throbber
;.. '
`
`'
`~
`
`Retina.
`v. 30, no. 9 (Oct. 201 0)
`General Collection
`W1 RE2498
`2010-10-15 10:25:28
`
`PATHWAY-BASED THERAPIES FOR AGE-RELATED MACULAR
`DEGENERATION
`Zarbin, Rosenfeld
`"TREAT AND EXTEND" FOR TYPE 3 CHOROIDAL NEOVASCULARIZATION
`Engelbert, Zweifel, Freund
`FREQUENCY OF MACULAR HEMORRHAGES
`Barbazetto, Saroj, Shapiro, Wong, Freund
`INTRAVITREAL ANTI-VEGF HEMORRHAGIC COMPLICATIONS
`Mason, III, Frederick, Neimhin, White, Jr, Feist, Thomley, Albert, Jr
`WET AGE-RELATED MACULAR DEGENRATION: LESION CHANGES WITH
`RANIBIZUMAB
`Sadda, Stoller, Boyer, Btodi, Shapiro, Ianchuley
`RANIBIZUMAB FOR CHOROIDAL NEOVASCULARIZATION SECONDARY TO
`PUNCTATE INNER CHOROIDOPATHY
`Menezo, Cuthbertson, Downes
`BEVACIZUMAB DURING PREGNANCY
`Tarantola, Folk, Boldt, Mahajan
`AQUEOUS HUMOR IN DIABETIC MACULAR EDEMA
`Funk, Schmidinger, Maar, Bolz, Benesch, Zlabinger, Schmidt-Erfurth
`BEVACIZUMAB IN PIGMENT EPITHELIUM DETACHMENT AS A RESULT
`OF CHOROIDAL NEOVASCULARIZATION IN AGE-RELATED MACULAR
`DEGENERATION
`Acft, Hoeh, Ruppenstein, Kretz, Dithmar
`INTRAVITREAL BEVACIZUMAB FOR AGE-RELATED MACULAR
`DEGENERATION
`Tao, Jonas
`BEVACIZUMAB-RELATED INFLAMMATION
`Chong, Anand, Williams, Qureshi, Callanan
`DRUSEN IMAGING
`Spaide, Curcio
`HIGH-RESOLUTION OPTICAL COHERENCE TOMOGRAPHY IN ADULT
`VITELLIFORM MACULAR DYSTROPHY
`Finger, Charbellssa, Kellner, Schmitz-Valckenbetg, Fleckenstein, Scholl, Holz
`CYTOKINE LEVELS IN CENTRAL SEROUS CHORIORETINOPATHY
`Lim, Kim, Shin
`HYPOXIA-INDUCIBLE FACTOR-I a DIABETIC AND NONDIABETIC PATIENTS
`Lim, Spee, Hinton
`EVOLUTION OF AUTOFLUORESCENCE IN MULTIPLE EVANESCENT WHITE
`DOT SYNDROME
`dell'Omo, Mantovani, Wong, Konstantopoulou, Kulwant, Pavesio
`
`Mylan Exhibit 1076
`Mylan v. Regeneron, IPR2021-00880
`Page 1
`
`

`

`jsocl Evaluation of Ultra Wide Angle "ora-ora" High Re fractive Index Self-Stabilizing
`Contact Lens for Vitreous Surgery . . ... . .. .. . . .. ... . . ................. . . . .. 1551
`Ravi K. Murthy, Vikram S. Bra" K. V Chalam
`
`CORRESPONDENCE . . ..... . . . .. . . ... . .. . . .. ..... . ... . . .... . .... . . . . ...... 1554
`
`For information on submitting a manuscript or for subscription information, please
`visit our website, www.retinajournal.com
`
`l'ermission to l'hotocO()Y Articles
`This publication is protected by copyright. Permission to photocopy must be secured in writing from:
`• Permissions Department, Lippi ncou Williams & Wilkins, 351 W. Camden Street, Baltimore, MD 2120 I; telephone 410-528-4050;
`email: journalpermissions@lww.com: URL: www.lww.comfresources/permissionsljournals.html or
`• Copyright Clearance Center (CCC), 222 Rosewood Dr.,Danvet:~. MA 01923; 978-750-8400; FAX:978-750-4470; Internet:
`www.copyright.com; or
`• UMI, Box 49. 300 North Zeeh Road, Ann Arbor, !VII 48106·1346; FAX: 3 I 3-761-1203.
`
`This m ateria I wgs co pied
`atrh<> NLM.,ndmay be
`Sub·je<t USCo·p'l"i~ Laws
`
`Mylan Exhibit 1076
`Mylan v. Regeneron, IPR2021-00880
`Page 2
`
`

`

`Review
`
`PATHWAY-BASED THERAPIES FOR AGE-
`RELATED MACULAR DEGENERATION
`
`An Integrated Survey of Emerging Treatment
`Alternatives
`
`MARCO A. ZARBIN, MD, PHD,* PHILIP J. ROSENFELD, MD, PHD†
`
`Purpose: To review treatments under development
`for age-related macular
`degeneration (AMD) in the context of current knowledge of AMD pathogenesis.
`Methods: Review of the scientific literature published in English.
`Results: Steps in AMD pathogenesis that appear to be good targets for drug
`development include 1) oxidative damage; 2)
`lipofuscin accumulation; 3) chronic
`inflammation; 4) mutations in the complement pathway; and 5) noncomplement
`mutations that
`influence chronic inflammation and/or oxidative damage (e.g.,
`mitochondria and extracellular matrix structure). Steps in neovascularization that
`can be targeted for drug development and combination therapy include 1) angiogenic
`factor production; 2) factor release; 3) binding of factors to extracellular receptors (and
`activation of intracellular signaling after receptor binding); 4) endothelial cell activation
`(and basement membrane degradation); 5) endothelial cell proliferation; 6) directed
`endothelial cell migration; 7) extracellular matrix remodeling; 8) tube formation; and 9)
`vascular stabilization.
`Conclusion: The era of pathway-based therapy for the early and late stages of AMD
`has begun. At each step in the pathway, a new treatment could be developed, but
`complete inhibition of disease progression will
`likely require a combination of the
`various treatments. Combination therapy will
`likely supplant monotherapy as the
`treatment of choice because the clinical benefits (visual acuity and frequency of
`treatment) will
`likely be superior
`to monotherapy in preventing the late-stage
`complications of AMD.
`RETINA 30:1350–1367, 2010
`
`A large number of treatments for exudative and
`
`nonexudative age-related macular degeneration
`(AMD) are in preclinical development or in early-
`stage clinical trials (Figure 1). In this review, six
`observations relevant to the pathogenesis of AMD will
`be described. Emerging and established AMD treat-
`ments will then be reviewed within the context of these
`pathogenic schemes. This information should be
`especially useful for the rational development of
`combination therapies.
`
`Pathogenesis of Age-Related
`Macular Degeneration
`
`Detailed consideration of the pathogenesis of AMD
`is beyond the scope of this perspective, but it has been
`discussed extensively elsewhere.1,2 Six concepts will
`be considered briefly.
`First, biochemical studies and histological studies of
`AMD have implicated oxidative damage as a possible
`cause of this disease. Eyes with geographic atrophy
`
`1350
`
`Mylan Exhibit 1076
`Mylan v. Regeneron, IPR2021-00880
`Page 3
`
`

`

`PATHWAY-BASED THERAPIES FOR AMD  ZARBIN AND ROSENFELD
`
`1351
`
`Disease Study (AREDS) (http://clinicaltrials.gov/ct2/
`show/NCT00000145?term=Age-Related+Eye+Disease+
`Study+%28AREDS%29&rank=3) results is that an-
`tioxidant supplementation reduces the risk of visual
`loss associated with AMD among properly selected
`patients, especially for patients with the CFHTT
`genotype.10
`Second, excessive accumulation of lipofuscin in the
`RPE may play an important role in the pathogenesis
`AMD.11 In RPE cells, the main source of lipofuscin is
`probably the undegradable components of phagocytized
`outer segments.12 In vertebrate photoreceptors,
`light
`causes isomerization of visual pigment chromophore, 11-
`cis-retinylidene,
`to all-trans-retinylidene, followed by
`release of all-trans-retinal from the opsin binding pocket
`(Figure 2).13
`and its reduction to all-trans-retinol
`ABCA4, an adenosine triphosphate–binding cassette
`transporter present in the outer segment of rods and
`transports N-retinylidene-phosphatidylethanol-
`cones,
`amine from the outer segment disks to the photoreceptor
`cytoplasm.14,15 Retinol dehydrogenase 8 (in outer
`segments) and retinal dehydrogenase 12 (in inner seg-
`ments) reduces all-trans-retinal to all-trans-retinol.16,17
`Vitamin A (all-trans-retinol) diffuses to RPE where it
`is esterified by lecithin/retinol acyltransferase (LRAT)
`to all-trans-retinyl esters and is stored in retino-
`somes.18,19 All-trans-retinyl esters are isomerized to
`in a reaction involving RPE-65.20–22
`11-cis-retinol
`Next, 11-cis-retinol is oxidized to 11-cis-retinal,23,24
`which then diffuses across the extracellular space to
`photoreceptors and recombines with rod-and-cone
`opsin proteins to regenerate visual pigments. Within
`the outer segment disks, ethanolamine can combine
`with two retinaldehyde molecules to form N-retiny-
`lidene-N-retinylethanolamine (A2E); A2E is a major
`fluorophore in lipofuscin found in the RPE.25
`Third, AMD is associated with chronic inflammation
`in the region of the RPE, Bruch’s membrane, and
`choroid.26 Several lines of evidence demonstrate this
`fact. Drusen, for example, contain many components of
`the activated complement cascade.27–29 Anatomical
`studies demonstrate the presence of inflammatory cells
`in Bruch membrane.30 Bioactive fragments of C3 (C3a)
`and C5 (C5a) are present in the drusen of AMD eyes and
`induce vascular endothelial growth factor
`(VEGF)
`expression in RPE cells.31 The latter findings may
`explain why confluent soft drusen are a risk factor for
`CNVs in AMD eyes. The presence of proinflammatory
`molecules in drusen constitutes a stimulus for chronic
`inflammation in the RPE–Bruch membrane–chorioca-
`pillaris complex that may result in some features of late
`AMD. One interpretation of the AREDS is that zinc, one
`of the main therapeutic ingredients of this treatment, also
`affects the complement system, which in turn may slow
`
`Fig. 1. Rate of AMD treatment growth. The number of treatments for
`AMD in preclinical testing, early clinical testing, or clinical practice
`has undergone exponential growth during the past 5 years.
`
`(GA) exhibit DNA strand breaks and lipoperoxida-
`tion.3 Antioxidant changes in the retinal pigment
`epithelium (RPE) of AMD eyes indicate that the RPE
`cells are under oxidative stress (e.g., increased levels
`of heme oxygenase-1 and heme oxygenase-2 and Cu-Zn
`superoxide dismutase).4 Advanced glycation end
`products occur in soft drusen, basal laminar and basal
`linear deposits, and the cytoplasm of RPE cells asso-
`ciated with choroidal neovascularization (CNV).5,6
`Carboxymethyl lysine is present in drusen and CNV5,7
`as are carboxyethyl pyrrole protein adducts.5 Addi-
`tionally, Fe2+—which is an essential element for
`enzymes involved in the phototransduction cascade,
`outer segment disk membrane synthesis, and conver-
`sion of all-trans-retinyl ester
`to 11-cis-retinol
`in
`RPE—also catalyzes the conversion of hydrogen per-
`oxide to hydroxyl radicals and is known to accumulate
`in Bruch’s membrane in AMD eyes.8,9 Epidemiologic
`studies indicate that one of the main risk factors for
`AMD is smoking, which is known to cause oxidative
`damage. One interpretation of the Age-Related Eye
`
`From the *Institute of Ophthalmology and Visual Science,
`University of Medicine and Dentistry of New Jersey, New Jersey
`Medical School, Newark, New Jersey; and †Bascom Palmer Eye
`Institute, University of Miami, Miller School of Medicine, Miami,
`Florida.
`M. A. Zarbin received grant support from the Lincy Foundation,
`Foundation Fighting Blindness, National Eye Institute, Advanced
`Cell Technology, Research to Prevent Blindness, Janice Mitchell
`Vassar and Ashby John Mitchell Fellowship, Joseph J. and
`Marguerite DiSepio Retina Research Fund, the New Jersey Lions
`Eye Research Foundation, and the Eye Institute of New Jersey. P. J.
`Rosenfeld received grant support from National Eye Institute,
`Alexion Pharmaceuticals, Othera Pharmaceuticals, Carl Zeiss
`Meditec, Potentia Pharmaceuticals, and CoMentis.
`M. A. Zarbin is a consultant to Novartis, Genentech, Wyeth/
`Pfizer, Lilly, and Bausch and Lomb. P. J. Rosenfeld is serving on
`the study advisory boards of Othera Pharmaceuticals, GlaxoS-
`mithKline, Sanofi-Aventis, Oraya, Potentia Pharmaceuticals, and
`Bristol-Myers Squibb.
`Institute of
`Reprint
`requests: Marco Zarbin, MD, PhD,
`Ophthalmology and Visual Science, New Jersey Medical School,
`Doctors Office Center, Room 6156, 90 Bergen Street, Newark, NJ
`07103; e-mail: zarbin@umdnj.edu
`
`Mylan Exhibit 1076
`Mylan v. Regeneron, IPR2021-00880
`Page 4
`
`

`

`1352 RETINA, THE JOURNAL OF RETINAL AND VITREOUS DISEASES  2010  VOLUME 30  NUMBER 9
`
`Fig. 2. The visual cycle. Re-
`produced with
`permission
`from http://lpi.oregonstate.edu/
`infocenter/vitamins/vitaminA/
`visualcycle.html. Courtesy of
`Jane Higdon, Linus Pauling
`Institute, Oregon State Uni-
`versity, copyright 2010.
`
`disease progression. Zinc inhibits C3 convertase
`activity,32 and levels of C3a des Arg, which is a cleavage
`product of C3a and reflects complement activation, are
`higher in patients with AMD (including patients with
`early as well as late AMD) versus controls.33 We are not
`aware of published data demonstrating that zinc
`supplements lower C3a des Arg levels in AMD patients.
`Fourth, drusen, GA, and CNV are associated with
`mutations in components of the complement pathway,
`which is part of the innate immune system (Figure 3).
`Protective and risk-enhancing mutations in compo-
`nents of the complement pathways have been reported
`and include the following loci: complement factor H
`(CFH), complement component 2 (C2), factor B
`(CFB), complement component 3 (C3), and factor I
`(CFI).27,35–47
`Fifth, oxidative damage can compromise regulation
`of the complement system by RPE cells. Thurman and
`Holers48 noted that the alternative complement pathway
`is continuously activated in the fluid phase, and tissue
`surfaces require continuous complement inhibition to
`prevent spontaneous autologous cell injury. Sohn et al49
`demonstrated that the complement system is continu-
`ously activated in the eye. Thurman et al50 showed that
`oxidative stress reduces the regulation of complement
`on the surface of ARPE-19 cells (i.e., reduces surface
`expression of the complement inhibitors, decay accel-
`erating factor [CD55] and CD59) and impairs comple-
`ment regulation at the cell surface by factor H. Sublytic
`activation of the complement cascade also causes VEGF
`release from the cells, which compromises RPE barrier
`function. Similarly, oxidative stress can reduce the
`ability of interferon-gamma to increase CFH expression
`in RPE cells.51 In vitro evidence indicates that products
`of the photooxidation of A2E in RPE cells can serve as
`a trigger for the complement system.52 Thus, the relative
`abundance of lipofuscin in the submacular RPE may
`predispose the macula to chronic inflammation and
`
`AMD, particularly in patients who cannot control
`complement activation because of inherited abnormal-
`ities in the complement system. Hollyfield et al53 have
`described an animal model that links oxidative damage
`and complement activation to AMD.
`Sixth, some AMD-risk enhancing mutations not
`directly involving the complement pathway are also
`linked to inflammation or oxidative damage.54–59
`A proposed pathogenesis (Figure 4) of AMD suggests
`the possibility of therapeutic intervention at different
`points in the natural history of
`the disease with
`antioxidants, visual cycle inhibitors, antiinflammatory
`agents, antiangiogenic agents, and neuroprotective agents.
`
`Treatment
`
`Antioxidants
`
`The AREDS did not show a statistically significant
`benefit of the AREDS formulation for either the
`development of new GA or the involvement of the
`fovea in eyes with preexisting atrophy.60 In part, this
`result may be because of the paucity of GA patients in
`the study. AREDS II
`(http://clinicaltrials.gov/ct2/
`show/NCT00345176?term=Age-Related+Eye+Disease+
`Study+%28AREDS%29&rank=1)
`is a randomized,
`multicenter, clinical trial to assess 1) the role of lutein
`(10 mg)/zeaxanthin (2 mg) and omega-3 long-chain
`polyunsaturated fatty acids (docosahexaenoic acid
`[DHA]/eicosapentaenoic acid [EPA]) in prevention of
`development of GA or CNV; and 2) the possible
`deletion of beta-carotene and lowering the daily zinc
`oxide dose to 25 mg. A Phase 3 clinical trial is underway.
`A recently terminated Phase 2 clinical study, known as
`the OMEGA Study (Othera, Pharmaceuticals Inc.,
`Conshohocken, PA) (http://clinicaltrials.gov/ct2/show/
`NCT00485394?term=OMEGA+Study&rank=1),inves-
`tigated an eyedrop with a prodrug, known as OT 551
`
`Mylan Exhibit 1076
`Mylan v. Regeneron, IPR2021-00880
`Page 5
`
`

`

`PATHWAY-BASED THERAPIES FOR AMD  ZARBIN AND ROSENFELD
`
`1353
`
`3. The
`Fig.
`complement
`pathway. Modified with per-
`mission from Donoso et al.2
`Components of the comple-
`ment system in which mu-
`tations have been associated
`with increased or decreased
`risk of drusen, GA, and CNV
`are circled. The coagulation
`system-activated
`intrinsic
`pathway is not shown.34
`
`(4-cyclopropanoyloxy-1-hydroxy-2,2,6,6-tetramethypi-
`peridine HCL), to treat GA. This prodrug penetrates the
`eye well and is converted to the active drug (TEMPOL-H),
`which has antioxidant, antiinflammatory (down regu-
`lates nuclear factor k-B), and antiangiogenic properties
`in preclinical models. OT 551 failed to slow the
`enlargement rate of GA after 18 months. We do not
`know if the failure to demonstrate a treatment benefit is
`because of inadequate posterior segment drug delivery
`or because of its mechanism of action.
`
`Visual Cycle Modulators
`
`Visual cycle modulators are intended to reduce the
`accumulation of toxic fluorophores (e.g., A2E) and
`lipofuscin in RPE cells. Retinol binding protein (RBP)
`possesses a high-affinity binding site for all-trans-
`retinol. The binding of retinol to RBP, in turn, creates
`a high-affinity binding site for transthyretin (TTR).
`
`Binding of TTR to the RBP–retinol complex creates
`a large molecular complex that resists filtration in the
`kidney and permits a high steady-state concentration of
`retinol in the circulation, which facilitates delivery of
`retinol to extrahepatic target tissues such as the eye.
`Unlike other extrahepatic tissues, the eye demonstrates
`a unique preference for uptake of retinol when it is
`presented in the RBP–TTR complex. N-(4-hydroxy-
`phenyl) retinamide (Fenretinide; Sirion Therapeutics,
`Inc, Tampa, FL) displaces all-trans-retinol from RBP in
`blood. Fenretinide possesses a bulky side chain on its
`terminal end that prevents interaction of the complex
`with TTR. In the absence of TTR binding, the RBP–
`fenretinide complex is eliminated through glomerular
`filtration (excreted in urine) because of its relatively
`small size. Thus, fenretinide treatment causes a dose-
`dependent reversible reduction in circulating RBP and
`retinol. The unique requirement of the eye for retinol
`delivered by RBP renders the eye more susceptible to
`
`Mylan Exhibit 1076
`Mylan v. Regeneron, IPR2021-00880
`Page 6
`
`

`

`1354 RETINA, THE JOURNAL OF RETINAL AND VITREOUS DISEASES  2010  VOLUME 30  NUMBER 9
`
`Fig. 4. Proposed pathophys-
`iology of AMD and locations
`in the pathway in which
`different
`therapeutic inter-
`ventions might be effective.
`Modified from Zarbin, M,
`Sunness JS. Dry age-related
`macular degeneration and
`age-related macular degener-
`ation pathogenesis. In: Levin
`LA, Albert DM, eds. Ocular
`Disease: Mechanisms
`and
`Management. China: Saunders
`(Elsevier); 2010:527–535.
`
`in women with breast cancer were 1) qualitative
`interaction between age and treatment duration and 2)
`plasma retinol.64 A trend toward mild inhibition of
`retinal photoreceptor function after prolonged duration
`of intervention was observed in the older women.64
`
`Fig. 5. Microscopic analysis of lipofuscin autofluorescence and cy-
`tostructure of the retina. Tissue sections were prepared from the eyes of
`ABCA42/2 albino and pigmented mice that had been treated with either
`DMSO or fenretinide (HPR) (10 mg/kg) for 42 days. Sections from
`albino mice were analyzed by fluorescence microscopy, while sections
`from pigmented mice were used for light microscopy. A. Analysis of
`lipofuscin autofluorescence revealed considerable accumulation within
`the RPE of DMSO-treated mice. B. In contrast, HPR-treated mice
`showed significantly reduced levels of lipofuscin fluorophores. C.
`Tissue sections prepared from an age-matched and strain-matched wild-
`type mouse are shown for comparison. Analysis of RPE and retina
`cytostructure by light microscopy revealed no aberrant morphology
`associated with either DMSO or HPR treatment (not shown). OS, outer
`segment. Reproduced with permission from Radu et al.61
`
`reductions in serum RBP retinol compared with other
`tissues. Consequently, during chronic fenretinide
`administration, levels of retinol within the eye will be
`reduced dramatically while other extrahepatic tissues
`will obtain retinol from alternate sources. Fenretinide
`reduces lipofuscin and A2E accumulation in the RPE of
`ABCA42/2 mice and causes modest delays in dark
`adaptation (Figure 5).61 A Phase 2 clinical trial of this
`oral agent is underway. Patients receive placebo, 100-
`mg, or a 300-mg dose. Interim analyses reported at
`scientific meetings suggest a possible therapeutic effect
`from the drug, but the results are preliminary. Regarding
`possible side effects from fenretinide, we note that
`fenretinide has been used in clinical trials for cancer
`therapy and prevention of malignant neoplasms.62–68 In
`these studies, which involve patients ranging in age from
`approximately 30 years to 60 years, the incidence of
`acquired night blindness ranges from 2% to 20%, with
`a median of approximately 15%.65,67–72 The incidence
`of dry eye ranges from 3% to 53%, with a median of
`approximately 5%.65,67,69,70,72 Typically, only a minority
`(approximately ,5%) of patients had to discontinue
`therapy because of
`these side effects. Reversible
`dark-adaptation changes
`and electroretinogram
`abnormalities were associated with fenretinide chemo-
`therapy (800 mg/day) for basal cell carcinoma73 and for
`breast cancer (200 mg/day),74 which is associated with
`a decline in plasma retinol concentration.75 Among
`breast cancer patients (mean age, 48 years) using 200
`mg/day,
`the changes in night vision were rarely
`symptomatic.76 Predictive factors with a significant
`effect on the electroretinogram at a dose of 200 mg/day
`
`Mylan Exhibit 1076
`Mylan v. Regeneron, IPR2021-00880
`Page 7
`
`

`

`PATHWAY-BASED THERAPIES FOR AMD  ZARBIN AND ROSENFELD
`
`1355
`
`Aging and obesity are risk factors for diminished night
`vision because of a strong association with lower plasma
`retinol concentrations.75 We emphasize, however, that
`the clinical trial in progress will determine whether
`observed benefits outweigh these potential side effects.
`Another visual cycle modulator is 13-cis-retinoic
`acid (Accutane, Hoffmann-La Roche, Inc., Nutley, NJ),
`which inhibits the conversion of all-trans-retinyl esters
`(in retinosomes) to 11-cis-retinol and the conversion of
`11-cis-retinol to 11-cis-retinal by retinol dehydrogenase
`and also reduces lipofuscin accumulation in ABCA42/2
`mice.77 This oral agent may be associated with a high
`nyctalopia.78 All-trans-retinylamine
`incidence
`of
`(ACU-4429; Acucela, Seattle, WA)
`is an orally
`administered compound that
`inhibits conversion of
`all-trans-retinyl ester to 11-cis-retinol via blockade
`of RPE65 or another protein needed for isomerization
`of all-trans-retinol. ACU-4429 also reduces lipofuscin
`and A2E accumulation in the RPE of ABCA42/2 mice.
`Because this molecule works as an enzyme inhibitor
`(rather than by reducing availability of precursor, thus
`reducing rhodopsin formation via mass action kinetics),
`its effects should last longer than fenretinide, thus
`permitting less frequent dosing. However, there may be
`greater risk of side effects, such as nyctalopia. Retinoids
`and farnesyl-containing isoprenoids (TDT and TDH)
`also block RPE65.
`Although the use of beta-carotene in the AREDS
`formulation and attempts to block the visual cycle as
`a treatment for AMD may seem contradictory, it is not
`clear that these treatment approaches are antagonistic.
`Normally, beta-carotene is metabolized to retinalde-
`hyde. Relatively high-dose beta-carotene supplemen-
`tation (not vitamin A) was used in the AREDS. In low
`doses, beta-carotene can act as an antioxidant.79 High
`doses of beta-carotene can reduce retinoic acid levels,
`possibly via stimulation of cytochrome P450 activity
`because of
`the formation of eccentric cleavage
`products (vs. the central cleavage of beta-carotene,
`which forms 2 retinal molecules).80 In addition, a free
`radical–rich environment also favors the formation of
`eccentric cleavage products, cytochrome P450 stim-
`ulation, and local retinoic acid deficiency. Thus, it is
`possible that in the local environment of the outer
`retina–RPE–Bruch membrane–choroid,
`fenretinide
`and beta-carotene may not have completely antago-
`nistic effects.
`
`Antiinflammatory Agents
`
`Corticosteroids have a number of antiangiogenic
`effects (Table 1). They have been used previously as
`sole treatment and as part of combination treatment for
`CNV.81 Iluvien (Alimera Sciences, Alpharetta, GA) is
`
`Table 1. Some Antiangiogenic Effects of Corticosteroids
`
`Induce capillary basement membrane dissolution (in
`growing capillaries).
`Alter the behavior of inflammatory cells that stimulate
`angiogenesis.
`Inhibit bFGF-stimulated choroidal endothelial cell
`migration and tube formation.
`Inhibit bFGF-induced activation of matrix
`metalloproteinase-2.
`Reduce oxidative stress–induced VEGF messenger RNA
`expression in ARPE-19 cells.
`Alter intercellular adhesion molecule expression of
`nonendothelial cells.
`Reduce blood–retinal barrier breakdown in rabbit eyes.
`Inhibit platelet-derived growth factor–induced VEGF
`expression.
`Reduce numbers of microglia in AMD-associated
`choroidal new vessels.
`
`Modified from Zarbin.81
`
`a nonbioerodible polyimide tube containing 180 mg
`of the corticosteroid fluocinolone acetonide. It is inserted
`via a 25-gauge intravitreal
`injector, which creates
`a self-sealing wound. A Phase 2 study is underway
`involving 40 patients with bilateral GA, and the primary
`outcome is a difference in the enlargement rate of GA in
`treated versus untreated eyes. The study eye is
`randomized to high (0.5 mg/day) or low (0.2 mg/day)
`dose Iluvien. The fellow eye serves as a control.
`A number of agents that modulate different parts of
`the complement system are in Phase 1 and Phase 2
`clinical trials (Figure 6). In general, these agents work
`either by replacing a defective complement component
`(e.g., providing normal factor H to patients with Y402H
`mutations) so that complement activation can be
`modulated properly or by blocking the complement
`pathway (e.g., POT-4, which inhibits C3). Several
`examples will be discussed because they illustrate some
`of the challenges associated with manipulation of this
`pathway.
`lectin, and alternative pathways
`The classical,
`generate bioactive fragments C3a and C5a and the
`membrane attack complex (C5b,6,7,8,9) via C3
`cleavage. As a result, C3 inhibition should be very
`effective at blocking complement activation that arises
`from many different mutations involving the comple-
`ment system (thus, targeting a relatively large popula-
`tion of AMD patients), which should be a therapeutic
`advantage. However, this degree of complement in-
`hibition may create risks such as an increased risk of
`injection-associated endophthalmitis.
`In a murine
`model, it seems that C3 deficiency does not increase
`the risk of Staphylococcus aureus endophthalmitis.82
`Conversely, in a guinea pig model, complement deple-
`tion with cobra venom factor does seem to increase the
`risk of S. aureus, Staphylococcus epidermidis, and
`
`Mylan Exhibit 1076
`Mylan v. Regeneron, IPR2021-00880
`Page 8
`
`

`

`1356 RETINA, THE JOURNAL OF RETINAL AND VITREOUS DISEASES  2010  VOLUME 30  NUMBER 9
`
`Fig. 6. Treatment of AMD
`through management
`of
`complement
`abnormalities.
`Depiction
`of
`complement
`pathways (on the right) is
`modified with
`permission
`from Donoso et al.2 Circles
`and the two red arrows in-
`dicate parts of the pathway
`that are targets of current
`therapy (listed on the left).
`
`Pseudomonas aeruginosa endophthalmitis.83,84 POT-4
`(Potentia Pharmaceuticals, Louisville, KY), a cyclic
`peptide of 13 amino acids that
`is a derivative of
`Compstatin, is a C3 inhibitor and is administered by
`intravitreal
`injection. An attractive feature of
`this
`preparation is that gel-like deposits will form in the
`vitreous when POT-4 is injected at high concentrations.
`These deposits last at least 6 months, thus providing
`a sustained-release delivery system. It is not known
`whether the doses administered intravitreally will have
`systemic effects, but a Phase 1 study of POT-4 in AMD
`eyes with CNV was completed successfully without
`any safety concerns (http://clinicaltrials.gov/ct2/show/
`NCT00473928?term=POT-4&rank=1).
`Inhibition of C5 is attractive because terminal
`complement activity is blocked, but proximal comple-
`ment
`functions
`remain intact,
`for example, C3a
`anaphylatoxin production, C3b opsonization, and im-
`mune complex and apoptotic body clearance. ARC1905
`(Ophthotech Corp., Princeton, NJ) is an anti-C5 aptamer
`delivered by intravitreal injection. It is in Phase 1 trials
`for
`nonexudative
`(http://clinicaltrials.gov/ct2/show/
`NCT00950638?term=ARC-1905&rank=1) and exuda-
`tive complications of AMD (http://clinicaltrials.gov/ct2/
`show/NCT00709527?term=ARC-1905&rank=2). Ecu-
`lizumab (SOLIRIS, Alexion Pharmaceuticals, Cheshire,
`CT) is a humanized monoclonal antibody that blocks C5
`and is administered intravenously. Eculizamab is
`already Food and Drug Administration–approved for
`the treatment of paroxysmal nocturnal hemoglobinuria
`and is in Phase 2 trials for treatment of nonexuda-
`tive complications of AMD (http://clinicaltrials.gov/ct2/
`show/NCT00935883?term=eculizumab&rank=2). C5a
`receptor blockade, for example, JPE1375 (Jerini AG,
`Berlin, Germany); PMX025 (Arana Therapeutics,
`
`Sydney, Australia); Neutrazimab (G2 Therapies, Dar-
`linghurst, New South Wales, Australia), might have an
`advantage or a disadvantage over direct C5a inhibition.
`C5a receptor blockade might inhibit some important
`inflammatory pathways31 without preventing membrane
`attack complex formation.
`Replacement of CFH should inhibit inflammation
`in AMD patients with risk-enhancing mutations in
`CFH. It is not clear whether patients with other
`mutations will benefit from this therapy. An attractive
`feature of this approach, which might require genetic
`screening before treatment,
`is that
`there is no
`increased risk of infection because we have innate
`systems that permit CFH to modulate C3 activation
`locally. The recombinant human form of the full-
`length CFH protein in its ‘‘protective’’ form is known
`as rhCFHp (Ophtherion, Inc, New Haven, CT). This
`protein can be administered intravenously or intra-
`vitreally. In preclinical models, intravitreal adenovi-
`ral vector delivery of the CFH gene has been effective
`and offers the promise of a sustained delivery system.
`(Our understanding is that Ophtherion, Inc., is not
`going to continue with its
`rhCFHp program.)
`Replacement of defective CFH is also being de-
`veloped by Taligen (TT30, a recombinant fusion
`protein, Taligen Therapeutics, Cambridge, MA)
`(http://www.taligentherapeutics.com/pipeline/index.
`html). Taligen is also exploring factor B inhibition
`using a humanized antibody fragment (TA106).
`Gene therapy to silence genes by preventing
`messenger RNA expression might be useful
`for
`treatment of AMD because deletion of genes closely
`related to CFH (i.e., CFHR1 and CFHR3) seems to be
`strongly protective against AMD.38 However, short-
`interfering RNA therapies in the eye may be toxic,85
`
`Mylan Exhibit 1076
`Mylan v. Regeneron, IPR2021-00880
`Page 9
`
`

`

`PATHWAY-BASED THERAPIES FOR AMD  ZARBIN AND ROSENFELD
`
`1357
`
`and it seems that the deletion of CFHR1 and CFHR3
`protects against development of AMD at least in part
`because the deletion tags a protective haplotype and
`does not occur in association with the Y402H single-
`nucleotide polymorphism.86
`Sirolimus (rapamycin; Macusight/Santen, Union City,
`CA)
`is a macrolide fungicide that
`targets mTOR
`(mammalian target of rapamycin) and is antiinflamma-
`tory, antiangiogenic, and antifibrotic; mTOR is a protein
`kinase that regulates proliferation, motility, survival, and
`protein synthesis. Rapamycin can be administered
`subconjunctivally and was in Phase 1/2 studies in
`patients with GA (http://clinicaltrials.gov/ct2/show/
`NCT00766649?term=rapamycin&cond=Macular+
`Degeneration&rank=3) as well as in monotherapy
`(http://clinicaltrials.gov/ct2/show/NCT00712491?term=
`rapamycin&cond=Macular+Degeneration&rank=1) and
`combination therapy trials with ranibizumab for exuda-
`tive
`complications
`of AMD (http://clinicaltrials.
`gov/ct2/show/NCT00766337?term=rapamycin&cond=
`Macular+Degeneration&rank=2). Glatiramer acetate
`(Copaxone; TEVA, Petach Tikva,
`Israel)
`induces
`glatiramer acetate–specific suppressor T cells and
`downregulates inflammatory cytokines.
`It can be
`administered subcutaneously and is in Phase 2
`and Phase 3 studies in patients with drusen (http://
`clinicaltrials.gov/ct2/show/NCT00466076?term=copax-
`one&rank=10). A small, randomized, controlled study
`demonstrated efficacy after 12 weeks of subcutaneous
`injections.87 It remains to be shown whether drusen
`disappearance, the end point of this study, represents
`an appropriate surrogate end point for long-term visual
`acuity preservation in AMD eyes. The Complications
`of Age-Related Macular Degeneration Prevention
`(CAPT) trial demonstrated no long-term visual benefit
`to laser photocoagulation-induced drusen resorption.88
`The mechanism of action of
`the two treatment
`modalities, however, is fundamentally different. Laser
`treatment induces inflammation, and glatiramer ace-
`tate is antiinflammatory.
`Amyloid-b oligomers are toxic to cells (soluble
`monomers are not). Amyloid diseases typically exhibit
`abundant fibrils of various lengths. These fibrils are an
`end product of stepwise protein/peptide misfolding,
`and they accumulate as long-lived extracellular deposits.
`Drusen vesicles probably contain fibrillar amyloid
`composed in part

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