throbber
Quan Dong Nguyen
`Eduardo Buchele Rodrigues
`Michel Eid Farah
`AARVoemmaMe\yNCCay
`
`nvaahel
`
`SPECeet
`
`Page 01 of 19
`
`SAUNDERS
`
`ELSEVIER
`
`Exhibit 2084
`
`€ e
`
`Mylan v. Regeneron, IPR2021-00880
`U.S. Pat. 9,669,069, Exhibit 2084
`
`Exhibit 2084
`Page 01 of 19
`
`

`

`
`
`Commissioning Editor: Russell Gabbedy
`Development Editor: Alexandra Mortimer
`Editorial Assistant: Poppy Garraway
`Project Manager: Beula Christopher
`Design: Charles Gray
`Mlustration Manager: Gillian Richards
`Illustrator: Cactus
`Marketing Managers (UK/USA): Richard Jones / Helena Mutak
`
`Exhibit 2084
`
`Page 02 of 19
`
`Exhibit 2084
`Page 02 of 19
`
`

`

`Pharmacotherapy
`
`Rvoahel
`
`!
`:
`
`Quan Dong Nguyen mp, MSc
`Associate Professor of Ophthalmology
`Diseases of the Retina and Vitreous, and Uveitis
`WilmerEyeInstitute
`Johns Hopkins University School of Medicine
`Baltimore, MD, USA
`
`Eduardo Buchele Rodrigues mp
`Department of Ophthalmology
`Vision Institute
`Federal University of Sao Paulo
`Sao Paulo, Brazil
`
`Michel Eid Farah mp
`Professor of Ophthalmology and Vice-chairman
`Retina and Vitreous Department
`Vision Institute
`Federal University of Sao Paulo
`Sao Paulo, Brazil
`
`William F. Mieler mp
`Professor and Vice-Chairman
`Department of Ophthalmology & Visual Sciences
`University ofIllinois at Chicago
`Chicago, IL, USA
`
`
`
` | ESTES
`
`Exhibit 2084
`
`Page 03 of 19
`
`Exhibit 2084
`Page 03 of 19
`
`

`

`SAUNDERS
`ELSEVIER
`
`SAUNDERSis an imprint of ElsevierInc.
`
`© 2010,Elsevier Inc. All rights reserved.
`
`First published 2010
`
`Chapter 35 in this publication is in the public domain and may be used and reprinted without special
`permission: citation of the source, however, is appreciated.
`
`No part of this publication may be reproduced or transmitted in any form or by any means,electronic or
`mechanical,
`including photocopying, recording, or any information storage and retrieval system, without
`permission in writing from the publisher. Details on how to seek permission, further information about
`the Publisher’s permissions policies and our arrangements with organizations such as the Copyright
`Clearance Center and the Copyright Licensing Agency, can be found at our website: www.elsevier.com/
`permissions.
`
`British Library Cataloguing in Publication Data
`Retinal pharmacotherapy.
`1. Retina-Diseases—Chemotherapy.
`|. Rodrigues, Eduardo.
`617.7’35061 -dce22
`
`ISBN-13: 9781437706031
`
` Exhibit 2084
`
`Library of Congress Cataloging in Publication Data
`A catalog record for this book is available from the Library of Congress
`
`Notices
`Knowledge and bestpracticein this field are constantly changing. As new research and experience broaden
`our understanding, changesin research methods, professional practices, or medical treatment may become
`necessary. Practitioners and researchers mustalwaysrely on their own experience and knowledgein evaluat-
`ing and using any information, methods, compounds, or experiments describedherein. In using suchinfor-
`mation or methods they should be mindful of their own safety and the safety of others, including parties for
`whom they have a professional responsibility.
`With respect to any drug or pharmaceutical productsidentified, readers are advised to check the most
`current information provided(i) on procedures featured or(ii) by the manufacturer of each product to be
`administered, to verify the recommended doseor formula, the method and duration of administration, and
`contraindications. It is the responsibility of practitioners, relying on their own experience and knowledge of
`their patients, to make diagnoses, to determine dosages and the best treatment for each individual patient,
`and to take all appropriate safety precautions.
`To the fullest extent of the law, neither the Publisher nor the authors, contributors, or editors, assume any
`liability for any injury and/or damage to personsor property as a matter of products liability, negligence or
`otherwise, or from any use or operation of any methods, products,instructions, or ideas contained in the
`material herein.
`
`The Publisher
`
`your source for books,
`OAC journals and multimedia
`in the health sciences
`www.elsevierhealth.com
`
`
`
`Working together to grow
`libraries in developing countries
`
`www.elsevier.com | www.bookaid.org | www.sabre.org
`BOOK AID
`International
`ELSEVIER
`Sabre Foundation
`
`
`
`The
`publisher's
`policy is to use
`paper manufactured
`from sustainable forests
`
`Printed in China
`Last digit is the printnumber:9 8
`
`7
`
`6
`
`5
`
`4
`
`3
`
`2
`
`1
`
`Page 04 of 19
`
`Exhibit 2084
`Page 04 of 19
`
`

`

`CHAPTER
`
` LEO
`
`
`
`
`Ocular angiogenesis: vascular
`endothelial growth factor
`and other factors
`
`Anthony P. Adamis, MD
`
`
`
`INTRODUCTION
`
`Theoriginal visionary proposal by Dr. Judah Folkman!that antiangio-
`genic therapy couldoffer an approachto the treatment of manycancers
`ultimately led to a major research effort into the mechanisms which
`control both physiological and pathological angiogenesis. His work
`also contemplated the use of antiangiogenic drugs in ophthalmology.
`A principal focus of this research effort has been the identification of
`Specific molecules involved in the promotion andinhibition of angio-
`enesis, an effort that has already led to the developmentof targeted
`therapies against vascular endothelial growth factor (VEGF). In addi-
`tion, manyother factors have been identified that act as promoters or
`inhibitors of angiogenesis (Table 4.1). This chapter will focus on those
`molecules whose roles have been best validated to date, and which
`possess particular relevance to ocular neovascularization.
`
`
`
`PROMOTERSOF ANGIOGENESIS
`
`VASCULAR ENDOTHELIAL
`GROWTH FACTOR
`
`VEGFin physiologic and pathologic
`angiogenesis
`VEGF(also known as VEGF-A)is a 45-kDa homodimeric glycoprotein
`belonging to a family that also includes VEGF-B through VEGF-E,
`platelet-derived growth factor (PDGF), and placental growth factor?
`Initially isolated as a vascular permeability factor, VEGF was
`subsequently cloned and found to be a potent proangiogenic factor,
`acting as a master regulator of angiogenesis (reviewed by Ferrara and
`Davis-Smyth’ and Ferrara’). VEGFhas subsequently been foundto act
`in a wide variety of other physiological contexts,’ some of which,
`such as neuroprotection, are completely independentof its role in
`angiogenesis.
`Alternative splicing of the human VEGFgeneyields six principal
`isoforms of 121, 145, 165, 183, 189, and 206 amino acids.> The corre-
`sponding rodent isoforms are one aminoacid shorter.? Many studies
`have focused on characterizing the functions of VEGF,2, VEGFy¢5, and
`VEGFigo. VEGF,.;is freely diffusible, while VEGF) and larger isoforms
`are found sequestered in the extracellular matrix; VEGFy5 exists in both
`diffusible and matrix-bound forms.’ VEGFacts as a ligand for VEGF
`teceptor 1 (VEGFR1) and VEGFR2; these receptor tyrosine kinases in
`turn activate downstream signaling cascades.
`VEGFacts in many capacities in angiogenesis, including as an endo-
`thelial cell mitogen’ and survivalfactor,” and as a chemoattractantfor
`bone marrow-derived endothelial progenitorcells.* In addition, VEGF
`induces the upregulation of extracellular matrix-degrading enzymes,
`such as matrix metalloproteinases (MMPs)’ and plasminogen activa-
`tor,'” as well as nitric oxide," a downstream mediator of VEGFsignal-
`ing.'’ Moreover, VEGFhas twoadditional properties whichareofdirect
`relevance for the pathophysiology of ocular neovascular diseases.First,
`
`Exhibit 2084
`
`Page 05 of 19
`
`it is the most potent known inducerof vascular permeability," an action
`related to the edema which often accompanies ocular neovasculariza-
`tion. Secondly, the retinal expression of VEGF, which is produced by
`a wide variety ofretinal cell types,'*"* is upregulated by hypoxia,'*"”
`a responsethatis believed to be important in maintaining the health
`of both retinal neurons® and the choriocapillaris” while also creating
`a proangiogenic environment.
`Reflecting the original focus of Dr. Folkman’s proposal on the impor-
`tance of angiogenesis in cancer growth and metastasis,’ initial investiga-
`tions of the role of VEGF in pathological angiogenesis demonstrated
`that interference with VEGFsignaling inhibited tumor growth.!? Over
`the course of a decade, a role for VEGFin ocular neovascular disease
`also wasestablished based on three main lines of evidence: (1) correla-
`tions of VEGFelevation with the presence of ocular neovascular disease
`in the eyesofpatients; (2) preclinical studies demonstrating that experi-
`mental elevation of VEGFlevels in the eye led to neovascularization;
`and(3) the converse experiment, in which inhibition of VEGFsignaling
`decreased neovascularization.
`Correlations betweenelevations in ocular levels of VEGF and ocular
`neovascular disease have been reported and include conditions such
`as iris neovascularization, retinal vein occlusion, diabetic retinopathy
`(DR), diabetic macular edema (DME), neovascular glaucoma,andreti-
`nopathy of prematurity (reviewed byStarita etal."). Elevated expres-
`sion of VEGFalso has been detected in surgically removed maculae”
`and choroidal neovascularization (CNV) membranesof eyes with age-
`related macular degeneration (AMD).”
`A variety of approaches have been employed to demonstrate that
`elevated ocular levels of VEGFare sufficient to induce ocular neovas-
`cularization. These haveincluded direct intravitreal injection of VEGE”
`and retinal vein photocoagulation” in monkeys; in rodent models,
`studies have included intravitreal
`injection of VEGF-expressing
`vectors,“4 and the use of transgenic mice engineered to overexpress
`VEGFin the retinal pigment epithelium (RPE).”
`The experiments demonstrating that VEGFelevations are necessary
`for the developmentof ocular neovascularization have also employed
`various techniques. Agents used to block the actions of VEGF have
`included VEGFR fusion proteins,”anti-VEGFantibodies,” an anti-
`VEGF monoclonalantibody antigen-binding fragment (Figure 4.1);
`an aptamer directed against VEGFys," and VEGF,sb, a VEGE variant
`which binds VEGFR2 but cannotactivateit.” Agents used to block the
`ocular production of VEGF or VEGFR1atthetranscriptional or trans-
`lational level have included small interfering RNAs (siRNAs) specific
`for VEGF® or VEGFR1,™ and antisense oligonucleotides specific for
`VEGE®*Blocking the actions of VEGF in the eye by various means
`inhibited neovascularization of theiris,” cornea,” retina," and
`choroid.2731334
`Further detailed investigations into the mechanisms underlying
`VEGF’s importance have revealedthat the isoform VEGF«5 is especially
`pathogenic. In a murine model of ischemia-associated ocular neovas-
`cularization, retinal expression of VEGF,,; was foundto be dramatically
`elevated compared to other isoforms; moreover, intravitreal injection
`of a VEGFi¢5-specific RNA aptamer wasasefficient at inhibiting the
`pathological neovascularization as injection of a VEGFR-Fc fusion
`protein that inactivated all VEGF isoforms (Figure 4.2).”* In addition,
`VEGF;5 acts as an especially potent
`inflammatory cytokine, a
`property of direct relevance given the importance of inflammation in
`
`23
`
`Exhibit 2084
`Page 05 of 19
`
`

`

`
`
`
`
`$10]08-]JBUIOPUP1O}Oe4YIMOJD[eljeyJOPUFJejnoseA‘siseueHo!BuyJIN°yHALdWHO
`
`
`
`
`
`
`
`
`
`
`
`Table 4.1 Proangiogenic and antiangiogenic factors
`
`Vasostatin (calreticulin fragment)
`
`
`
`Proangiogenic
`factors
`
`Angiogenin
`Angiopoietin- 1
`Complementfactors C3
`and C5
`
`Cryptic collagen IV
`fragment
`Developmentally
`regulated endothelial
`locus 1 (Del-1)
`Fibroblast growth factors:
`acidic (aFGF) and basic
`(bFGF)
`Follistatin
`
`Granulocyte colony-
`stimulating factor
`(G-CSF)
`Hepatocyte growth factor
`(HGF)/scatter factor (SF)
`Interleukin-8 (IL-8)
`a5 integrins
`Leptin
`Midkine
`
`Pigment epithelium-
`derived growth factor
`Placental growth factor
`Platelet-derived
`endothelial cell growth
`factor (PDECGF)
`Platelet-derived growth
`factor-BB (PDGF-BB)
`Pleiotrophin (PTN)
`Progranulin
`Proliferin
`
`Transforming growth
`factor-a (TGF-a)
`Transforming growth
`factor-B (TGF-B)
`Tumor necrosis factor-a.
`(TNF-a)
`Vascular endothelial
`growth factor (VEGF)
`
`Antiangiogenic factors
`
`Angioarrestin
`Angiostatin (plasminogen
`fragment)
`Antiangiogenic antithrombin Ill
`Cartilage-derived inhibitor (CDI)
`CD59 complement fragment
`Endostatin (collagen XVIII
`fragment)
`Fibronectin fragment
`Growth-related oncogene (Gro-B)
`Heparinases
`Heparin hexasaccharide
`fragment
`Human chorionic gonadotropin
`(hCG)
`Interferon o/B/y
`Interferon-inducible protein
`(IP-10)
`Interleukin-12
`
`Kringle 5 (plasminogen fragment)
`Metalloproteinase inhibitors
`(TIMPs)
`2-Methoxyestradiol
`Pigment epitheliurn-derived
`growth factor
`Placental ribonuclease inhibitor
`
`Plasminogenactivator inhibitor
`Platelet factor-4 (PF4)
`Prolactin 16-kDa fragment
`Proliferin-related protein (PRP)
`Retinoids
`Soluble VEGFR-1
`
`Tryptophanyl-tRNA synthase
`fragment
`VEGF0
`Tetrahydrocortisol-S
`Thrombospondin-1 (TSP-1)
`Transforming growth factor-B
`(TGF-B)
`Vasculostatin
`
`prematurity (Figure 4.4)* and in laser-induced CNV.* VEGF,; was
`more potent at chemotaxis of monocyte/macrophages than VEGF,).”
`Since macrophages produce VEGE,” their infiltration serves as an
`amplification mechanism in further promoting angiogenesis.
`
`Investigational approaches to VEGF inhibition
`in ocular neovascularization
`
`The extensive research effort into elucidating VEGF’s role in ocular
`neovascularization has provided a sound foundation for the develop-
`mentof anti-VEGFtherapies. Three agents, pegaptanib,” ranibizumab,"
`and bevacizumab,” are already in widespread use, and are discussed
`in dedicated chapters of this text. A brief account of other approaches
`currently under evaluation in clinicaltrials follows.
`
`RNAinterference
`
`RNAinterference abrogates gene expression througha cellular defense
`mechanism mediated by double-stranded RNA sequencesofatleast
`21 nucleotides long, resulting in targeted destruction of specific mRNA
`species.” RNA interference has been used to target VEGF mRNAin
`animal models, leading to suppression of corneal neovascularization™
`as well as CNV induced either by laser® or by overexpression of VEGF
`from a transgene."* Sirna-027, an agent targeting the expression of
`VEGEFR1,also has been shown to suppress both retinal and CNV in
`murine models.™
`Currently there are two siRNA agents undergoing evaluation in
`clinical trials for treatment of neovascular AMD.Bevasiranib (Ophi
`Health), directed against VEGF, has successfully completed a phase II
`trial and is currently recruiting patients for the phase III COBALTtrial
`in which it will be combined with ranibizumab.” In addition, a phase
`I trial of the anti-VEGFR1 agent AGN211745 (Allergan; previously
`Sirna-027) has been completed,” and enrollment for a phaseII trial is
`ongoing.** Recent evidence suggests that antiangiogenic siRNAs work
`nonspecifically and through a nonclassical siRNA mechanism in sup-
`pressing CNV.”
`
`Soluble VEGFRfusion protein: VEGF-Trap —
`Work demonstrating the potential of soluble VEGFR fusion proteins to
`suppress retinal neovascularization” provided a basis for the develop-
`ment of VEGF-Trap, a fusion protein combining components of both
`VEGFR1 and VEGFR2.” VEGF-Trap, which was engineered with a
`view to optimizing pharmacokinetic properties as well as efficacy,
`binds to all isoforms of VEGF as well as placental growth factor.”
`Intravitreal injection of VEGF-Trap inhibited laser-induced CNV in
`mice, as well as preventing VEGF-induced blood-tetinal barrier break-
`down.”It is now being evaluated in a phase III study.”
`
`Anecortave acetate
`
`Anecortaveacetate is a memberof a group of corticosteroids,first iso-
`lated in Dr. Folkman’slaboratory,” that have angiostatic properties but
`lack conventional anti-inflammatory activity.” In a rat retinopathy of
`prematurity model, anecortave significantly reduced pathologic retinal
`neovascularization without affecting normalretinal angiogenesis.” In
`other studies with this model, anecortave was foundto reduceretinal
`expression of VEGE®andofinsulin growth-factor-1 andits receptor.”
`Anecortave also inhibited VEGFR2 expression in a murine model of
`retinoblastoma.” These findings suggest that the angiostatic effects of
`anecortave mayatleast in part be mediated through VEGFsignaling
`pathways.”
`Anecortave acetate has shown some promise as a treatment for
`neovascular AMD, administered as a juxtascleral depoteither alone®
`or in combination with photodynamic therapy.” Although anecortave
`acetate did not meetits efficacy endpointin a phase II noninferiority
`trial comparing it
`to photodynamic therapy with verteporfin,® it
`remains understudy as a prophylactic treatment to slow the progres-
`sion of neovascular AMD.”
`
`Adapted from: Angiogenesis Foundation. Understanding angiogenesis.
`List of known angiogenic growth factors. Available online at: http://www.
`angio.org/understanding/content_understanding. html.
`
`pathological neovascularization. Laser injury has been shown to up-
`regulate retinal expression of intercellular cell adhesion molecule-1
`(ICAM1), thereby promoting leukocyte adhesion to the vascular endo-
`thelium through CD18, the leukocyte ligand for ICAM1.* Genetic
`ablation of either molecule significantly reduced the formation of
`laser-induced CNV (Figure 4.3). In this context, it is noteworthy that
`VEGF,,; was found to be significantly more potent at upregulating
`ICAM1expression on endothelial cells than VEGF;>,.” In addition,
`depletion of macrophages has been found to inhibit the development
`of pathological neovascularization in a rat model of retinopathy of
`
`24
`
`Exhibit 2084
`
`Page 06of 19
`
`Exhibit 2084
`Page 06 of 19
`
`

`

` |
`
`
`
`
`
`BuljyoYyUlS99USIDSJISege|NOILOAS
`
`| Prevention
`
`| Control
`
`No.ofgrade4CNVlesions
`
`35
`
`42
`
`Days
`
`Control/Treatment
`
`30
`
`No.ofgrade4CNVlesions
`
`Days
`
`Es
`
`x
`Figure 4.1 Inactivation ofall vascular endothelial growth factor (VEGF) isoforms potently inhibits laser-induced choroidal neovascularization
`(CNV) in the nonhuman primate. (A) Cynomolgus monkeys(n = 10) received 500 pig of recombinant humanized monoclonal anti-VEGF
`
`antibody (rhuFab VEGF) in one eye andvehicle in the other, every 2 weeks. On day 21, CNV wasinducedby laser wounding. The bar
`“graph showsthe total number of grade 4 CNV lesions in the eyes receiving rhuFab VEGF(gold bar) compared to thosein control eyes that
`‘Teceived vehicle (blue bar); assessments were made 2 weeksafter laser induction (day 35), and 3 weeksafter the laser induction (day 42)
`Adapted from Krzystolik MG, Afshari MA, Adamis AP,et al. Prevention of experimental choroidal neovascularization with intravitreal anti-
`vascular endothelial growth factor antibody fragment. Arch Ophthalmol 2002;120:338-346.
`1.0-
`
`
`
`
`
`(mm?) on oSie)
`
`|Control 4
`|
`[BB VEGF46q-selective blockade
`a8
`_ |) Nonselective VEGF blockade
`
`baadfo)
`
`Area
`
`0.0-
`
`especially critical for the recruitment of mural cells (pericytes and
`smooth-muscle cells) to the developing vasculature.” Genetic ablation
`of PDGF-Bleads to perinatal death from hemorrhages and vascular
`system abnormalities” while ablation of the PDGFR-generesults in a
`similar phenotype.” Proliferation of mural cells was significantly
`reduced in mice lacking either PDGF-B or PDGER-B.* Also, administra-
`tion of an aptamerspecific for PDGF-Bledfirst to pericyte loss and then
`to regression of tumorvessels in a murine tumor model. These find-
`ings indicate that PDGF-B produced by endothelial cells is essential for
`the proliferation, migration, and recruitmentof muralcells to the devel-
`oping capillaries (Figure 4.5)."°
`Studies of ocular neovascularization in mice have provided further
`evidence in support of this model. Inhibition of PDGF-B signaling,
`whether by genetic ablation in endothelial cells or PDGFR kinase
`inhibitors,” led to deficient pericyte recruitment in models ofretinal”
`and corneal” neovascularization.
`Studies using three different models of ocular neovascularization, in
`which PDGF-B and VEGFsignaling were blocked by administration of
`an antibody to PDGFR-f or pegaptanib,respectively, have furtherdelin-
`eated the respective roles of these molecules.” Physiological retinal
`angiogenesis wasinhibited on postnatal day 3 by blocking PDGF-B, but
`notby blocking VEGF,«:; however, combined blockadeprovided greater
`inhibition. Conversely, VEGFblockadealoneinhibited the development
`of laser-induced CNV, whereas blocking PDGF-B signaling was ineffec-
`tive on its own,again,greater inhibition occurredifboth pathways were
`blocked. Finally, in a corneal model of neovascularization, PDGF-B
`blockade between days 10 and 20 postinjury led to detachmentof mural
`cells from corneal neovessels; in contrast, VEGF blockade reduced neo-
`vascularization when applied immediately after wounding, butit did
`not induce regression ofvessels after they were established. However,
`vessel regression was enhancedif both inhibitors were given (Figure
`4.6).These experiments suggest that a combination strategy targeting
`both VEGF and PDGF-B may be more effective, both in treating estab-
`lished neovascularization and in preventing new vessel growth.
`
`FIBROBLAST GROWTH FACTOR2 (FGF2)
`
`FGF2(also knownasbasic FGF)is a heparin-binding growth factorthat
`occurs in several isoforms. FGF2 signals throughfour receptor tyrosine
`kinases (FGFreceptor 1 through FGFreceptor 4) andacts ina variety
`of developmentalprocesses, including angiogenesis.”
`
`25
`
`
`
`
`The PDGF family consists offour related dimeric polypeptides (PDGF-A
`thtough PDGF-D)" that are structurally related to VEGF? In general
`
`they occur as homodimers, although the PDGF-AB heterodimer has
`also been identified." PDGFs are ligands for two receptor tyrosine
`
`Kinases, PDGFR-o. and PDGER-B, of which PDGER-B is principally
`Tesponsible for signal transduction on cells associated with the vascular
`system,
`including endothelial cells, pericytes, and smooth-muscle
`
`cells. Similarly, PDGFalso has a widespread distribution among these
`“samecell types.” In addition to its central role in vascular system
`_development, PDGF signaling is important for processes such as
`_Woundhealing and central nervous system development.”
`
`__
`Studies have revealed a central role for the PDGF-B homodimerin
`vascular development, as it was found to stimulate the proliferation,”
`
`and inducecapillary tube formation™ of endothelial cells. PDGF-B is
`
`e Figure 4.2 Vascular endothelial growth factor (VEGF64/165) is
`€specially potent in promoting pathological neovascularization. In a
`_fat modelof ischemia-induced retinal neovascularization, intravitreal
`injection of an aptamer specific for VEGF164165 Was as effective in
`"inhibiting pathological neovascularization as a VEGFR1-Fe fusion
`_ protein which binds all VEGF isoforms. Adapted from Ishida S, Usui
`T, Yamashiro K,et al. VEGF164-mediated inflammation is required
`
`for pathological, but not physiological, ischemia-induced retinal
`
`“neovascularization. J Exp Med 2003;198:483-489.
`
`LATELET-DERIVED GROWTH FACTOR
`
`
`Exhibit 2084
`
`Page 07 of 19
`
`Exhibit 2084
`Page 07 of 19
`
`

`

`CD18-/-
`
`Wildtype C57BL/6J
`ICAM-1 -/-
`Percent Week1
`
`P=0.02
`
`P=0.015
`
`P=0.042
`
` 9= 4+mJa
`
`ae@
`OQO
`
`c= 52o©@
`
`Q
`=,
`QO
`De
`
`9<»Q9
`

`cS
`
`=m
`
`M2
`
`Q 2aDa ) >T
`
`Figure 4.3 Evidenceofthe role of inflammation in the model
`oflaser-induced choroidal neovascularization (CNV). (A) Genetic
`ablation of either CD18 or intercellular cell adhesion molecule-1
`(ICAM1) led to marked diminution of the size of laser-induced CNV.
`Two weeks following laser injury, stacked confocal images were
`takenof fluorescein Griffonia simplicifolia lectin |-labeled tissue within
`the laser scars. CNV membranes were significantly reduced in both
`mutant strains, compared to wild-type mice. Scale bar, 100 um.
`(B) Loss of either CD18 or ICAM1 resulted in fewer lesions of
`pathological significance. Fluorescein angiography performedat
`1, 2, and 4 weeksafter laser photocoagulation demonstrated that
`ablation of either CD18 (blue bar) or ICAM1 (purple bar) resulted in
`Significantly fewer grade 2B lesions (those showing pathologically
`significant leakage) than were seen in wild-type mice (gold
`lbet)
`bar) (mean + SEM: n =5forall groups). Adapted from Sakurai E
`=°x
`Week2
`Week 4
`Taguchi H, AnandA,etal. Targeted disruption of the CD18 or
`(B)
`ICAM-1 geneinhibits choroidal neovascularization. Invest Ophthalmol!
`»=2)
`Vis Sci 2003;44:2743-2749,
`
`The role of FGF2 in ocular neovascular disease is not well defined.
`Elevated expression of FGF2 has been detected in CNV membranes
`from patients with AMD” and in epiretinal membranes from patients
`with proliferative DR.” However, exogenous administration of FGE2
`produced only subretinal neovascularization that did not penetrate
`Bruch’s membrane in an experimental model of CNV. Other studies
`foundthat transgenic mice with elevated retinal FGF2 expression devel-
`oped CNV following low-intensity laser (sufficient to disrupt photo-
`receptors but not Bruch’s membrane) while wild-type mice did not.”
`Taken together with studies demonstrating that genetic ablation of the
`FGF2 genedid notinhibit the formation oflaser-induced CNV,”these
`findings suggest that FGF2isin itself not sufficient to provoke CNV in
`the absence of an additional stimulus and that FGF2 mayalso not be
`required to induce CNV.
`
`TUMOR NECROSIS FACTOR-o (TNF-a)
`TNF-a is the prototypic member ofa superfamily of cytokines that
`mediate a variety of biological functions, signaling through a corre-
`spondingly large family of receptors.” Several studies have examined
`the role of TNF-c. as a mediatorof angiogenesis, but a unified picture
`is not yet apparent.
`TNF-o. has been found to stimulate angiogenesis in the corneas of
`rats® and rabbits.” It is notclear if these representdirect or indirect
`effects since TNF-a has been demonstrated to induce expression of
`VEGF” and VEGFR2"potently in cultured endothelial cells. TNEF-o.
`also upregulates the synthesis of other factors associated with angio-
`genesis, including angiopoietin 1 and angiopoietin 2as well as MMP2
`and MMP9.*
`
`a Sa
`
`e
`@Q=
`
`ob!
`
`Qoa
`
`26
`
`Several studies have assessed therole of TNF-o. signaling in angio-
`genesis. In ischemic-induced neovascularization in the limbs of mice,
`TNF-a wasessentialfor the mobilization andsurvival ofbone marrow-
`derived endothelial progenitorcells, induction of VEGF expression and
`collateral vessel development.® In another report, administration of
`infliximab (a monoclonal antibody to TNF-a)* or etanercept(a soluble
`TNF receptor fusion protein) both inhibited the size of laser-induced
`CNV in mice. Gene knockout studies, however, have been inconsis-
`tent; somestudies found a dependenceofretinal neovascularization on
`TNF-a function® whereas others did not.
`In clinical studies, elevated levels of TNF-o. have been found in
`fibrovascular membranes of patients with proliferative DR®’ and
`in surgically excised CNV membranes. Intriguingly,
`intravenous
`administration of infliximab for treatment of rheumatoid arthritis
`caused regression of CNV in patients with AMD”; moreover, intrave-
`nous infliximab also led to reductions in macular edemain patients
`with DME.” It is notclear if these effects of TNF-a are independent of
`its upregulation of VEGF:if separate pathwaysare involved, TNF-a
`inhibition aloneorin combination with VEGFinhibition could provide
`an additional therapeutic option.
`
`EPHS AND EPHRINS
`ee
`Ephs comprisea large family of receptortyrosine kinases that are acti-
`vated upon binding with their cognate membrane-bound ligands, the
`ephrins.””” EphrinAsare attached to the cell membrane by a glyco-
`sylphosphatidy] anchor while the ephrinBs have transmembrane and
`cytoplasmic signaling domains (Figure 4.7).The Ephs alsofall into two
`
`|
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`Serre
`Page 08 of 19
`
`Exhibit 2084
`Page 08 of 19
`
`

`

`(a o
`
`an
`E
`g°°
`oc
`
`304
`mo
`a
`
`0.2
`
`0
`
`7 (clodronate)
`
`u
`
`P>0.5
`
`(D) 30
`
`=
`E 20
`8
`os
`
`z
`= 10
`a
`
`Clodronate-
`aes liposome
`
`:
`
`PBS-
`liposome
`.
`
`Clodronate-
`;
`
`
`
`
`
` BulyeyUlSBOUSIDSJIsege|NOILOAS
`liposome (kK)
`
`Normoxia hypoxia
`
`
`164 —>
`120 —>
`
` 188 —>|
`
`
`Figure 4.4 Monocytes contribute to pathological retinal neovascularization. In a retinopathy of prematurity model, postnatal day zero (PO)
`rats were maintained for 10 days in 80% oxygen, interrupted daily by 30 minutes in room air, followed by a progressive return to 80%
`Oxygen. This treatment led to an avascular retina. On P10, corresponding to study day 0 (D0), retinal revascularization was induced by
`maintaining the rats in room air for an additional 7 days (D7). (A-C) At D7, pathological neovascularization (PaNV; arrowsin A and B) was
`Significantly inhibited by treatment with clodronate liposomes compared to controlliposomes (n = 8 for both treatments; means + standard
`deviation). (D) Physiological neovascular area (PhRV) wasnotsignificantly affected by treatment with clodronateliposomes(P > 0.05).
`(E-J) Influx of monocytes was observed just before and during pathological neovascularization. (H-J) Monocytes were labeled with a
`fluorescein conjugated antibody to CD13 (E and H), while rhodamine-conjugated Concanavalin A was used to label the retinal vasculature
`and adherent leukocytes (F and |). As shown by superposition of these figures (panels G and J), the concanavalin A and CD13 staining
`co-localized, indicating that the adherent leukocytes were monocytes. (K) In cultured peripheral blood monocytes obtained from
`retinopathologic rats at D7, exposure to hypoxia (1% oxygen) led to marked increase in expression of vascular endothelial growth factor
`mRNA compared to exposure to normoxia (21% oxygen). PBS, phosphate-buffered saline. Scale bars: (A and B) 0.5 mm and (E-J) 50 um.
`Reproduced from Ishida S, Usui T, Yamashiro K,et al. VEGF164-mediatedinflammation is required for pathological, but not physiological,
`ischemia-inducedretinal neovascularization. J Exp Med 2003;198:483-489.
`
`Wild type
`
`Figure 4.5 Platelet-derived growth factor (PDGF)-B regulates
`the developmentof blood vessel walls. During blood vessel
`development, the nascent endothelial tube (yellow) is surrounded
`by undifferentiated mesenchymalcells (gray) which are induced to
`
`differentiateintovascularsmooth-musclecells(VSMC), andtoform
`
`a surrounding sheath (red). During further development of the
`vascular network, with concomitant growth and sprouting of blood
`vessels, PDGF-B derived from the endothelium further promotes
`VSMCproliferation and migration. These proliferative and migratory
`responses are reducedin mice in which PDGF-B or PDGFR-B have
`been genetically ablated, leading to defective coating of capillaries
`by pericytes, as well as to VSMC hypoplasiain larger vessels.
`Reproduced from Hellstrom M, Kalen M, Lindahl P, et al. Role of
`PDGF-B and PDGFR-betain recruitment of vascular smooth muscle
`Cells and pericytes during embryonic blood vessel formation in the
`mouse. Development 1999;126:3047-3055.
`
`Exhibit 2084
`
`Page 09 of 19
`
`PDGF-Bdriven
`VSMCproliferation
`and migration
`
`vSMC
`induction
`
`
`o~«@eG“”
`PDGF-B
`and migration
`
`Reduced vSMC
`proliferation
`
`PDGFR-B
`knock-out
`
`27
`
`Exhibit 2084
`Page 09 of 19
`
`

`

`
`
`
`
`
`
`$10}0e4JA8UJOPUeJOJOB4YIMOIHJeljayyOpuUyZJejnose/\ssiseueBolbuyJeINdOefHALdVHO
`
`
`
`
`
`
`
`
`
`
`
`PBS
`control
`
`Anti-VEGF
`aptamer
`
`Ant-PDGFR-B
`antibody
`
`Ant-VEGF aptamer +
`anti-PDGFR-6 antibody
`
`Figure 4.6 The role of platelet-derived growth factor (PDGF)-B on blood vessel growth and muralcell coverage in a corneal
`neovascularization model. (A) Endothelial cells were labeled by staining with lectin (green) and mural cells were stained with an antibody
`against smooth-muscle actin (red). Starting at 10 days following cornealinjury, mice received daily intraperitoneal injections of an anti-
`PDGFR-B antibody or phosphate-buffered saline (PBS), and were sacrificed at 20 days postinjury. Treatment with the anti-PDGF-B antibody
`led to reduced mural cell coverage compared to controls (arrow). Scale bar = 20 um. (B) Following induction of cornealinjury, mice received
`daily intraperitonealinjections of one of the following: PBS, a polyethylene-glycolated anti-vascular endothelial growth factor (VEGF) aptamer,
`an anti-PDGFR-B antibody, or both the anti-VEGF aptamer and the anti-PDGFR-B antibody. Neovasculature (green) was stained by
`fluorescein isothiocyanate-concanavalin A. Neovascularization wassignificantly reduced by the anti-VEGF aptamer compared with either
`PBSorthe anti-PEGFR-B antibody (P < 0.01); inhibition of both VEGF and PDGF-Bsignaling led to a furthersignificant reduction (P < 0.05),
`comparedto inhibition of VEGF signaling alone. Scale bar = 100 um. Adapted from Jo N, Mailhos C, Ju M, etal. Inhibition of platelet-
`derived growth factor B signaling enhancestheefficacy of anti-vascular endothelial growth factor therapy in multiple models of ocular
`neovascularization. Am J Pathol 2006; 168:2036-2053.
`
`broad groups, EphA and EphB, with the EphAs binding primarily,
`although notexclusively, to members of ephrinA subclass, while EphBs
`similarly tend to bind preferentially to ephrinB ligands.
`Owing tothe association of ephrins to cell

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