration;
`diabetic retinopathy; retinopathy of prematurity
`
`1. Introduction
`1.1 Vascular Endothelial Growth Factor (VEGF)
`Vascular endothelial growth factor (VEGF), a dimeric glycoprotein of approximately 40
`kDa, is a potent, endothelial cell mitogen tJ1at stimulates proliferation, migration and tube
`fonnation leading to angiogenic growth of new blood vessels. It is essential for angiogenesis
`during development; the deletion of a single allele arrests angiogenesis and causes
`embryonic 1.etliality (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 growtl1 factor) (Fig. IA). Alternative
`
`© 2008 Elsevier Ltd. All rights reserved.
`Corresponding auihor: John S. Penn, Ph.D., Vanderbilt Eye Institute, 8016 Medical Center East, North Tower, Vanderbilt University
`School of Medicine, Nashville, 1N 37232-8808.
`Publisbet·'s Disclaimer: 111is is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our
`customers we are providing this early version of the manuscript. 111e manuscript will undergo copyediting, typesetting, and review of
`the resulting proof before it is published in its final c itable fom1. Please note that during the production process errors may be
`discovered whi ch could affect the content, and all legal disclaimers that apply to the journal pertain.
`
`Regeneron Exhibit 1055.001
`
`

`

`z
`I
`I
`-0
`)>
`)>
`
`C --::r
`
`0 -,
`
`z
`I
`I
`-0
`)>
`)>
`
`C --::r
`0 -, s:
`
`D.>
`::,
`C
`CJ)
`() -,
`
`-5· --
`
`z
`I
`I
`-0
`)>
`)>
`
`C --::r
`0 -, s:
`
`D.>
`::,
`C
`CJ)
`() -,
`
`-5· --
`
`Penn et al.
`
`Page2
`
`splicing results in several VEGF variants. In humans, these include the relatively abundant
`VEGF121, VEGF16s, VEGF189 and VEGF206, and several less abundant forms (Fig. lB).
`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 e>..1racellular matrix. VEGF J6S 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 confinn that the relative solubility of VEGF splice
`variants strongly affects their specific bioactivities (Taka11ash.i and Shibuya, 2005).
`Moreover, plasmin and various metalloproteinases can cleave VEGF l6S, resulting in an N(cid:173)
`terminal 113-amino 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 isoforms tmder normal or pathological conditions and the molecular
`regulators of VEGF alternative splicing is relatively limited.
`
`Recently, discovery of the so called " VEGFxX,'Cb isoforms" has sparked new interest in the
`molecular events iliat regulate VEGF expression (for review, see Ladomery et al., 2007).
`The VEGFxx.,'<b isofonus share approxin1ately 94-98% homolo!,ry with the corresponding
`VEGF>,,'XX isoforms and have the same length. However, due to alterations in the C(cid:173)
`terminus they bind to VEGF receptors, but do not fully activate tllem and act as " dominant
`negative splice variants" (Bates et al., 2002). Studies showing that VEGF l6Sb is
`downregulated in angiogenic tissues (Ladomery et al., 2007) suggest a primary role for this
`isofonn in controlling VEGF activity in health and disease. Administrntion 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-1/Flt-l (ftns-like tyrosine kinase) and VEGFR-2/KDR/Flk-l (kinase insert domain(cid:173)
`containing receptor/fetal liver kinase), along witl1 structurally related receptors, Flt-3/Flk-2
`and VEGFR-3/Flt-4, belong to the receptor tyrosine kinase family (Fig. IA) (Hanks and
`Quinn, 1991; Blume-Jensen and Hunter, 2001). VEGFR-1 and -2 are primarily involved in
`angiogenesis, (Yancopoulos et al., 2000) whereas Flt-3 and Flt-4 are involved in
`hematopoiesis and ly mphangiogenesis (Jussila and Alitalo, 2000). The VEGFRs contain an
`approximately 750-amino-acid-residue e>..tracellular domain, which is organized into seven
`i1mnunoglobulin-like folds. Adjacent to the e>..tracellular domain is a single tnmsmembrane
`region, followed by aju>..1an1e1nbrane domain, a split tyrosine-kinase domain that is
`interrupted by a 70-amino-acid kinase insert, and a C-tenninal tail.
`
`VEGF receptor activation requires dimerization. Guided by the binding properties of tl1e
`ligands, VEGFRs fonn both homodimers and heterodimers (Rahimi, 2006). The signal
`transduction properties of the VEG FR heterodimers, compared with homodimers, remain to
`be fully elucidated. Dime1ization of VEGFR is accompanied by activation of receptor kinase
`activity, leading to autophosphorylation. Site-directed mutagenesis studies have
`demonstrated tliat the Tyrl214 residue, located in the crubo>..-y tenninus of VEGFR-2, is
`required for the ligand-dependent autophosphorylation of tl1e receptor and its ability to
`activate signaling proteins. Signal trdilSduction is propagated when activated VEGF
`receptors phosphorylate SH2 domain-containing protein substrates.
`
`In addition to VEGFRs, VEGF serves as a ligand to anotller family of receptors, the
`neuropilins. Neuropilins are 120- to 130-kDa non-tyrosine kinase receptors that mediate
`critical functions in tumor cells and the nervous and vascular systems. In endotl1elial cells,
`neuropilins serve as receptors for the class 3 semaphorins and co-receptors for VEGF family
`members. The role ofNeuropilin-1 (NRP-1) in mediating VEGF activity is now being
`
`ProgRelii1 Eye Res. Author manuscript; availabl e in PMC 2013 .lune 14.
`
`Regeneron Exhibit 1055.002
`
`

`

`z
`I
`I
`-0
`)>
`)>
`
`C --::r
`
`0 -,
`
`z
`I
`I
`-0
`)>
`)>
`
`C --::r
`0 -, s:
`
`D.>
`::,
`C
`CJ)
`() -,
`
`-5· --
`
`z
`I
`I
`-0
`)>
`)>
`
`C --::r
`0 -, s:
`
`D.>
`::,
`C
`CJ)
`() -,
`
`-5· --
`
`Penn et al.
`
`Page 3
`
`elucidated. VEGF signaling through NRP-1 stimulates endothelial cell migration and
`adhesion. The addition of an anti-NRP-1 antibody suppressed the mitogenic effects of
`VEGF 165 on bovine retinal endothelial cells (RECs) (Oh et al., 2002). In another il1 vitro
`model, the VEGF-dependent differentiation of a subset of lmman bone marrow-derived cells
`into vascular precursors, and subsequent prolifenition of these cells, required the activation
`of a VEGFR-2/NRP-l-dependent signaling pathway (Fons et al., 2004). Finally, VEGF
`promotion of the synthesis and release of prostacyclin (PGI2), an important mediator of
`angiogenesis, is thought to be mediated via NRP-1 binding (Neagoe et al., 2005). The
`angiogenic effects regulated tlrrough VEGF binding to NRP-2 are less well chanicterized
`and appear to be modulated differently from tl1e effects controlled by NRP-1. For example,
`VEGF selectively up-regulates NRP-1, but not NRP-2, on endoilielial cells (Oh et al., 2002).
`
`BIAcore analysis has shown NRP-1 to intenict wiili VEGFR-1, greatly reducing its binding
`affinity for VEGF 165 (Fuh et al., 2000). Co-culture systems of endotl1elial cells and breast
`carcinoma cells indicate that NRP-1 significantly enhances VEGF l6S binding to VEGFR-2
`(Soker et al., 2002). In aortic endotl1elial cells, NRP-2 interacted with VEGFR-1, but less is
`known at present about how this influences VEGF bioactivity (Gluzman-Poltordk et al ,
`2001). Finally, using multiple in vitJv systems, NRP-2 was shown to intenict witl1
`VEGFR-3, leading to lymphangiogenic activity, but no internction was seen between NRP-2
`and VEGFR-2 (Karp~men et al., 2006).
`
`1.3 VEGF Signaling
`
`Few SH2 domain-containing proteins have been shown to interact directly with VEGFR-2.
`Phospholipase C-y (PLC-y) binds to phosphorylated Tyrll75 (Tyrll73 in tl1e mouse), and
`mediates the activation of tl1e mitogen-activated protein kinase (MAPK) cascade, leading to
`proliferation of endotl1elial cells (Takal1ashi et al, 2001) (Fig. 2). PLCy activates protein
`kinase C via tl1e production of diacylglycerol and increased concentmtions of int:r'dcellular
`calcium. A Tyr l l 73Phe mutation of VEGFR-2 causes embryonic lethality due to vascular
`defects, mimicking tl1e defects of VEGFR-r 1- mice (Sakurai et al., 2005). These data
`demonstrate an essential function of the Tyr 1173 residue du.ring vascular development.
`
`In addition to PLCy, the adaptor molecule, Shb, also binds to phosphory lated Tyr 117 5.
`VEGF-induced migmtion and PI3K activation is inhibited by small interfering RNA
`(siRNA)-mediated knockdown of Shb in endotl1elial cells (Holmqvist et al., 2004). The
`serine/threonine kinase, Akt, is activated downstream of PI3K and mediates endothelial cell
`survival (Fujio and Walsh, 1999). Akt also regulates nitric oxide (NO) production by direct
`phosphorylation and activation of endothelial NO synthase (eNOS). Finally, phosphorylated
`Tyrl 175 is known to interact witl1 Sek (lgarashi et al., 1998; Sakai et al., 2000), an adaptor
`molecule that binds Grb2, and participates in MAPK signaling in tl1e epidennal growth
`factor pathway (Thelemann et al., 2005).
`
`Another important phosphorylation site in VEGFR-2 is Tyr95 l (Tyr949 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 growtll in vRAP-/- mice confirm an essential function for
`iliis residue in endothelial cells of tl1e angiogenic phenotype (Matsumoto et al., 2005).
`VEGF induces the fonnation 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 a Tyrl212Phe (corresponding to the human Tyrl214) VEGFR-2 mutant
`are viable and fertile (Sakurai et al., 2005). However, phosphorylation ofTyrl212/I214 has
`
`ProgRelii1 Eye Res. Author manuscript; available in PMC 2013 .lune 14.
`
`Regeneron Exhibit 1055.003
`
`

`

`z
`I
`I
`-0
`)>
`)>
`
`C --::r
`
`0 -,
`
`z
`I
`I
`-0
`)>
`)>
`
`C --::r
`0 -, s:
`
`D.>
`::,
`C
`CJ)
`() -,
`
`-5· --
`
`z
`I
`I
`-0
`)>
`)>
`
`C --::r
`0 -, s:
`
`D.>
`::,
`C
`CJ)
`() -,
`
`-5· --
`
`Penn et al.
`
`Page 4
`
`been implicated in VEGF-induced actin remodeling through the sequential activation of
`CDC42 and p38 MAPK (Lamalice et al., 2004). lnhibition of p38 MAPK augments VEGF(cid:173)
`induced angiogenesis in the chicken chorioallantoic membmne (CAM) assay (lssbmcker et
`al., 2003 ; Matsumoto 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 re!,'lllates VEGF(cid:173)
`induced actin reorganization and migration (McMullen et al., 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 si!,•naling. 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 seveml different mechanisms (Dor et
`al., 2001). These mechanisms include increased transcription, rnRNA stability, and protein
`translation using an internal ribosomal entry site, as well as increased expression of ox.-ygen
`regulated protein 150, a chaperone required for intracellular tnmsport of proteins from the
`endoplasmic reticulum to the Golgi apparatus prior to secretion (Chen and Shyu, 1995;
`Forsythe et al., 1996; Levy et al., 1996; Levy et al., 1998; Ozawa et al., 2001).
`
`The increase in VEGF trc:lllscription is largely mediated via hypoxia inducible factor-1
`(HIF-1) (Fig. 3). HIF-1 is a heterodirneric transcription factor composed of two subunits -
`the constitutively produced HIF-1[3 subunit and the inducible component, HIF-la. (Wang
`and Semenza, 1995). Under normoxic conditions, HIF-la. is inactivated and targeted for
`proteasomal degradation by hydrox.-ylation, whereas under hypoxic conditions the specific
`hydrox.-ylases are inhibited, resulting in the rescue ofHIF-la. from degmdation (Schofield
`and Ratcliffe, 2004). When this occurs, HIF-la. complexes with HIF-1[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) (Ikeda et al., 1995; Laughner et al., 2001; Shima et al.,
`1996; Wenger, 2002; Forsythe et al., 1996). The in1portance of interaction between HIF-la.
`and the VEGF promoter has been confinned in studies of HIF-la. - /- mouse embryonic stem
`cells, in which basal expression of VEGF mRNA remains low in response to hypoxia
`(Canueliet et al., 1998; Iyer et al., 1998).
`
`Two additional isoforms of HIF, known as HIF-2a. and HIF-3a., have been identified by
`screening for proteins that associate with HIF-1[3 (Ratcliffe, 2007). HIF-2a. appears to be
`closely related to HIF-la. and can promote HRE-dependent gene transcription. While
`structurally and fimctionally sintilar, HIF-la. and HIF-2a. appear to exert different biological
`functions, as demonstrated in studies using knockout mice (Hu et al., 2003). For example,
`while HIF-la. antagonizes c-Myc function, inhibiting renal cell carcinoma (RCC) growth,
`HIF-2a. promotes cell cycle progression in hypoxic RCC and many other cell lines (Gordan
`et al., 2007). Interestingly, the most distantly related isofonn, HIF-3a., appears to antagonize
`HRE-dependent gene expression, suggesting a possible negative influence on hypoxia(cid:173)
`induced gene expression. Additional study is needed to detennine if HIF-2a. or HIF-3a. is
`involved in the regulation of retinal VEGF ex.'Pression.
`
`Clearly, post-transcriptional events are also important in the regulation ofVEGF production
`in tl1e diseased retina, as underscored by tl1e correlation of polymorphisms within the 5' -
`untranslated region (UTR) of t11e VEGF gene with the occurrence of age-related macular
`degeneration (AMD). The 3'UTR and t11e 5'UTR of the VEGF gene are important sites of
`
`ProgRelii1 Eye Res. Author manuscript; availabl e in PMC 201 3 .lune 14.
`
`Regeneron Exhibit 1055.004
`
`

`

`z
`I
`I
`-0
`)>
`)>
`
`C --::r
`
`0 -,
`
`z
`I
`I
`-0
`)>
`)>
`
`C --::r
`0 -, s:
`
`D.>
`::,
`C
`CJ)
`() -,
`
`-5· --
`
`z
`I
`I
`-0
`)>
`)>
`
`C --::r
`0 -, s:
`
`D.>
`::,
`C
`CJ)
`() -,
`
`-5· --
`
`Penn et al.
`
`Page 5
`
`regulation controlling mRNA 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 mRNA stability in response to hypoxia comes from i11
`v1t1v mRNA degradation assays that have led to the identification of adenylate/uridylate-rich
`elements (AREs) in the 3 1 UTR of VEGF mRNA. YEGF mRNA is extremely labile in
`nom10xic conditions, with a half-life of less than 1 hour, as compared with the average half(cid:173)
`life of 10 to 12 hours for eukaryotic mRNA. During hypoxia, the half-life of VEGF mRNA
`increases by two to three-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
`rnRNA, protecting it from degradation by endonucleases (Brennan and Steitz, 2001 ;
`Robinow et al., 1988).
`
`Post-transcriptional regulation can also occur in tJ1e in 51 -UTR of VEGF 1nRNA. This
`region contains multiple IRES. These are specific sites of attachment to the ribosomal
`machinery, which provide sites for initiation of translation alternative to the classical 5' cap(cid:173)
`and elF-dependent translational system (for review, see van der Velden and Thomas, 1999).
`Several IRES have been identified in the 5' -UTR of YEGF mRNA, and these provide
`alternative sites of translational control ofVEGF e>.'J)ression (Bomes et al , 2004). Notably,
`evidence suggests that IRES sites in the VEGF 5' -UTR can potentially control tl1e
`generation of alternatively spliced VEGF (Bornes et al., 2004; Huez et al., 2001 ).
`
`AnotJ1er regulatory mechanism consists of increased production of oxygen regulated protein
`150 (ORPI50) in response to hypoxia. Studies using human macrophages transfected with
`adenovirus coding for ORPl 50 showed that overexpression of ORP150 resulted in increased
`VEGF secretion in hypoxia. Evidence suggests that under hypoxic conditions, ORP150
`functions as a molecular chaperone to facilitate YEGF protein transport and secretion
`(Ozawa et al., 2001). VEGF is not only regulated by hypoxia.
`
`VEGF function is also affected by insulin like growtl1 factor 1 (IGF-1) which plays an
`important role in retinal vasculariz.ation. Several lines of evidence, including in vitro studies,
`support the notion tJ1at IGF-1 is critical for vessel development (King et al, 1985; Grant et
`al., 1993). Pretenn infants with reduced serum levels ofTGF-1 have a higher incidence of
`development of retinopathy (Hellstrom et al., 2003). Mice null for the IGF-1 gene have
`retarded retinal vascular growth, compared to wild type controls (Hellstrom et al., 2001).
`However, the action ofIGF-1 is not mediated by decreasing VEGF expression, as the
`amount ofVEGF mRNA is si1nilar in knock out and wild type control mice; instead IGF-1
`acts by decreasing YEGF activation of tl1e Akt signaling pathway. Both MAPK and A.kt
`pathways have been shown to be necessary for endothelial cell survival (Smith et al., 1999).
`
`1.5 Retinal Expression of VEGF and VEG FR
`
`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 conunon 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 inl1erited
`retinal dystrophies (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 YEGF. These include
`the retinal pigmented epithelium (RPE) (Miller et al., 1997), astrocytes (Stone et al., 1995),
`Mi.iller cells (Robbins et al., 1997), vascular endothelium (Aiello et al., 1995) and ganglion
`
`ProgRelii1 Eye Res. Author manuscript; available in PMC 2013 .lune 14.
`
`Regeneron Exhibit 1055.005
`
`

`

`z
`I
`I
`-0
`)>
`)>
`
`C --::r
`
`0 -,
`
`z
`I
`I
`-0
`)>
`)>
`
`C --::r
`0 -, s:
`
`D.>
`::,
`C
`CJ)
`() -,
`
`-5· --
`
`z
`I
`I
`-0
`)>
`)>
`
`C --::r
`0 -,
`s:
`
`D.>
`::,
`C
`CJ)
`() -,
`
`-5· --
`
`Penn et al.
`
`Page 6
`
`cells (Ida et al., 2003). These cells differ widely in their responses to hypoxia; i11 vitro
`studies show that Miiller cells and astrocytes generally produce the !,>reatest 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 throughout 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 nonnal in the retinas of mice expressing only VEGF !64
`( VEGF1641164), indicating that this variant is sufficient for directing nonnal vascular growth
`and remodeling. In contrast, retinas of VEGF1201120 mice exhibited severe vascular defects,
`displaying retarded venous and severely flawed arterial development. VEGF1881188 mice had
`normal development of retinal veins but little or no arterial !,>rowth.
`
`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
`e:x-pressed by neural, glial and vascular elements. In adults, expression is generally restricted
`to the inner nuclear layer (Miiller cells and amacrine cells), the ganglion cell layer, and the
`relinal vasculature (Stitt et al. , 1998). However, during retinal neurogenesis VEGFR-2 is
`also expressed by neural progenitor cells (Hashimoto et al. , 2006). Notably, neural cell
`VEGFR-2 can be activated by VEGF iI1 vitro (Yang et al., 1996). In cultured retinal
`pericytes VEGFR-1 , but not -2, is expressed (Takagi et al. , 1996), whereas in cultured RPE
`cells, both receptors are expressed and are induced by oxidative stress (Sreekumar at al.,
`2006). In the mouse, ganglion cells express both receptors, but only VEGFR-2 is increased
`by intraocular inoculation with herpesvirus (Vinores et al , 2001). Studies in newborn mice
`using the VEGFR-specific kinase inhibitor, SU5416, indicate that Millier 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-1 expression dominated in
`nonnal retina, but was not increased in the diabetic retina, while VEGFR-2 levels were
`increased, particularly in the vascular elements (Smith et al., 1999). Finally, VEGFR-1 and
`-2 are found on uterine smooth muscle cells in vivo. When these cells are cultured in vitro,
`VEGFR-1 can be phosphorylated and is capable of inducing smooth muscle cell
`proliferation (Brown et al., 1997). To date, neither VEGFR-1 nor -2 has been identified in
`retinal smooth muscle cells.
`
`This article will review the role of VEGF in angiogenesis related to three blinding
`conditions: retinopathy of prematurity, diabetic retinopathy and age-related macular
`degeneration. These conditions constitute Lhe 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 p rofound impact in eye
`disease. VEGF antagonists have already proven their value in tumor angiogenesis and
`cho.roidal neovascularization, and new VEGF antagonists are being tested pre-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 neovascularization.
`
`2. Retinopathy of Prematurity
`
`Retinopathy of prematurity (ROP), a neovascularizing disease affecting preterm infants, is
`one of the most common 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 grams will develop ROP. Thirty six percent of these
`
`ProgRelii1 Eye Res. Author manuscript; available in PMC 2013 .lune 14.
`
`Regeneron Exhibit 1055.006
`
`

`

`z
`I
`I
`-0
`)>
`)>
`
`C --::r
`
`0 -,
`
`z
`I
`I
`-0
`)>
`)>
`
`C --::r
`0 -, s:
`
`D.>
`::,
`C
`CJ)
`() -,
`
`-5· --
`
`z
`I
`I
`-0
`)>
`)>
`
`C --::r
`0 -, s:
`
`D.>
`::,
`C
`CJ)
`() -,
`
`-5· --
`
`Penn et al.
`
`Page7
`
`infants will progress to severe ROP, a condition that can lead to retinal detaclunent 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 nonnally occurs in the
`hypoxic uterine environment, but it must occur in a relatively hyperoxic e>.'tra-uterine
`environment in these infants. For reasons defined in the following paragraphs, this leads to
`arrested growth of retinal blood vessels, followed by their unregtllated growth into the
`vitreous cavity, with potentially catastrophic consequences.
`
`2.1 Pathogenesis of ROP
`
`The pathogenesis of ROP is hypothesized to consist of two distinct phases (Madan and
`Penn, 2003). In the initial phase, nonnal 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, and/or to the premature
`withdrawal of certain maternally derived factors at tJ1e time of birt11. Relative retinal hypoxia
`results from the increasing metabolic demands of the developing neural retina that are unmet
`secondal)1 to the attenuation of blood vessels. This leads to the second phase ofROP,
`consisting of the release of VEGF and other angiogenic factors, producing excessive growth
`of abnonnal 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 occur. Understanding the process of nonnal retinal vascularization can provide
`important clues regarding the molecular mechanisms underlying the pathogenesis of the two
`phases ofROP.
`
`2.2 Development of the Retinal Vasculature
`The process of nonnal vascularization has been extensively examined in the kitten, mouse
`and rat retinas (Ashton et al., 1957; Ashton, 1961; Ashton, 1970; Blanks and Johnson, 1983;
`Connolly et al., 1988; Chan-Ling et al., 1990; Smith et al, 1994; Stone et al., 1995; DorrelJ
`and Friedlander, 2006). Several studies have also examined the process in the human fetal
`relina (Michaelson, 1948: Nilausen, 1958; Cogan, 1963; Ashton, 1970; Nishimura and
`Taniguchi, 1982; Penfold et al.. 1990; Gariano et al.. 1994: Hughes et al., 2000).
`Development of the retinal vasculature follows a common pattern. in all species (Dorre!J and
`Friedlander, 2006), but there are some dissimilarities as well (Ashton, 1968; 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, 1970; Stone et aJ., 1995; Engem1an and Meyer, 1965).
`Generally, the completion of retinal vascular development is coincident witJ1 eye opening in
`mammalian species.
`
`Retinal tissue is provided witl1 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, 2001). The superficial
`plexus lies immediately beneath the inner limiting membrane, while t11e deep plexus
`penneates the inner nuclear layer. The endothelial cells lining tl1e vessels in tJ1e retinal
`capillary plexuses fonn tight junctions and provide an important part of the blood-retinal
`barrier, while the choroidal vessels are fenestrated, lacking this barrier (Raviola, 1977;
`Campochiaro, 2000). At the choroid/retina interface, barrier function is provided by tight
`
`ProgRelii1 Eye Res. Author manuscript; availabl e in PMC 2013 .lune 14.
`
`Regeneron Exhibit 1055.007
`
`

`

`z
`I
`I
`-0
`)>
`)>
`
`C --::r
`
`0 -,
`
`z
`I
`I
`-0
`)>
`)>
`
`C --::r
`0 -, s:
`
`D.>
`::,
`C
`CJ)
`() -,
`
`-5· --
`
`z
`I
`I
`-0
`)>
`)>
`
`C --::r
`0 -,
`s:
`
`D.>
`::,
`C
`CJ)
`() -,
`
`-5· --
`
`Penn et al.
`
`Page 8
`
`junctions between adjacent retinal pi1:,11nent epithelial cells. Additionally, prior to the
`fonnation of the retinal vasculature, a trnnsient network of vessels called the hyaloid
`vascular system forms to nourish the immature lens. The vessels of the hyaloid ex1:end from
`their source at the optic nerve head to the posterior surface of the lens, where they bifurcate
`to fonn the dense capillary arbor known as the ttmica 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 I:,'llide the formation of the retinal
`vasculature in manunals. Inuuediately prior to the development of the vascular plex'Us, a
`network of astrocytes rndiates from the optic nerve head across the surface of the imma11.1re
`retina in a central to peripheral pattern. This network forms the scaffold upon which the
`primary vascular plex11s is fom1ed (Ling and Stone, 1988; Fmttiger et al., 1996). Astrocytes
`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.
`In rodents, the development of the superficial ple,ms begins at birth, while in humans the
`process begins at about 16 weeks gestation (Provis, 200 l ; Saint-Geniez and D ' Amore, 2004;
`Dorrell and Friedlander, 2006).
`
`VEGF expression in the retina is closely linked to retinal vascular development. On post(cid:173)
`natal day zero (PO), intense VEGF expression is noted in the innennost nerve fiber layer in
`the mouse retina. From PO - P7, expression increases and is i:,rreater at the leading edge of
`blood vessel growth (Gariano et al., 2006). As blood vessel growth proceeds, there is a
`grndual decrease in VEGF expression (Fig. 4). Th

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