throbber
Article
`
`Ocular Angiogenesis: The Science Behind the Symptoms
`With a variety of factors contributing to neovascularization, many therapies
`are being developed
`
`January 1, 2011
`
`PEER REVIEWED
`
`Ocular Angiogenesis: The Science Behind the Symptoms
`
`With a variety of factors contributing to neovascularization, many therapies
`are being developed.
`
`Matthew Dombrow, MD · Ron A. Adelman, MD, MPH, FACS
`
`With most things in nature, a balance must be struck between two opposing forces, each necessary for a
`functional end product. Enormously complex interactions between production, degradation and reformation are
`necessary for any living organism. The eye, being one of the most complex and evolved organs, must also adhere
`to this delicate balance. Angiogenesis and antiangiogenesis are one of these many pairings that must achieve
`balance. In his groundbreaking 1971 article, Judah Folkman introduced the idea of a “diffusible message” released
`from solid tumor cells to invoke a robust capillary-sprouting response, more robust than ordinary wound healing or
`inflammation, to help support its cancerous growth. In the same light, Folkman introduced the concept of
`1
`antiangiogenesis.
`
`A distinction between vasculogenesis and angiogenesis bears mention. Vasculogenesis is blood-vessel formation
`via endothelial progenitor cells and hemangioblast differentiation, while angiogenesis is the formation of new
`capillaries from pre-existing blood vessels. Angiogenesis requires endothelial cell migration, proliferation, survival,
`vessel maturation, vessel-wall remodeling and degradation of the extracellular matrix. It is a normal part not only of
`development but also of healing. Endothelial cell number is normally stable without significant proliferation, likely
`due to a balance between proangiogenic factors (ie, vascular endothelial growth factor) and antiangiogenic
`(angiostasis) factors (ie, pigment epithelium–derived factor). When there is an imbalance between these two,
`pathology ensues.
`
`Ocular angiogenesis is a major cause of much ocular disease and blindness. It is a significant contributing factor in
`diabetic retinopathy, exudative AMD, corneal graft rejection, corneal neovascularization, retinopathy of prematurity,
`retinal vein occlusion, neovascular glaucoma and sickle cell retinopathy. In this article, different forms of ocular
`angiogenesis, their mediators and implications for treatment will be reviewed.
`
`RETINAL AND CHOROIDAL NEOVASCULARIZATION
`Retinal Neovascularization
`
`Samsung et al. v. Regeneron IPR2023-00884
`Regeneron Pharmaceuticals, Inc. Exhibit 2113 Page 1
`
`

`

`Proliferative diabetic retinopathy (Figures 1-3), RVO and sickle cell proliferative vitreoretinopathy are the most
`common forms of retinal neovascularization. Other causes include familial exudative vitreoretinopathy, sarcoidosis,
`pars planitis, radiation retinopathy, ocular ischemic syndrome and Eales disease. A combination of angiogenesis
`and vasculogenesis occurs during retinal neovascularization. Normally, minimal endothelial cell proliferation occurs
`in the retina. “Hypoxia is believed to be the initial stimulus that causes an upregulation of growth factors, integrins
`2
`and proteinases, which result in endothelial cell proliferation and migration.” Hypoxia upregulates VEGF mRNA in
`3
`retinal endothelial cells, RPE cells, pericytes, Müller cells and ganglion cells.
`4
`
`Figure 1. Severe proliferative diabetic retinopathy with trac-tional retinal detachment.
`
`Samsung et al. v. Regeneron IPR2023-00884
`Regeneron Pharmaceuticals, Inc. Exhibit 2113 Page 2
`
`

`

`Figures 2 and 3. Early phase fluorescein angiography of leaking neovascular membranes and hemorrhage
`in patients with PDR.
`
`Choroidal Neovascularization
`
`Despite alterations in choroidal blood flow in AMD, it may not be enough to produce significant hypoxia to induce
`CNV.
` Like retinal neovascularization, CNV is a combination of angiogenesis and vasculogenesis. CNV is most
`3,5,6
`7
`commonly seen in wet AMD (Figures 4 and 5). Other common causes are high myopia, choroidal rupture, angioid
`streaks, ocular histoplasmosis syndrome, multifocal choroiditis, punctate inner choroidopathy and iatrogenic
`causes (intense laser photocoagulation). Wet AMD is the leading cause of severe vision loss in the elderly in the
`United States. Oxidative damage and inflammation (and less so hypoxia) are thought to tip the pro- and
`8
`antiangiogenic-factor balance in favor of angiogenesis.
`
`Samsung et al. v. Regeneron IPR2023-00884
`Regeneron Pharmaceuticals, Inc. Exhibit 2113 Page 3
`
`

`

`Figures 4 and 5. Fundus photography and mid-phase fluorescein angiography of an active choroidal
`neovascular membrane in a patient with exudative age-related macular degeneration.
`
`INFLAMMATION AND COMPLEMENT
`
`In experimental models, a robust immune response is seen quickly after choroidal neovascular membrane (CNVM)
`is induced by laser. Within 72 hours, there is an influx of neutrophils, macrophages, natural killer cells, microglial
`cells and edema. Macrophages are found near ruptured or thin areas in Bruch's membrane and have been
`9
`isolated from CNVM removed via submacular surgery.
` Activated macrophages secrete collagenase and
`10
`elastase, which may erode Bruch's membrane, allowing for vascular mobilization. Recently, inhibition of vascular
`adhesion protein-1 decreased macrophage accumulation in CNV lesions and reduced CNV size and expression
`11
`of other inflammatory molecules including TNF-α, ICAM-1 and MCP-1.
`11
`
`Samsung et al. v. Regeneron IPR2023-00884
`Regeneron Pharmaceuticals, Inc. Exhibit 2113 Page 4
`
`

`

`Drusen are likely the byproducts of RPE cells with some protein components, possibly arising from the choroid. It
`is still not truly known if drusen are epiphenomena of AMD, an active player in the inflammatory cascade, or a
`passive player via its physical barrier and disruption of transport across Bruch's membrane.
` Shen was able to
`12
`produce a CNV membrane by subretinal injection of Matrigel (BD Biosciences, Sparks, MD), a soluble basement
`membrane preparation mimicking drusen formation.
` In the past decade, noting similarities between drusen found
`13
`in AMD and drusen found in patients with membranoproliferative glomerulonephritis type 2, Hageman and
`colleagues formed the basis of complement system dysfunction and its role in CNVM formation.
` Specifically,
`14-16
`variants of complement factor H (CFH) and alterations in the alternative complement pathway have been
`genetically linked to AMD.
` A myriad of targets of the complement sys tem exist and several phase 1 and 2
`14-17
`trials are ongoing. Potential agents work by either replacing a defective component (ie, CFH with the Y402H
`mutation) or by blocking a complement pathway (C3 and C5 inhibitors).
`18
`
`VEGF
`
`Vascular endothelial growth factor was initially named vascular permeability factor, as it originally was isolated in
`tumor ascites fluid from guinea pigs.
` VEGF is 50,000 times more potent as a vasodilator than histamine.
` It
`19
`20
`includes a family of growth factors — VEGF-A, VEGF-B, VEGF-C, VEGF-D and PlGF (placental growth factor).
`The major mediator of tumor angiogenesis is VEGF-A. Despite a very high affinity between VEGF and VEGF
`receptor 1 (VEGFR-1), most transduction occurs with VEGFR-2. VEGFR-1 may be a “decoy” receptor, preventing
`VEGF binding to VEGFR-2.
` Thus, the interaction of VEGF and VEGFR-2 is crucial.
`3,21
`
`Vascular endothelial growth factor is expressed in most types of human cancers and is highly selective for
`endothelial cells.
` As adapted from Hicklin, VEGF is involved in: (1) endothelial cell proliferation via activation of
`10,22
`mitogen-activated protein kinases; (2) endothelial cell permeability via opening of endothelial fenestrations and cell
`junctions; (3) tumor-cell invasion via induction of metallo-proteinases (MMPs) and urokinase plasminogen
`activators (UPA), hence promoting extracellular matrix degradation; (4) migration through activation of FAK, p38
`and nitric oxide; (5) survival of new endothelial cells by inhibiting apoptosis; and (6) activation and stabilization of
`the vascular network.
`23
`
`Induction of VEGF results from hypoxia via hypoxia-inducible factor 1 (HIF-1), low pH, inflammatory cytokines (IL-
`6), growth factors (basic fibroblast growth factor), sex hormones (androgens and estrogens), chemokines,
`oncogene activation and decreased activity of tumor suppressor gene activity.
` Numerous studies have shown
`22,23
`increased aqueous and vitreous levels of VEGF and VEGFR-1 in a variety of ocular proliferative conditions.
`24 31
`Surgically removed CNVMs from AMD patients show increased VEGF expression in fibroblasts and RPE cells.
`32 34
`
`But is VEGF itself sufficient enough to produce retinal and choroidal neovascularization? VEGF is found in the
`Bruch's membrane–choriocapillaris complex of normal healthy donor eyes.
` Some animal models had difficulty
`10,35
`producing both retinal and choroidal neovascularization. Ozaki implanted intravitreal, sustained-release VEGF
`pellets that induced retinal neovascularization in rabbits but not in primates.
` Tolentino showed that intravitreal
`36
`injections of VEGF in monkey eyes produced capillary nonperfusion and vessel dilation, but preretinal
`neovascularization in only the peripheral retina and not posterior pole.
` Transgenic mice in which rhodopsin
`37
`promoter was coupled to a VEGF gene, hence producing increased intraretinal VEGF, did form intraretinal and
`subretinal neovascularization. However, as Campochiaro has demonstrated, when increased VEGF levels are
`combined with photoreceptor degeneration, CNVM may form. Thus, healthy photoreceptors may somehow prevent
`the choriocapillaris from responding to elevated VEGF levels.
`38,39
`
`Basic FGF
`
`Samsung et al. v. Regeneron IPR2023-00884
`Regeneron Pharmaceuticals, Inc. Exhibit 2113 Page 5
`
`

`

`Fibroblast growth factor (FGF) is a heparin-binding growth factor with a high affinity for heparin sulfate
`proteoglycans. FGF and associated receptor tyrosine kinases (RTKs) are expressed in virtually all cells —
`overexpression plays a role in many cancers.
` A recent question is whether it is truly involved in retinal and
`40
`choroidal neovascularization. Studies have shown increased levels of FGF and FGF-like peptides in ischemic
`retinopathy and laser-induced CNV. Following FGF-impregnated microsphere injection into the subretinal space,
`AMD-like neovascular membranes formed in rabbit eyes.
` However, other studies suggest that FGF is only
`3,41-43
`angiogenic when it accompanies other cell injury, thus unmasking “control mechanisms that sequester FGF2.”
`3,44
`Newer evidence may show that FGF may not, at least alone, be a factor in ocular angiogenesis. FGF2-deficient
`transgenic mice developed the same amount of retinal and choroidal neovascularization as in wild-type mice.
`FGF2 retinal overexpression did not produce retinal neovascularization in mice.
`45,46
`
`IGF-1
`
`The role of insulin-like growth factor (IGF)-1 in angiogenesis was seen as early as 1953, when Poulsen noted
`regression of neovascularization in proliferative diabetic retino- pathy patients after pituitary infarction.
` Pituitary
`47
`ablation was even a treatment for PDR at one time. IGF-1 is elevated in the serum and vitreous in PDR.
`48,49
`Elevated growth hormone/IGF-1 has been implicated in retinal neovascularization in mouse models independent of
`VEGF levels.
` More recently, Hellstrom studied the relationship between IGF-1 and VEGF in retinopathy of
`50
`prematurity. Knock-out IGF-1 mice's normal retinal vascular development was arrested despite adequate VEGF
`levels. Infants with higher levels of IGF-1 had more normal vasculature development. Hellstrom proposed that
`infants with a low level of IGF-1 develop an avascular retina, and in conjunction with subsequent elevated VEGF
`levels, proliferative ROP ensues.
`51
`
`MMP/UPA
`
`Extracellular proteinases are vital for the migration of endothelial cells through the extracellular matrix. Urokinase
`plasminogen activator and the matrix metalloproteinase (MMP) family are primarily involved in angiogenesis.
`MMPs are upregulated in angiogenic lesions and “their inhibition or genetic ablation diminishes angiogenic
`switching, tumor size and growth.”
`3,11,52-54
`
`Prinomostat (AG3340), an MMP inhibitor, was shown to inhibit CNVM growth and leakage when injected
`intravitreally in an animal model, but early human trials studying use for cancers were terminated. It is currently
`being studied for possible ocular use.
` Intravitreal injection of A6, a urokinase system inhibitor, showed promise
`55,56
`in successful treatment of monkey CNVM — up to a 76% reduction in CNVM size was safely observed.
`57
`
`IL-8 AND TNF-α
`
`Interleukin-8 is also thought to play a role in the inflammatory component of ocular neovascularization. It is a
`cytokine that is a chemotactic factor for neutrophils and lymphocytes. IL-8 levels are significantly elevated in the
`vitreous of patients with proliferative neovascularization, including PDR, RVO and Eales disease.
` It is also
`31,58
`thought to aid in the inflammatory component of angiogenesis as shown in rat cornea studies.
` Elner showed that
`59
`stimulated RPE cells synthesized IL-8, thereby possibly augmenting the inflammatory cascade in angiogenesis.
`60
`
` CNV membranes
`Elevated levels of TNF-α have been isolated from fibrovascular membranes in PDR eyes,
`61
`62
`and animal models of hypoxia-induced retinal neovascularization.
` TNF-α antagonists have been successful in
`63
`treating T-cell–mediated noninfectious uveitis and juvenile idiopathic arthritis, but its role in ocular angiogenesis
`has not been well established.
`
`PEDF
`
`Samsung et al. v. Regeneron IPR2023-00884
`Regeneron Pharmaceuticals, Inc. Exhibit 2113 Page 6
`
`

`

`Pigment epithelium–derived factor (PEDF) is believed to be the most potent angiogenesis inhibitor. Hypoxia has
`been shown to downregulate PEDF, thus permitting neovascularization.
` Vitreous samples from PDR and RVO
`64,65
`patients showed lower PEDF levels than in controls, and PDR eyes treated with PRP showed the ability to
`replenish their levels of PEDF.
` Systemic and intravitreal PEDF administration decreases retinal
`31,66
`neovascularization in a hypoxia-induced neovascularization mouse model.
` Mori showed decreased CNV using
`67,68
`an adenoviral vector transfer of PEDF.
` A phase 1 trial of an adenoviral vector delivered intravitreal PEDF
`69-71
`(AdPEDF) completed in 2006 showed stabilization of CNVM in wet AMD patients for up to 12 months.
`72
`
`TGF-β
`
`Transforming growth factor–β's exact role remains somewhat more elusive in angiogenesis. It is believed to be
`both pro- and antiangiogenic, depending on its concentration and surrounding environment. Low levels lead to
`angiogenesis by upregulating angiogenic factors and proteases. High levels stabilize basement membrane
`formation, recruit smooth muscle cells, and inhibit endothelial cell growth.
` Ogata showed TGF-β levels remained
`73
`elevated in photocoagulated RPE cells, while angiogenic factors, including VEGF and IL-8, were reduced.
` In the
`74
`context of cancer and tumor angiogenesis, Bernebeu describes TGF-β's “dual and paradoxical role”; first it acts as
`a tumor suppressor in a premalignant stage, and then its role becomes “exploited” by active tumor cells to aid in its
`own growth. Tumor cells can become resistant to TGF-β via mutating TGF-β receptors, thus reacting as if in a low-
`level TGF-β environment.
`75
`
`Uveal Melanoma
`
`Uveal melanoma cells are one of the few cell types that use a process coined “molecular mimicry” to enhance their
`microcirculation. Folberg and Maniotis found de novo vascular channels that were completely devoid of endothelial
`cells in aggressive melanoma cells.
` This suggested tumor cell ability to form new circulatory channels
`76
`independent of angiogenesis, hence discovering a potential new way to diagnose and treat tumor growth. In
`addition to this novel concept, angiogenesis also plays a role in uveal melanoma's growth. In 1979, Folkman
`implanted aqueous humor aspirated from eyes with uveal melanoma, eyes with retinoblastoma and control eyes
`into the chorioallantoic membrane of chick embryos. The majority of tumor-laden samples produced an angiogenic
`response in the chick embryo, while only one sample from a control eye did (this patient later developed
`lymphocytic leukemia).
`77
`
`Recent reports of VEGF expression in uveal melanoma have been highly variable. Generally, elevated VEGF
`levels have been seen in samples with a necrotic tumor component but do not appear to be related to tumor size
`or presence of metastases.
` This year, bevacizumab (Avastin, Genentech) and gene transfer of PEDF (an
`78-80
`angiogenesis suppressor) decreased uveal melanoma growth and hepatic micrometastases in a mouse
`model.
`81,82
`
`DISCUSSION
`
`In addition to the above factors, several other growth factors, cytokines and environmental conditions are involved
`in the normal and abnormal cascade of angiogenesis. As with any disease process, it is vital to understand the
`pathophysiology in order to better target treatments. With regard to the eye, preventing angiogenesis is
`multifactorial and complex. Despite our lack of full understanding of these diseases (eg, AMD), we have been able
`to halt or at least slow the progression of angiogenesis.
`
`Laser photocoagulation has been the mainstay of treatment for retinal neovascularization. It has been used for
`CNV and then was replaced with photodynamic therapy, which in turn has been largely replaced by anti-VEGF
`injections. Currently, anti-VEGF treatment has taken center stage due to the primary role VEGF has in ocular
`neovascularization. In the past decade, tremendous advancements have been made in the treatment of wet AMD.
`
`Samsung et al. v. Regeneron IPR2023-00884
`Regeneron Pharmaceuticals, Inc. Exhibit 2113 Page 7
`
`

`

`Newer anti-VEGF agents are under investigation, such as aflibercept (VEGF Trap-Eye, Regeneron). It has a
`longer duration of action compared to ranibizumab (Lucentis, Genentech) and thus may allow for a less frequent
`dosing schedule. There are also clinical trials evaluating small-interfering RNA and combination therapy with
`brachytherapy to address VEGF-induced angiogenesis.
`
`Additionally, other targets of angiogenesis are under investigation, such as receptor tyrosine kinase inhibitors to
`target VEGF and PDGF receptors, complement modulators, MMP and UPA inhibitors, and even doxycycline.
`Recently, Cox showed that oral doxycycline, through its anti-inflammatory properties and MMP-suppression
`effects, reduced angiogenesis in CNVM and pterygium mouse models.
` For now, anti-VEGF agents are the
`52
`mainstay for the treatment of choroidal angiogenesis. For retinal angiogenesis, such as in PDR, laser
`photocoagulation and/or anti-VEGF agents may be used.
`
`CONCLUSIONS
`
`A delicate balance must be struck between angiogenesis and its counterpart, angiostasis. Retinal
`neovascularization and CNV may ensue when this balance goes awry. Retinal neovascularization is usually
`initiated in hypoxic states (eg, PDR), whereas CNV is usually a result of aging, chronic inflammation and oxidative
`damage (eg, AMD). VEGF has been strongly implicated in both, but other contributing factors include complement,
`fibroblast growth factor, insulin-like growth factor 1, matrix metalloproteinase, urokinase, interleukin-8, tumor
`necrosis factor-α and pigment epithelium–derived growth factor.
`
`Due to its enormous complexity, there are many pathways to target when treating ocular neovascularization.
`Although not a very elegant treatment, laser photocoagulation has been a successful treatment for retinal
`neovascularization. A bit more refined, targeting VEGF and its resulting molecular cascade of events has shown
`great hope in the treatment of both retinal and choroidal neovascularization. Potentially longer-lasting treatments
`that target more specific pathways may come about in the treatment of ocular neovascularization, thus reducing
`angiogenic-related eye disease and blindness. RP
`
`REFERENCES
`
`1. Folkman J. Tumor Angiogenesis: therapeutic implications. N Engl J Med. 1971;285:1182-1186.
`2. Engerman RL, Pfaffenbach D, Davis MD. Cell turnover of capillaries. Lab Invest. 1967;17:738-43.
`3. Das A, McGuire PG. Retinal and choroidal angiogenesis: pathophysiology and strategies for inhibition. Prog
`Retin Eye Res. 2003;22:721-748.
`4. Aiello L P, Northrup JM, Keyt BA, Takagi H, Iwamoto MA. Hypoxic regulation of vascular endothelial growth
`factor in retinal cells. Arch Ophthalmol. 1995;113:1538-1544.
`5. Grunwald JE, Hariprasad SM, DuPont J, et al. Foveolar choroidal blood flow in age-related macular
`degeneration. Invest Ophthalmol Vis Sci. 1998; 39:385-390.
`6. Campochiaro PA. Retinal and choroidal neovascularization. J Cell Physiol. 2000; 184:301-310.
`7. Sengupta N, Caballero S, Mames RN, Butler JM, Scott EW, Grant MB. The role of adult bone marrow-derived
`stem cells in cho roidal neovascualrization. Invest Ophthalmol Vis Sci. 2003;44:4908-4913.
`8. Age-related macular degeneration: what you should know. National Eye Institute Web site.
`http://www.nei.nih.gov/health/maculardegen/nei_wysk_amd.PDF. Accessed October 31, 2010.
`9. Nusenblatt RB, Ferris F 3rd.. Age-related macular degeneration and the immune response; implications for
`therapy. Am J Ophthalmol. 2007; 144:618-626.
`10. Bressler SB. Introduction: Understanding the role of angiogenesis and antiangiogenic agents in age-related
`macular degeneration. Ophthalmology. 2009; 116(Suppl):S1-S7.
`11. Noda K, She H, Nakazawa T, et al. Vascular adhesion protein-1 blockade suppresses choroidal
`neovascualrization. FASEB J. 2008;22:2928-2935.
`12. Nozaki M, She H, Nakazawa T, et al. Drusen complement components c3a and c5a promote choroidal
`
`Samsung et al. v. Regeneron IPR2023-00884
`Regeneron Pharmaceuticals, Inc. Exhibit 2113 Page 8
`
`

`

`neovascularization. Proc Natl Acad Sci U S A. 2006; 103:2328-2333.
`13. Shen D et al. Exacerbation of retinal degeneration induced by subretinal injection of Matrigel in CCL2/MCP-1
`deficient mice. Paper presented at: Annual meeting of the Association of Research for Vision and Ophthalmology;
`April 30-May 4, 2005; Fort Lauderdale, FL.
`14. Gehrs KM, Jackson JR, Brown EN, Allikmets R, Hageman GS. Complement, age-related macular
`degeneration and a vision of the future. Arch Ophthalmol. 2010;128:349-358.
`15. Hageman GS, Luthert PJ, Victor Chong NH, Johnson LV, Anderson DH, Mullins RF. An integrated hypothesis
`that considers drusen as biomarkers of immune-mediated processes at the RPE-Bruch's membrane in ter face in
`age-related macular degeneration. Prog Retin Eye Res. 2001;20:705-732.
`16. Anderson DH, Mullins R F, Hageman GS, Johnson LV. A role for local inflammation in the formation of drusen
`in the aging eye. Am J Ophthalmol. 2002; 134:411-431.
`17. Haines JL, Hauser MA, Schmidt S, et al. Complement factor H variant increases the risk of age-related
`macular degeneration. Science. 2005; 308:419-421.
`18. Zarbin MA, Rosenfeld PJ. Pathway-based therapies for age-related macular degeneration: an integrated
`survey of emerging treatment alternatives. Retina. 2010;30:1350-1367.
`19. Senger DR, Galli SJ, Dvorak AM, Perruzzi CA, Harvey VS, Dvorak HF. Tumor cells secrete a vascular
`permeability factor that promotes accumulation of ascites fluid. Science. 1983;219:983-985.
`20. Dvorak HF. Vascular permeability factor/vascular endothelial growth factor: a critical cytokine in tumor
`angiogenesis and a potential target for diagnosis and therapy. J Clin Oncol. 2002;20:4368-4380.
`21. Ferrara N, Gerber HP, LeCouter J. The biology of VEGF and its receptors. Nat Med. 2003;9:669-676.
`22. Kerbel RS. Tumor angiogenesis. N Engl J Med. 2008;358:2039-2049.
`23. Hicklin DJ, Ellis LM. Role of vascular endothelial growth factor pathway in tumor growth and angiogenesis. J
`Clin Oncol. 2005;23:1011-1027.
`24. Adamis AP, Miller JW, Bernal MT, et al. Increased vascular endothelial growth factor levels in the vitreous of
`eyes with proliferative diabetic retinopathy. Am J Ophthalmol. 1994;118:445-450.
`25. Aiello L P, Avery RL, Arrigg PG, et al. Vascular endothelial growth factor in ocular fluid of patients with diabetic
`retinopathy and other retinal disorders. N Engl J Med. 1994;331:1480-1487.
`26. Malecaze F, Clamens S, Simorre-Pinatel V, et al. Detection of vascular endothelial growth factor messenger
`RNA and vascular endothelial growth factor-like activity in proliferative diabetic retinopathy. Arch Ophthalmol.
`1994;112:1476-1482.
`27. Pe'er J, Folberg R, Itin A, Gnessin H, Hemo I, Keshet E. Upregulated expression of vascular endothelial growth
`factor in proliferative diabetic retinopathy. Br J Ophthalmol. 1996;80:241-245.
`28. Ambati J, Chalam KV, Chawla DK, et al. Elevated gamma-aminobutyric acid, glutamate, and vascular
`endothelial growth factor levels in the vitreous of patients with proliferative diabetic retinopathy. Arch Ophthalmol.
`1997; 115:1161-1166.
`29. Hattenbach LO, Allers A, Gümbel HO, Scharrer I, Koch FH. Vitreous concentrations of TPA and plasminogen
`activator inhibitor are associated with VEGF in proliferative diabetic vitreoretinoapthy. Retina. 1999;19:383-389.
`30. Matsunaga N, Chikaraishi Y, Izuta H, et al. Role of soluble vascular endothelial growth factor-1 in the vitreous
`in proliferative diabetic retinopathy. Ophthalmology. 2008;115:1916-1922.
`31. Murugeswari P, Shukla D, Rajendran A, Kim R, Namperumalsamy P, Muthukkaruppan V. Proinflammatory
`cytokines and angiogenic and anti-angiogenic factors in vitreous of patients with proliferative retinopathy and eales’
`disease. Retina. 2008;28:817-824.
`32. Amin R, Puklin JE, Frank RN. Growth factor localization in choroidal neovascualr membranes of age-related
`macular degeneration. Invest Ophthalmol Vis Sci. 1994;35:3178-3188.
`33. Frank RN, Amin RH, Eliott D, Puklin JE, Abrams GW. Basic fibroblast growth factor and vascular endothelial
`growth factor are present in epiretinal and choroidal neovascualr membranes. Am J Ophthalmol. 1996;122:393-
`403.
`34. Lopez PF, Sippy BD, Lambert HM, Thach AB, Hinton DR. Transdifferentiated retinal pigment epithelial cells are
`immunoreactive for vascular endothelial growth factor in surgically excised age-related macular degeneration-
`
`Samsung et al. v. Regeneron IPR2023-00884
`Regeneron Pharmaceuticals, Inc. Exhibit 2113 Page 9
`
`

`

`related choroidal neovascular membranes. Invest Ophthalmol Vis Sci. 1996;37:855-868.
`35. Bhutto IA, McLeod DS, Hasegawa T, Kim S Y, Merges C, Tong P, Lutty GA. Pigment epithelium-derived factor
`(PEDF) and vascular endo thelial growth factor (VEGF) in aged human choroids and eyes with age-related
`macular degeneration. Exp Eye Res. 2006;82:99-110.
`36. Ozaki H, Hayashi H, Vinores SA, Moromizato Y, Campochiaro PA, Oshima K. Intravitreal sustained release of
`VEGF causes retinal neovascularization in rabbits and breakdown of the blood-retinal barrier in rabbits and
`primates. Exp Eye Res. 1997;64:505-517.
`37. Tolentino MJ, McLeod DS, Taomoto M, Otsuji T, Adamis AP, Lutty GA. Pathologic features of vascular
`endothelial growth factor-induced retinopathy in the nonhuman primate. Am J Ophthalmol. 2002;133:373-385.
`38. Yamada H, Yamada E, Kwak N, et al. Cell injury unmasks a latent proangiogenic phenotype in mice with
`increased expression of FGF2 in the retina. J Cell Physiol. 2000;185:135-142.
`39. Campochiaro PA. Retinal and choroidal neovascularization. J Cell Physiol. 2000;184:301-310.
`40. Cook KM, Figg WD. Angiogenesis inhibitors: Current strategies and future prospects. CA Cancer J Clin.
`2010;60:222-243.
`41. Zhang NL, Samadani EE, Frank RN. Mitogenesis and retinal pigment epithelial cell antigen expression in the
`rat after krypton laser photocoagulation. Invest Ophthalmol Vis Sci. 1993;34:2412-2424.
`42. Kimura H, Spee C, Sakamoto T, et al. Cellular response in subretinal neovascularization induced by bFGF
`impregnated microspheres. Invest Ophthalmol Vis Sci. 1999;40:524-528.
`43. Sivalingham A, Kenney J, Brown GC, Benson WE, Donoso L. Basic fibroblast growth factor levels in the
`vitreous of patients with proliferative diabetic retinopathy. Arch Ophthalmol. 1990;108:869-872.
`44. Yamada H, Yamada E, Kwak N, et al. Cell injury unmasks a latent proangiogenic phenotype in mice with
`increased expression of FGF2 in the retina. J Cell Physiol. 2000;185:135-142.
`45. Ozaki H, Okamoto N, Ortega S, et al. Basic fibroblast gowth factor is neither necessary nor sufficient for the
`development of retinal neovascularization. Am J Pathol. 1998;153:757-765.
`46. Tobe T et al. Targeted disruption of the FGF2 gene doe not prevent choroidal neovascularization in a murine
`model. Am J Pathol. 1998 Nov;153(5):1641-6.
`47. Poulsen JE. Recovery from retinopathy in a case of diabetes with Simmonds’ disease. Diabetes. 1953 Jan-
`Feb;2(1);7-12.
`48. Merimee TJ et al. Insulin-like growth factors. Studies in diabetics with and without retinopathy. N Engl J Med.
`1983 Sep 1;309(9):527-30
`49. Grant M et al. Insulin-like growth factors in vitreous. Studies in control and diabetic subjects with
`neovascularization. Diabetes. 1986 Apr;35(4);416-20.
`50. Smith LE, Kopchick JJ, Chen W, et al. Essential role of growth hormone in ischemia-induced retinal
`neovascularization. Science. 1997;276;1706-1709.
`51. Hellstrom A, Perruzzi C, Ju M, et al. Low IGF-1 suppresses VEGF-survival signaling in retinal endothelial cells:
`direct correlation with clinical retionopathy of prematurity. Proc Natl Acad Sci U S A. 2001;98;5804-5808.
`52. Cox CA, Amaral J, Salloum R, et al. Doxycycline's effect on ocular angiogenesis: an in vivo analysis.
`Ophthalmology. 2010;117:1782-1791.
`53. Plantner JJ, Smine A, Quinn TAl. Increase in interphotoreceptor matrix gelatinase A (MMP-2) associated with
`age-related macular degeneration. Exp Eye Res 1998;67:637-645.
`54. Bergers G, Brekken R, McMahon G, et al. Matrix metalloproteinase-9 triggers the angiogenic switch during
`carcinogenesis. Nat Cell Biol. 2000;2:737-744.
`55. El Bradey M, Cheng L, Bartsch DU, et al. Preventive versus treatment effect of Ag3340, a potent matrix
`metalloproteinase inhibitor in a rat model of choroidal neovascularization. J Ocular Pharm Ther. June;20:217-236.
`56. Blodi BA. AG3340 Study Group. Effects of prinomastat (AG3340), an angiogenesis inhibitor, in patients with
`subfoveal choroidal neovascularization associated with age-related macular degeneration. Invest Ophthalmol Vis
`Sci. 2001 42:S311.
`57. Koh HJ, Freeman WR, Azen S P, et al. Effect of a novel octapeptide urokinase fragment, A6, on experimental
`choroidal neovascularization in the monkey. Retina. 2006;26:202-209.
`
`Samsung et al. v. Regeneron IPR2023-00884
`Regeneron Pharmaceuticals, Inc. Exhibit 2113 Page 10
`
`

`

`58. Yoshida A, Yoshida S, Khalil AK, Ishibashi T, Inomata H. Role of NF-kappaB-mediated interleukin-8 expression
`in intraocular neovascularization. Invest Ophthalmol Vis Sci. 1998;39:1097-1106.
`59. Koch AE, Polverini PJ, Kunkel SL, et al. Interleukin-8 as a macrophage-derived mediator of angiogenesis.
`Science. 1992;258:1798-1801.
`60. Elner VM, Strieter RM, Elner SG, Baggiolini M, Lindley I, Kunkel SL. Neutrophil chemotactic factor (IL-8) gene
`expression by cytokine-treated retinal pigment epithelial cells. Am J Pathol. 1990;136:745-750.
`61. Limb GA, Chignell AH, Green W, LeRoy F, Dumonde DC. Distribution of TNF alpha and its reactive vascular
`adhesion molecules in fibrovascular membranes of proliferative diabetic retinopathy. Br J Ophthalmol.
`1996;80:168-173.
`62. Oh H, Takagi H, Takagi C, et al. The potential angiogenic role of macrophages in the formation of choroidal
`neovascular membranes. Invest Ohpthalmol Vis Sci. 1999;40:1891-1898.
`63. Majka S, McGuire PG, Das A. Regulation of matrix metalloproteinase expression by tumor necrosis factor in a
`murine model of retinal neovascularization. Invest Ophthalmol Vis Sci. 2002;43:260-266.
`64. Lange J, Yafai Y, Reichenbach A, Wiedemann P, Eichler W. Regulation of pigment epithelium-derived factor
`production and release by retinal glial (Muller) cells under hypoxia. Invest Ophthalmol Vis Sci. 2008;49:5161-5167.
`65. Dawson DW, Volpert OV, Gillis P, et al. Pigment epithelium-derived factor: a potent inhibitor of angiogenesis.
`Science. 1999;285:245-248.
`66. Spranger J, Osterhoff M, Reimann M, et al. Loss of antiangiogenic pigment epithelium-derived factor in
`patients with angiogenic eye disease. Diabetes. 2001;50:2641-2645.
`67. Stellmach V, Crawford SE, Zhou W, Bouck N. Prevention of ischemia-induced retinopathy by the natural ocular
`antiangiogenic agent pigment epithelium-derived factor. Proc Natl Acad Sci U S A. 2001;98:2593-2597.
`68. Duh EJ, Yang HS, Suzuma I, et al. Pigment epithelium-derived factor suppresses ischemia-induced retinal
`

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