throbber
Series Editors: D. Neil Granger & Joey Granger
`
`COLLOQUIUMSERIES IN
`INTEGRATED SYSTEMS PHYSIOLOGY:
`FROM MOLECULE TO FUNCTION
`
`Angiogenesis
`
`Thomas H.Adair
`Jean-Pierre Montani
`
`LIFE SCIENCES
`
`WS MORGAN & CLAYPOOL LIFE SCIENCES
`
`

`

`Angiogenesis
`
`Samsung et al. v. Regeneron IPR2023-00884
`Regeneron Pharmaceuticals, Inc. Exhibit 2136 Page 2
`
`

`

`Samsung et al. v. Regeneron IPR2023-00884
`Regeneron Pharmaceuticals, Inc. Exhibit 2136 Page 3
`
`

`

`
`
`Editors
`D. Neil Granger, Louisiana State University Health Sciences Center-Shreveport
`Joey P. Granger, University ofMississippi Medical Center
`Physiology is a scientific discipline devoted to understanding the functions of the body. It addresses
`function at multiple levels, including molecular, cellular, organ, and system. An appreciation of the
`processes that occur at each level is necessary to understand function in health and the dysfunc-
`tion associated with disease. Homeostasis and integration are fundamental principles ofphysiology
`that accountfor the relative constancy of organ processes and bodily function even in the face of
`substantial environmental changes. This constancy results from integrative, cooperative interactions
`of chemical andelectrical signaling processes within and betweencells, organs and systems. This
`eBookseries on the broad field of physiology covers the major organ systems from an integra-
`tive perspective that addresses the molecular andcellular processes that contribute to homeostasis.
`Material on pathophysiology is also included throughout the eBooks. The state-of the-art treatises
`were produced by leading experts in the field of physiology. Each eBook includes stand-alone in-
`formation andis intended to be of value to students, scientists, and clinicians in the biomedical
`sciences. Since physiological concepts are an ever-changing work-in-progress, each contributorwill
`have the opportunity to make periodic updates of the covered material.
`
`Publishedtitles
`(for future titles please see the website, www.morganclaypool.com/page/lifesci)
`
`IPR2023-00884
`Samsung et al. v. Regeneron
`Regeneron Pharmaceuticals, Inc. Exhibit2136
`Page 4
`
`

`

`Copyright © 2011 by Morgan & Claypool Life Sciences
`
`All rights reserved. No part of this publication may be reproduced, stored in a retrieval system, or transmitted in
`any form or by any means—electronic, mechanical, photocopy, recording, or any other except for brief quotations
`in printed reviews, without the prior permission of the publisher.
`
`Angiogenesis
`Thomas H. Adair and Jean-Pierre Montani
`www.morganclaypool.com
`
`ISBN: 9781615043309 paperback
`
`ISBN: 9781615043316 ebook
`
`DOI: 10.4199/C00017ED1V01Y201009ISP009
`
`A Publication in the
`
`CoLLoqUiUM SerieS on inTeGrATeD SYSTeMS PHYSioLoGY: FroM MoLeCULe To
`FUnCTion To DiSeASe
`
`Lecture #10
`
`Series Editors: Joey Granger, University of Mississippi Medical Center and D. Neil Granger, Louisiana State
`University Health Sciences Center-Shreveport
`
`Series ISSN
`ISSN 2154-560X
`ISSN 2154-5626
`
`print
`electronic
`
`Samsung et al. v. Regeneron IPR2023-00884
`Regeneron Pharmaceuticals, Inc. Exhibit 2136 Page 5
`
`

`

`Angiogenesis
`
`ThomasH.Adair
`University ofMississippi Medical Center
`
`Jean-Pierre Montani
`University of Fribourg, Switzerland
`
`COLLOQUIUM SERIES ON INTEGRATEDSYSTEMS PHYSIOLOGY:
`FROM MOLECULETO FUNCTIONTO DISEASE #10
`
`Ne MORGAN QCLAYPOOL LIFE SCIENCES
`
`IPR2023-00884
`Samsung et al. v. Regeneron
`Regeneron Pharmaceuticals, Inc. Exhibit2136
`Page6é
`
`

`

`ABSTRACT
`Angiogenesis is the growth ofblood vessels from theexisting vasculature. Thefield of angiogenesis
`has grown enormouslyin the past 30 years, with only 40 papers published in 1980 and nearly 6000
`in 2010. Whyhas there been this explosive growth in angiogenesis research? Angiogenic therapies
`provide a potential to conquer cancer, heart diseases, and more than 70oflife’s most threatening
`medical conditions. Thelives ofat least 1 billion people worldwide could be improved with angio-
`genic therapy, according to the Angiogenesis Foundation. In this little book, we provide a simple
`approach to understandtheessential elements of the angiogenic process, we critique the most pow-
`erful angiogenesis assays that are used to discover proangiogenic and antiangiogenic substances,
`and weprovide an in-depth physiological perspective on how angiogenesis is regulated in normal,
`healthy tissues of the human body. All tissues of the body require a continuous supply of oxygen to
`burn metabolic substrates that are needed for energy. Oxygen is conducted to these tissues by blood
`capillaries: more capillaries can improve tissue oxygenation and thus enhance energy production;
`fewer capillaries can lead to hypoxia and even anoxia in the tissues. This means that angiogenic
`therapies designed to control the growth and regression ofbloodcapillaries can be used to improve
`the survival of poorly perfusedtissues that are essential to the body (heart, brain, skeletal muscle,
`etc.) and to rid the body of unwantedtissues (tumors).
`
`
`
`IPR2023-00884
`Samsung et al.v. Regeneron
`Regeneron Pharmaceuticals, Inc. Exhibit2136
`Page7
`
`

`

`
`
`Theauthorsare grateful for the editorial assistance of Karen A. Richards and Leslie S. Adair. This
`work was supported by a grant from the National Heart, Lung, and Blood Institute (HL-51971).
`
`IPR2023-00884
`Samsung et al. v. Regeneron
`Regeneron Pharmaceuticals, Inc. Exhibit2136
`Page 8
`
`

`

`Samsung et al. v. Regeneron IPR2023-00884
`Regeneron Pharmaceuticals, Inc. Exhibit 2136 Page 9
`
`

`

`
`
`Overview ofAngiogenesis ...........ssccsssesssecesseeesseceeeeeceeceseeeseseeeesseceeeeceeeseneeeeeseeeees 1
`LL History oo... ceeeeececceesesescseeceseeeecsesesensnenececessesessnsnsnsceenssesecseseseseusnecececeesesesanensanenecess 1
`1.2 Origin of Blood Vessels ..........s:ssssssssesesessesesesesesesceeeecscseseseseseesesceceesaeeeesesensanenecess 2
`1.3 The Angiogenic Process .........sssscsssecsssseseseesscscscssseseesescseesescecsssneessesesnsesseeseeeseees 3
`1.3.1 Types ofAngiogenesis .........c.scscccessesssescsssesessscsesesesescscecseseecseececseaneeeeecaees 3
`1.3.2 Sprouting Angiogenesis...........scscesssscssseseesseseseeceeseseseececsesesceeseecsesneceeeaees 4
`1.3.3
`Intussusceptive Angiogenesis...........cccesescsceseseseeesesessssesceceeeseseecseseeenenesees 6
`
`ATIQIOGCTICRIS ASBBYB 50...ocscscss0cscecsccsssscessovansvseecssecseessusseusonensssescssoosnsesonecusosecsseoses 9
`2.1
`In Vitro Assays.......ccccessecsssscsssesescececsesesescscecsnsesnesesessssnesesesesensnssesesansneessseessneneeees 9
`2.1.1 Endothelial Cells Are Heterogeneous..........c:c:ccesesesesessesesesesesescesnsceceeeees 9
`2.1.2 In Vitro Conditions Rarely Reflect In Vivo Environment..............0:000+ 9
`2.1.3 Endothelial Cell Proliferation Assays..........sssssssssesssssesesssesesescsnscseeees 10
`2.1.4 Endothelial Cell Migration Assays.........:cccsssssesesescscsesesesesesesssenseeeeees 11
`2.1.5 Endothelial Tube Formation Assays ........cscscsessssseeesesceseeeseseeceeesseeeees 12
`2.1.6 Rat and Mouse Aortic Ring Assay ........cccccsssessesesescscesesesesesesesseenseeeeees 14
`In Vivo Assay......cscscesesesesesesssseececeeseseseaesescscesessssecsesesesessnecacecseseassnseseneneseceses 15
`2.2.1 Corneal Angiogenesis Assay....
`veces 15
`2.2.2 Chick Chorioallantoic Membrane(CAM)‘AngiogenesisAssay...
`.. 16
`2.2.3 Matrigel Plug Assay ........s.scscsesccssesssesesesssnsnseseesenencessssesescesesseseseeeesasaeees 17
`
`2.2
`
`Regulation: Metabolic Factors ..........::scssscsssseesseeessecerseeceseseeeecsseeeseeceeeeeeeeeeneees 19
`3.1 Capillary Growth Is Proportional to Metabolic Activity.........ccccsseseeseseeeeeees 19
`3.2
`Increasing Metabolic Activity Stimulates Blood Vessel Growth... 20
`3.3. Decreasing Metabolic Activity Causes Vascular Regression....
`ssssesenes 22
`3.4 Long-Term Increases in Blood Pressure Lead to Vascular Rarefaction..
`23
`3.5 Oxygen Is a Master Signal in Growth Regulation ofthe Vascular Systenn 24
`3.5.1
`Increases in Muscular Activity Cause Decreases in
`Muscle Oxygemation..........cscccsssesssssesssescssscessesescscecsesesnscececsnsneceeseeeneneess24
`3.5.2 Oxygen Regulates Angiogenic Growth Factor Production............:000+ 24
`
`IPR2023-00884
`Samsung et al. v. Regeneron
`Regeneron Pharmaceuticals, Inc. Exhibit2136
`Page 10
`
`

`

`x ANGIoGENESIS
`
`4.
`
` VEGF-A Released From Hypoxic Tissues Is a Key Regulator
`3.5.3
`of Angiogenesis ..................................................................................... 25
`
`3.5.4 Negative Feedback Regulation of VEGF-A .......................................... 26
`3.5.5
` Oxygen Plays a Central Role in Feedback Regulation of Vascular
`
`Growth and Regression ......................................................................... 27
`3.6 Role of Adenosine in Metabolic Regulation of Vascular Growth ...................... 28
`
`Regulation: Mechanical Factors ......................................................................... 31
`4.1 Control of Blood Vessel Growth ....................................................................... 31
`4.1.1 Epithelial Sodium Channel Protein Biology ......................................... 31
`4.1.2 Epithelial Sodium Channels Can Form a Mechanosensory
`
`Complex ................................................................................................ 32
`4.1.3
` Epithelial Sodium Channels Can Mediate Mechanotransduction in
`
`Mammals ............................................................................................... 32
`4.1.4 Do Epithelial Sodium Channels Mediate Angiogenesis? ...................... 33
`4.1.5 Physical Forces Acting on the Walls of Blood Vessels ........................... 33
`4.1.6 Shear Stress Is Sensed by the Endothelium ........................................... 34
`4.1.7 Increased Blood Flow (Shear Stress) Can Stimulate Angiogenesis ....... 35
`4.1.8
` Possible Role of Endothelial Cell Shape in Regulating Blood Vessel
`
`Growth and Regression ......................................................................... 36
`4.1.9 Mechanical Factors Have an Accessory Role in Angiogenesis .............. 37
`4.2 Control of Lymphangiogenesis ......................................................................... 38
`4.2.1 Flow-Guided Lymphangiogenesis ........................................................ 39
`4.2.2 High Salt Load Stimulates Lymphangiogenesis in Skin ....................... 40
`
`Glossary .................................................................................................................... 41
`
`References ................................................................................................................. 47
`
`Author Biographies .................................................................................................... 71
`
`Samsung et al. v. Regeneron IPR2023-00884
`Regeneron Pharmaceuticals, Inc. Exhibit 2136 Page 11
`
`

`

`CHAPTER 1
`
`
`
`Angiogenesis is the growth of bloodvessels from the existing vasculature. It occurs throughoutlife
`in both health and disease, beginning in utero and continuing on through old age. No metabolically
`active tissue in the body is more than a few hundred micrometers from a bloodcapillary, which is
`formedbythe process of angiogenesis. Capillaries are neededin all tissues for diffusion exchange of
`nutrients and metabolites. Changes in metabolic activity lead to proportional changes in angiogen-
`esis and, hence, proportional changes in capillarity. Oxygen plays a pivotal role in this regulation.
`Hemodynamicfactors are critical for survival ofvascular networks andforstructural adaptations of
`vesselwalls.
`
`Recognition that control of angiogenesis could have therapeutic value has stimulated great
`interest during the past 40 years. Stimulation of angiogenesis can be therapeutic in ischemic heart
`disease, peripheral arterial disease, and woundhealing. Decreasingor inhibiting angiogenesis can be
`therapeutic in cancer, ophthalmic conditions, rheumatoidarthritis, and other diseases. Capillaries
`grow andregress in healthytissues according to functional demands. Exercise stimulates angiogen-
`esis in skeletal muscle andheart. A lack of exercise leads to capillary regression. Capillaries grow
`in adipose tissue during weight gain and regress during weight loss. Clearly, angiogenesis occurs
`throughoutlife.
`
`1.1
`HISTORY
`The Scottish anatomist and surgeon John Hunterprovided thefirst recorded scientific insights into
`the field of angiogenesis. His observations suggested that proportionality between vascularity and
`metabolic requirements occurs in both health and disease. This belief is summarized in his Treatise
`published in 1794 [1] as follows: “In short, whenever Nature has considerable operations going on,
`andthose are rapid, then wefind the vascular system in a proportionable degree enlarged.” Although
`the term angiogenesis does not appear in his writings [1,2], Hunter was the first to recognize that
`overall regulation of angiogenesis follows a basic law of nature founded byAristotle [3], which in es-
`sence is “form follows function.” The modern history of angiogenesis began with the work ofJudah
`Folkman, who hypothesized(and published in 1971) that tumor growth is angiogenesis-dependent
`[4]. Recognition that control of angiogenesis could lead to cancer therapies stimulated intensive
`
`IPR2023-00884
`Samsung et al. v. Regeneron
`Regeneron Pharmaceuticals, Inc. Exhibit2136
`Page 12
`
`

`

`2 ANGIoGENESIS
`
`research in the field, e.g., only two manuscripts dealing with angiogenesis were published in 1970 and
`over 5200 articles were published in 2009. For detailed histories of angiogenesis, see Refs. [5–11].
`
`1.2 oRIGIN oF BLooD VESSELS
`The cardiovascular system is the first organ system to develop in the embryo [12]. The luminal
`surface of the circulatory system in contact with blood is a single layer of
`: these
`are derived from
` (Figure 1.1).
` differentiate from
`
`and give rise to
` and
`. Angioblasts are a cell type with potency
`to differentiate into endothelial cells but have not yet acquired all characteristic markers of endo-
`thelial cells.
` (Figure 1.2) is the de novo formation of blood vessels from
`
`[12–14]. It occurs in the extraembryonic and intraembryonic tissues of embryos [12,14]. Vasculo-
`genesis is a dynamic process that involves cell–cell and cell–
` (ECM) interactions
`directed spatially and temporally by
` and
` [14–17]. This process includes
`differentiation of mesodermal stem cells into angioblasts, growth factor directed migration of angio-
`blasts to form
` where angioblasts give rise to
` [12–14].
`.
`,
`Other types of vascular growth include
`, and
`The term
` means the formation of any blood vessel in the adult regardless of its
`size or type.
`
`FIGURE 1.1: Origin of endothelial cells and hematopoietic cells [14]. Mesodermal stem cells are the
`source of hematopoietic stem cells and angioblasts in the developing embryo. The hemangioblast is a
`precursor to both angioblasts and hematopoietic stem cells. Angioblasts differentiate into endothelial
`cells. Hematopoietic cells can differentiate into all cell types found in circulating blood
`
`Samsung et al. v. Regeneron IPR2023-00884
`Regeneron Pharmaceuticals, Inc. Exhibit 2136 Page 13
`
`endothelial cells
`
`mesoderm
`
`Hemangioblasts
`
`mesodermal stem cells
`
`hematopoietic stem cells
`
`angioblasts
`
`Vasculogenesis
`
`angioblasts
`
`extracellular matrix
`
`growth factors
`
`morphogens
`
`blood islands
`
`endothelial cells
`
`neovascularization
`
`arteriogenesis
`
`venogenesis
`
`lymphangiogenesis
`
`arteriogenesis
`
`arteriogenesis
`
`venogenesis
`
`venogenesis
`
`lymphangiogenesis
`
`lymphangiogenesis
`
`

`

`oVERVIEw oF ANGIoGENESIS 3
`
`FIGURE 1.2: Vasculogenesis in the vertebrate embryo. (a) Angioblasts derived from lateral mesoderm
`are committed to become arteries (red) or veins (blue). The cardinal veins assemble from precursor cells
`(blue) that remain in a lateral position. (b) Artery precursor cells migrate toward a vascular endothelial
`growth factor type A (
`) stimulus secreted from cells in the midline. (c) The migrating arterial
`angioblasts align into cords forming a plexus. (d) Arterial angioblasts coalesce forming the dorsal aorta.
`(e) Intersomite vessels are assembled from three types of endothelial cells with different morphologies
`indicated as blue, purple, and green. Used with permission from Nature Publishing Group: Hogan
`(2002) [18].
`
`THE ANGIoGENIC PRoCESS
`1.3
`1.3.1 Types of Angiogenesis
`Sprouting angiogenesis and intussusceptive angiogenesis both occur in utero and in adults. Sprout-
`ing angiogenesis is better understood having been discovered nearly 200 years ago: intussusceptive
`angiogenesis was discovered by Burri [19,20] about two decades ago. Figure 1.3 shows the basic
`morphological events for both types of angiogenesis. As implied by its name, sprouting angiogenesis
`is characterized by sprouts composed of endothelial cells, which usually grow toward an angiogenic
`stimulus such as VEGF-A. Sprouting angiogenesis can therefore add blood vessels to portions of
`tissues previously devoid of blood vessels. On the other hand, intussusceptive angiogenesis involves
`formation of blood vessels by a splitting process in which elements of interstitial tissues invade exist-
`ing vessels, forming transvascular tissue pillars that expand. Both types of angiogenesis are thought
`to occur in virtually all tissues and organs.
`
`Samsung et al. v. Regeneron IPR2023-00884
`Regeneron Pharmaceuticals, Inc. Exhibit 2136 Page 14
`
`VEGF-A
`
`

`

`4 ANGIoGENESIS
`
`FIGURE 1.3: Basic types of primary vascular growth. Redrawn after Carmeliet and Collen (2000) [21].
`
`1.3.2 Sprouting Angiogenesis
`The basic steps of sprouting angiogenesis include enzymatic degradation of capillary basement
`membrane, endothelial cell (EC) proliferation, directed migration of ECs, tubulogenesis (EC tube
`formation), vessel fusion, vessel pruning, and pericyte stabilization. Sprouting angiogenesis is ini-
`tiated in poorly perfused tissues when oxygen sensing mechanisms detect a level of
` that
`demands the formation of new blood vessels to satisfy the metabolic requirements of
`
` (Figure 1.4). Most types of parenchymal cells (myocytes, hepatocytes, neurons, astrocytes, etc.)
`respond to a hypoxic environment by secreting a key proangiogenic growth factor called vascular
`endothelial growth factor (VEGF-A). There does not appear to be redundant growth factor mecha-
`nisms that can replace the role of VEGF-A in hypoxia-induced angiogenesis.
`An endothelial
` guides the developing capillary sprout through the ECM toward an
`angiogenic stimulus such as VEGF-A [22–25]. Long, thin cellular processes on tip cells called
` secrete large amounts of proteolytic enzymes, which digest a pathway through the ECM
`for the developing sprout [26,27]. The filopodia of tip cells are heavily endowed with VEGF-A
`receptors (
`), allowing them to “sense” differences in VEGF-A concentrations and causing
`them to align with the VEGF-A gradient (Figure 1.5). When a sufficient number of filopodia on
`a given tip cell have anchored to the substratum, contraction of actin filaments within the filopodia
`literally pull the tip cell along toward the VEGF-A stimulus. Meanwhile, endothelial
`
`proliferate as they follow behind a tip cell causing the capillary sprout to elongate. Vacuoles develop
`and coalesce, forming a lumen within a series of stalk cells. These stalk cells become the trunk of the
`newly formed capillary. When the tip cells of two or more capillary sprouts converge at the source of
`VEGF-A secretion, the tip cells fuse together creating a continuous lumen through which oxygen-
`ated blood can flow. When the local tissues receive adequate amounts of oxygen, VEGF-A levels
`
`Samsung et al. v. Regeneron IPR2023-00884
`Regeneron Pharmaceuticals, Inc. Exhibit 2136 Page 15
`
`hypoxia
`
`parenchymal
`
`cells
`
`tip cell
`
`filopodia
`
`VEGFR2
`
`stalk cells
`
`

`

`oVERVIEw oF ANGIoGENESIS 5
`
`FIGURE 1.4: VEGF-A directed capillary growth to poorly perfused tissues. (A) Endothelial cells ex-
`posed to the highest VEGF-A concentration become tip cells (green). Hypoxic tissue is indicated by
`the circular blue fade. (B) The tip cells lead the developing sprout by extending numerous filopodia.
`(C) The developing spout elongates by proliferation of endothelial stalk cells (purple) that trail behind
`the tip cell. (D) The tip cells from two developing sprouts fuse and create a lumen. (E) Blood flowing
`through the new capillary oxygenates the tissues, thus reducing the secretion of VEGF-A. (F) The newly
`developed capillary is stabilized by pericyte recruitment (red), deposition of ECM (gray), shear stress and
`other mechanical forces associated with blood flow and blood pressure. Redrawn after Carmeliet et al.
`(2009) [24].
`
`
`
`return to near normal. Maturation and stabilization of the capillary requires recruitment of
`and deposition of ECM along with
` and other mechanical signals [28].
`Delta-Notch signaling is a key component of sprout formation (Figure 1.5). It is a cell–cell sig-
`naling system in which the ligand,
` (Dll4) mates with its
` on neighboring
`cells. Both the receptor and ligand is cell bound and thus act only through cell–cell contact. VEGF-A
`induces Dll4 production by tip cells, which leads to activation of notch receptors in stalk cells.
`Notch receptor activation suppresses VEGFR2 production in stalk cells, which dampens migratory
`behavior compared with that of tip cells. Hence, endothelial cells exposed to the highest VEGF-A
`concentration are most likely to become tip cells [24,25,30]. Although tip cells are exposed to the
`highest VEGF-A concentration, their rate of proliferation is far less compared with that of stalk
`cells.
`
`Samsung et al. v. Regeneron IPR2023-00884
`Regeneron Pharmaceuticals, Inc. Exhibit 2136 Page 16
`
`pericytes
`
`shear stress
`
`Delta-like-4
`
`notch receptor
`
`

`

`6 ANGIoGENESIS
`
`FIGURE 1.5: Microanatomy of a capillary sprout and tip cell selection. (A) An interstitial gradient for
`VEGF-A and an endothelial cell gradient for VEGFR2 are shown. Tip cell migration is thought to de-
`pend upon the VEGF-A gradient and stalk cell proliferation is thought to be regulated by the VEGF-A
`concentration. Redrawn after Carmeliet and Tessier-Lavigne (2005) [29]. (B) Delta-Notch signaling is
`critical for tip cell selection. Activation of notch receptors on stalk cells induces proteolytic cleavage and
`release of the intracellular domain, which enters the nucleus and decreases gene expression of VEGFR2.
`National Institutes of Health, public domain image.
`
`Not all aspects of the Delta-Notch signaling pathway are fully understood, but it is clear
`that production of a normal vasculature is heavily dependent upon the concentration of VEGF-A
`in the tissues. A 50% reduction of VEGF-A expression is lethal embryonically because of vascular
`defects [31,32], and excess VEGF-A in tumors induces overproduction of tip cells leading to a dis-
`organized vasculature [33]. This critical dependence on physiological concentrations of VEGF-A
`for construction of viable blood vessels might help explain why attempts to induce angiogenesis
`in poorly perfused tissues with VEGF-A administration and gene therapy have not been highly
`successful.
`
`1.3.3 Intussusceptive Angiogenesis
`Intussusceptive angiogenesis is also called splitting angiogenesis because the vessel wall extends into
`the lumen causing a single vessel to split in two. This type of angiogenesis is thought to be fast and
`efficient compared with sprouting angiogenesis because, initially, it only requires reorganization of
`existing endothelial cells and does not rely on immediate endothelial proliferation or migration.
`Intussusceptive angiogenesis occurs throughout life but plays a prominent role in vascular develop-
`
`Samsung et al. v. Regeneron IPR2023-00884
`Regeneron Pharmaceuticals, Inc. Exhibit 2136 Page 17
`
`

`

`oVERVIEw oF ANGIoGENESIS 7
`
`ment in embryos where growth is fast and resources are limited [34–36]. However, intussusception
`mainly causes new capillaries to develop where capillaries already exist.
`Evidence for the occurrence of intussusceptive angiogenesis is based upon the presence of
`transcapillary tissue pillars (Figure 1.6). Identification of tissue pillars requires scanning electron
`micrographs of vascular casts or three-dimensional reconstruction of serial micrographs. This type
`of angiogenesis was discovered in postnatal lungs of rats and humans [19,20], but it also occurs in
`many other tissues and organs, especially in capillary networks that abut an epithelial surface, e.g.,
`choroid of the eye, vascular baskets around glands, intestinal mucosa, kidney, ovary, and uterus
`[37,38]. It also occurs in skeletal muscle, heart, and brain. In addition to forming new capillary
`structures, intussusceptive growth plays a major role in the formation of artery and vein bifurcations
`as well as pruning of larger microvessels.
`The control of intussusceptive angiogenesis is poorly understood compared with sprouting
`angiogenesis. This difference is only partly due to its recent discovery in 1986 [20]. A rate-limiting
`step in intussusceptive growth research can be pinned to the laborious methods required to
`prove its presence, which, again, involve determining the frequency of tissue pillars from scan-
`ning electron micrographs of vascular casts. However, it is known that intussusceptive angiogenesis
`can be stimulated in the chick
` (CAM) with application of VEGF-A
`(Figure 1.7), and there is little doubt that many growth factors and signaling systems are involved
`[34,37]. Mechanical stresses related to increases in blood flow can initiate intussusceptive growth in
`some high flow regions of the circulation, as discussed in Chapter 4 [34,35].
`
`FIGURE 1.6: Scanning electron micrographs of Mercox casts. (a) Fetal chicken lung microvasculature.
`(b) Rat lung microvasculature at postnatal day 44. The small holes indicated by arrows have diameters
`of about 2 µM. The holes correspond to tissue pillars that extend across the capillary lumina. Scale bars:
`(a) 12 and (b) 20 µM. Used with permission from Wiley-Blackwell: Djonov, Kurz, and Burri (2003) [35].
`
`Samsung et al. v. Regeneron IPR2023-00884
`Regeneron Pharmaceuticals, Inc. Exhibit 2136 Page 18
`
`chorioallantoic membrane
`
`

`

`8 ANGIoGENESIS
`
`FIGURE 1.7: Intussusceptive angiogenesis in three dimensions (a–d) and two dimensions (a'–d'). (a,b,
`a',b') The process begins with protrusion of opposing endothelial cells into the capillary lumen. (c,c') An
`interendothelial contact is established and endothelial junctions are reorganized. (d,d') The endothelial
`(EC) bilayer and basement membranes (BM) are perforated centrally allowing growth factors to enter.
`Fibroblasts (Fb) and pericytes (Pr) migrate into the site of perforation where they produce collagen
`fibrils (Co) and other components of ECM forming a tissue pillar. Used with permission from Wiley-
`Blackwell: Djonov, Kurz, and Burri (2003) [35].
`
`• • • •
`
`Samsung et al. v. Regeneron IPR2023-00884
`Regeneron Pharmaceuticals, Inc. Exhibit 2136 Page 19
`
`

`

`CHAPTER 2
`
`
`
`The most significant advancements in angiogenesis in the past 25 years have been the discovery
`of proangiogenic and antiangiogenic molecules. The development of angiogenesis assays has been
`essential to the discovery of these molecules. In vitro assays have been expeditious and quantitative
`but should be viewedasfirst approximationsthat need to be confirmedin vivo. In vivo tests are more
`time-consuming anddifficult to quantitate, but because of the complex interactions between mul-
`tiple cell types necessary to form functional bloodvessels,all in vitro findings need to be confirmed
`in the intact animal.
`
`INVITRO ASSAYS
`2.1.
`2.1.1 Endothelial Cells Are Heterogeneous
`Most endothelial cell assays utilize human umbilical vein endothelial cells (HUVECs) or bovine
`aortic endothelial cells (BAECs) not becausethesecells are good representatives ofvascular endo-
`thelial cells in vivo, but because they arerelatively easy to harvest from large blood vessels. Endo-
`thelial cells are heterogeneous [39-43]: there are differences among endothelial cells from large and
`small blood vessels, arteries, and veins, species differences, organ differences, differences between
`host and tumor, andeven differences within a given organ. Also, endothelial cells used in laboratories
`are virtually alwaysin a proliferative state rather than the normal quiescentstate of the established
`vasculature in the intact animal. Even most “primary cultures” of endothelial cells require extensive
`proliferation to obtain enoughcells for an experiment with the assumption that they retain their
`normal in vivo physiological characteristics. This assumption is often incorrect. It is well-known
`that cells in vitro can both gain andlose attributes compared with parentcells in intact animals.
`
`2.1.2 In Vitro Conditions Rarely Reflect In Vivo Environment
`Anotherissue is the environmental conditions in which endothelial cells are cultured. Endothelial
`cells in vivo are exposed to shear stress and other hemodynamic forces that activate multiple sig-
`naling pathways. Endothelial cells in vitro are usually cultured in room air (21% oxygen), which
`is hyperoxic compared with the in vivo oxygen tension, especially in the microcirculation. Two-
`and three-dimensional scaffolds using Matrigel, collagen,or fibrin are used to simulate the normal
`
`IPR2023-00884
`Samsung et al. v. Regeneron
`Regeneron Pharmaceuticals, Inc. Exhibit2136
`Page 20
`
`

`

`10
`
` ANGIoGENESIS
`
`FIGURE 2.1: (a–c) Human umbilical vein endothelial cells (HUVEC) and (d–f ) human dermal micro-
`vascular endothelial cells (HuDMEC). (a, d) Phase contrast microscopy. (b, e) CD31 or (c, f ) von Wille-
`brand factor staining: nuclei are counterstained with DAPI. Photographs courtesy of Promocell GmbH,
`Heidelberg, Germany. Used with permission from Wiley-Blackwell: Staton et al. (2009) [42].
`
`extracellular matrix, but it is clear that complex interactions between endothelial cells and their in
`vivo physical environment cannot be fully simulated in culture. Also, in vivo interactions between
`endothelial cells and other cell types (smooth muscle cells, pericytes, fibroblasts, macrophages, etc.)
`are difficult to simulate in vitro. For these reasons, in vitro angiogenesis assays should be viewed as
`a starting point rather than an endpoint for discovery, depending on the purpose of the experiment
`(Figure 2.1).
`
`2.1.3 Endothelial Cell Proliferation Assays
`Proliferation of endothelial cells is needed for developing capillaries in the intact animal. The ac-
`tions of proangiogenic and antiangiogenic molecules on proliferation can be assessed by direct cell
`counts, DNA synthesis, or metabolic activity [39–42,44]. For testing potential proangiogenic mol-
`ecules, it is often necessary to reduce the rate of proliferation by decreasing serum levels in culture
`media, and it is usually more effective to test antiangiogenic molecules on cells that have a substan-
`tial rate of proliferation.
`The rate of cell proliferation can be determined by counting cells at 24-h intervals after seed-
`ing multiple cultures with a defined number of cells (Figure 2.2). Cells can be counted using a hemo-
`cytometer and light microscope or an electronic Coulter counter or similar device. Cell proliferation
`is often determined using a colorimetric method (MTT assay) in which mitochondrial enzymes
`
`Samsung et al. v. Regeneron IPR2023-00884
`Regeneron Pharmaceuticals, Inc. Exhibit 2136 Page 21
`
`

`

`ANGIoGENESIS ASSAyS 11
`
`FIGURE 2.2: Typical growth curve for HUVECs in culture media containing 10% fetal bovine se-
`rum (FBS). Media were changed daily. Cells were counted using a Coulter counter. Redrawn after Lee
`(2006) [44].
`
`reduce MTT to formazan dyes in proportion to cell number. MTT (3-(4,5-dimethylthiazol-2-yl)-
`2,5-diphenyltetrazolium bromide) is a yellow tetrazole that is reduced to purple formazan in living
`cells: absorbance is read by a spectrophotometer. DNA synthesis is often used to reflect cell prolif-
`eration. With the commonly used [3H]thymidine incorporation method, the amount of radioactiv-
`ity in cells is proportional to the amount of newly synthesized DNA. A similar but nonradioactive
`method utilizes bromodeoxyuridine (BrdU), which competes with thymidine for incorporation into
`DNA. Despite their popularity, these latter methods are not fully reliable. Changes in metabolic
`rates of individual cells and/or compounds that affect mitochondrial enzyme activity can cause gross
`miscalculations of cell proliferation with the MMT assay, and several investiga

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