angiogenesis and antiangiogenesis in the neonate relevance to retinopathy of prematurity

Upload: andreamoscosocontreras

Post on 02-Jun-2018

216 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/10/2019 Angiogenesis and Antiangiogenesis in the Neonate Relevance to Retinopathy of Prematurity

    1/8

    Angiogenesis and Antiangiogenesisin the Neonate: Relevance to Retinopathyof PrematurityAshima Madan, MD*

    Objectives After completing this article, readers should be able to:1. Delineate the major stimulus for growth of retinal blood vessels by angiogenesis in

    utero.

    2. Describe the pathogenesis of retinal neovascularization in preterm infants.

    3. List the angiogenic factors that regulate growth of new blood vessels.

    4. List potential antiangiogenesis therapies.

    IntroductionThe molecular mechanisms underlying new blood vessel growth currently are being

    investigated in several laboratories. Angiogenesis or neovascularization has been impli-

    cated in various unrelated disease processes, such as retinopathy of prematurity (ROP),diabetic retinopathy, choroidal neovascularization, macular degeneration, and tumor

    angiogenesis. On the other hand, growth of new blood vessels is desired and beneficial in

    wound healing and myocardial and limb ischemia. All of these unrelated conditions likely

    share several common mechanisms in the final pathway that culminates in angiogenesis.

    Identification of a number of growth factors along with the ability to manipulate the

    mouse embryo genetically has resulted in increased understanding of the molecular

    mechanisms regulating angiogenesis. This review summarizes current knowledge of the

    factors regulating new blood vessel growth and its relevance to retinal neovascularization

    in preterm infants.

    Vascularization of the Retina and Retinopathy of PrematurityThe outer layers of the retina are supplied by the choroid plexus, which lies between theoutermost layer of the retina and the retinal pigment epithelium (RPE). The inner layers

    are supplied by a superficial plexus beneath the inner limiting membrane and a deep plexus

    in the inner nuclear layer (Figs 1 and 2). Vascularization of the retina in utero, where

    arterial oxygen tension is less than 30 mm Hg, occurs by a process of vasculogenesis and

    angiogenesis. Vasculogenesis, the de novo formation of capillaries from endothelial cells

    that have differentiated from mesodermal precursors at approximately 16 weeks gestation

    in the posterior region around the optic disc, is seen only during embryonic development.

    Angiogenesis, the formation of blood vessels from existing blood vessels, begins around

    25 weeks gestation in the foveal region and is responsible for increasing vascular density of

    the superficial plexus and formation of the deep plexus.

    Newly formed blood vessels spread across the surface of the retina following the central

    peripheral gradient of retinal ganglion cell maturation toward the peripheral retina. Bloodvessels initially develop in the superficial plexus, followed by growth radially outward into

    the deep plexus only as far as the junction between the inner nuclear and peripheral layers

    (Fig. 2). Physiologic hypoxia, created by the increased metabolic demands of the fetal

    retina, leads to the release of vascular endothelial growth factor (VEGF) from neural

    cellsthe Muller cells in the inner nuclear layer and the astrocytes in the ganglion cell

    layer. VEGF acts in a paracrine manner and is the major stimulus for the growth of retinal

    blood vessels by angiogenesis in utero.

    Several lines of evidence indicate that vasculogenesis in the human retina is independent

    of metabolic demand and hypoxia-induced VEGF expression. First, VEGF expression has

    not been detected in the human retina before 20 weeks gestation. Second, neuronal

    *Associate Professor of Pediatrics, Stanford University School of Medicine, Stanford, CA.

    Article ophthalmology

    e356 NeoReviews Vol.4 No.12 December 2003

  • 8/10/2019 Angiogenesis and Antiangiogenesis in the Neonate Relevance to Retinopathy of Prematurity

    2/8

    maturation is greatest around 15 to 18 weeksgestation

    in the perifoveal region, which is avascular at this stage of

    gestation. Third, although VEGF knockout mice form

    vessels by vasculogenesis, such vessels are very abnormal.

    Although there is considerable variation in the time

    course of retinal vascularization among infants, approxi-mately 70% of the retina is vascularized at 27 weeks

    gestation; complete vascularization occurs by 36 weeks

    gestation on the nasal side and by 40 weeks gestation on

    the temporal side. The susceptibility of preterm infants of

    a younger gestational age to retinopathy compared with

    more mature infants is due to the presence of a larger

    region of avascular retina. The question of why the retina

    exclusively is affected by administered oxygen has per-

    plexed scientists. It has been hypothesized to be due to

    the unique relationship between the retinal and choroi-

    dal blood vessels as well as the unusual properties of the

    choroidal vasculature.The pathogenesis of retinal neovascularization in pre-

    term infants occurs in two phases. In the first phase,

    hyperoxia produces vasoconstriction and vaso-

    obliteration of developing retinal blood vessels. This

    phase likely represents an exaggerated natural protective

    response to a surplus of oxygen. The second phase is

    initiated with concomitant development of the photore-

    ceptor retinal layer with advancing postnatal age and the

    resulting increase in oxygen requirements. Photorecep-

    tors have a high metabolic demand because they need to

    resynthesize photosensitive pigment continuously, con-

    stantly rebuild their cilia, and maintain membrane mech-

    anisms for neural signaling. It is interesting to note that

    retinal consumption of oxygen is much higher in the dark

    than in the light. Retinal hypoxia, created by the inability

    of the vaso-obliterated blood vessels to meet the meta-

    bolic demands of the retina, results in the release of

    angiogenic growth factors and subsequent revasculariza-tion. However, this exuberant blood vessel growth is

    pathologic; the blood vessels tend to be leaky and friable,

    resulting in hemorrhage and exudation followed byfi-

    broplasia and ultimately retinal detachment and loss of

    vision. Although it has been suggested that the fibropla-

    sia may be caused by either invasion of the vitreous by

    mesenchymal precursors or the effect of fibroblast

    growth factor, the cause offibroplasia in ROP remains

    unknown.

    Angiogenesis

    Angiogenesis is a complex process that begins with mi-gration of inflammatory cells toward the hypoxic or

    inflammatory signal and the release of angiogenic factors

    (Table 1). Endothelial cells release proteolytic enzymes

    such as matrix metalloproteinases that disrupt the vascu-

    lar basement membrane and extracellular matrix. Vascu-

    lar adhesion molecules enhance adhesion of the endothe-

    lial cells to the extracellular matrix. The endothelial cells

    then migrate in the form of a solid column called the

    vascular sprout toward the angiogenic stimulus. By pro-

    ducing proteolytic activity, the advancing cell column

    migrates through the extracellular matrix. Behind the

    advancing region, cells begin to differentiate, adhere to

    Table 1.Angiogenic Factors Regulating Growth of New Blood Vessels

    Angiogenic Factors Function

    Vascular endothelial growth factor (VEGF) Endothelial cell (EC)-specific mitogen, prevents apoptosisBasic fibroblast growth factor, hepatocyte growth

    factor (HGF), tumor necrosis factor, nuclearfactor-kappaB, interleukin-8

    Increase proliferation of endothelial cells, potentiate effect ofVEGF (HGF)

    Placental growth factor-1 Selective agonist for Flt-1Hypoxia-inducible factor-1 alpha Transcription factor, upregulates several angiogenic factors in

    response to hypoxiaMetalloproteinases Proteases that degrade endothelial cell matrix (ECM)Alpha v beta 3 and alpha v beta 5 integrins Vascular adhesion molecules mediating binding of EC to ECME-Selectin Mediates cell-to-cell contactPlatelet-derived growth factor Recruits pericytesTransforming growth factor-beta Stabilizes blood vessels by induction of pericyte

    differentiation, basement membrane productionFlk-1, Flt-1, neuropilin VEGF tyrosine kinase (TR) receptors, initiate signaltransduction cascade

    Tie1 and Tie2 TR receptors for angiopoietinsAngiopoietins Ligands for Tie2, remodelling and stabilization of vessels

    ophthalmology angiogenesis

    NeoReviews Vol.4 No.12 December 2003 e357

  • 8/10/2019 Angiogenesis and Antiangiogenesis in the Neonate Relevance to Retinopathy of Prematurity

    3/8

    one another, form a new basement membrane, and de-

    velop a lumen to form a new capillary. Anastomosis of

    adjacent sprouting vessels establishes the vascular supply

    to the region. Formation of the early vascular network is

    followed by several maturation steps involving attraction

    of smooth muscle cells and pericytes around the endo-

    thelium as well as vascular remodeling involving changes

    in lumen diameter and vessel wall thickness to accommo-

    date the needs of the local tissue.

    Angiogenesis is a staged process that is tightly con-

    trolled by a balance between angiogenic and angiostatic

    factors. The endothelial cell growth factor family consists

    of the vascular endothelial growth factor and angiopoi-

    etin groups. All factors in this complex pathway must

    function in coordination to form functional vessels.

    Vascular Endothelial Growth FactorVEGF is the key growth factor driving ocular blood

    vessel growth. Both Michaelson and Ashton had pro-

    posed in the early 1950s that an angiogenic factor was

    released from the retina. However, it was not until the

    1980s that VEGF was identified as the key molecule in

    angiogenesis. VEGF-A exists in at least four different

    isoformsVEGF 121, VEGF 165, VEGF 189, and

    VEGF 206 that are produced by alternate splicing of

    the VEGF mRNA. VEGF binds to tyrosine kinase cell

    surface receptors, VEGF receptor-1 or Flt-1, and VEGFreceptor-2 or Flk-1. Neuropilin-1, a neuronal cell recep-

    tor that mediates neuronal guidance, has been identified

    as a coreceptor for VEGF and is critically involved in

    vascular development.

    The distribution of Flt-1 and Flk-1 receptors differs

    markedly during normal retinal development, and the

    receptors have different roles in endothelial cell prolifer-

    ation and differentiation. In situ hybridization and im-

    munohistochemistry experiments in mice that are 5 days

    postpartum have shown that the Flt-1 protein colocalizes

    with retinal vessels, with Flk-1 detectable only in the

    neural retina. These studies showed a 60-fold inductionof Flt-1 from postnatal day 3 to day 26, but no significant

    change in Flk-1 expression. Mice deficient in Flt-1 and

    Flk-1 died in utero between postnatal days 8.5 and 9.5.

    Flt-1 knockout mice developed disordered vascularity

    due to overgrowth of endothelial cells; mice that had

    targeted disruption of the Flk-1 gene failed to develop a

    vasculature and had very few endothelial cells.

    VEGF gene expression is regulated by oxygen ten-

    sion. Expression increases in response to hypoxia and is

    downregulated by hyperoxia. The mechanism of

    hypoxia-inducible expression is mediated at least partially

    via hypoxia-inducible factor-1 alpha (HIF-1-alpha), a

    transcription factor that transactivates several hypoxia-

    inducible genes.

    Several lines of evidence indicate that VEGF is the key

    molecule involved in development of normal blood ves-

    sels as well as in retinopathy. First, knockout mice that

    have disruption of even one allele of the VEGF gene have

    impaired blood vessel formation. On the other hand,

    transgenic mice that have overexpression of VEGF in the

    photoreceptor layer develop intraretinal and subretinal

    neovascularization. Increased expression of VEGF

    mRNA is seen anterior to the developing blood vessels

    on postnatal day 7 in the normal mouse retina (retinal

    vascularization in mice occurs after birth and is complete

    by 14 days). Second, VEGF levels are increased in the

    vitreous of patients who have diabetic retinopathy and

    other neovascular disorders of the eye as well as in a

    primate model of iris neovascularization. In situ hybrid-

    ization experiments have localized the site of production

    of VEGF during the process of neovascularization to the

    inner nuclear layer of the retina. The attenuation of

    blood vessels that is seen in the first phase of hyperoxia-

    induced retinal vessel loss can be inhibited by intravitreal

    administration of exogenous VEGF in animal models.

    Conversely, the increase in VEGF expression and neo-

    vascularization seen in the second phase of retinopathy

    can be inhibited in the mouse model by use of antisense

    oligonucleotides, receptor binding chimeric proteins,and monoclonal antibodies. Thus, VEGF is not just a

    mitogen; it also acts as an endothelial cell survival factor

    by preventing apoptosis of endothelial cells. This survival

    effect appears to be developmentally regulated. VEGF is

    not required for the maintenance of blood vessels in the

    adult vasculature.

    Current knowledge suggests that under normoxic

    conditions, VEGF is produced at a maintenance level

    adequate to support existing blood vessels. During hyp-

    oxia, VEGF levels increase above this level to induce

    growth of new blood vessels. Under hyperoxic condi-

    tions, VEGF levels decrease below the maintenance leveland lead to attenuation of existing blood vessels.

    Angiogenic FactorsOther angiogenic factors, such asfibroblast growth fac-

    tor (FGF), transforming growth factors (TGF) alpha and

    beta, hepatocyte growth factor, tumor necrosis factor-

    alpha, and interleukin-8 (IL-8), also are involved in

    angiogenesis. Although basic fibroblast growth factor

    (bFGF, or FGF2) previously was implicated in ocular

    neovascularization, FGF2-deficient mice developed the

    same degree of neovascularization as control animals in

    the oxygen-induced mouse model of retinopathy. IL-8

    ophthalmology angiogenesis

    e358 NeoReviews Vol.4 No.12 December 2003

  • 8/10/2019 Angiogenesis and Antiangiogenesis in the Neonate Relevance to Retinopathy of Prematurity

    4/8

    levels are increased in the vitreous fluid of patients who

    have proliferative diabetic retinopathy. Nuclear factor-

    kappaB (NF-kappaB) and a rat homolog of IL-8 are

    increased in endothelial and glial cells in oxygen-induced

    retinopathy. Knockout mice lacking the hypoxia-

    inducible factor-2 alpha gene (HIF-2 alpha) do not

    develop neovascularization when exposed to the

    hyperoxia/normoxia paradigm used to generate mice

    that have retinopathy. These mice have decreased levels

    of erythropoietin (Epo), indicating a role for Epo in

    experimental ROP.

    ProteolysisBinding of VEGF to the receptor is followed by degra-

    dation of the endothelial cell basement membrane by

    proteases that degrade collagen and other extracellular

    matrix components and disrupt the basement membrane

    barrier, thus enabling endothelial cells to migrate from

    the vessels and proliferate. Expression of protease genes

    is induced by cytokines and angiogenic growth factors

    such as bFGF and VEGF-A. The matrix metalloprotein-

    ases (MMP) are a family of proteases that selectively

    degrade extracellular matrix. They consist of secreted

    and membrane-associated endopeptidases that act on

    various substrates. The endothelial sprout must traverse

    several types of extracellular matrixes during the process

    of growth: first the basement membrane, then thefibrinogen-rich provisional matrix created by leakiness of

    the surrounding vessels, and finally the collagen and

    fibronectin-rich intersitial matrix. MMP activity helps in

    this process. The most convincing evidence for the role

    of MMP in angiogenesis is that use of MMP inhibitors

    inhibits in vivo and in vitro angiogenic responses. Also,

    MMP-deficient mice have a delayed angiogenic response

    during development. Although there is consensus that

    MMP activity clearly is implicated in angiogenesis, their

    precise role in this process and the details of their inter-

    action with other endothelial cell functions (see endothe-

    lial cell adhesion) is not yet defined completely. Someevidence suggests that MMPs may help to produce an-

    giogenesis inhibitors such as angiostatin. Thus, MMP

    activity possibly has a dual role (facilitating angiogenesis

    and releasing angiogenesis inhibitors) in new blood ves-

    sel growth.

    Endothelial Cell Adhesion and MigrationVascular cell adhesion molecules such as integrins alpha v

    beta 3 and alpha v beta 5 are critical to the formation and

    maintenance of the newly formed blood vessel. The

    integrins are a family of heterodimeric transmembrane

    receptors consisting of an alpha and beta subunit that

    each recognizes a unique set of extracellular matrix li-

    gands. They assist in the migration of endothelial cells by

    mediating binding of the endothelial cell to the extracel-

    lular matrix. Integrin-mediated adhesion initiates several

    intracellular signaling events that regulate cell survival

    and division. Inhibition of either matrix hydrolysis or

    integrin/ligand interaction can inhibit normal and

    pathologic retinal blood vessel formation. Integrin alpha

    v beta 3 is a receptor for a wide variety of extracellular

    ligands and is expressed at high levels on activated endo-

    thelial cells. Studies suggest that alpha v beta 3 can bind

    MMP-2 and localize the active form of this enzyme on

    the surface of angiogenic vessels, thus enabling the en-

    dothelial cells to degrade the extracellular matrix during

    invasion. However, the relationship between endothelial

    cells and the extracellular matrix is complex, and further

    studies are needed to elucidate the signaling pathway

    induced by interaction between vascular adhesion mole-

    cules and endothelial cells.

    It has been difficult to reconcile the results of studies

    using monoclonal antibodies to block alpha v beta 3 or

    alpha v beta 5 integrins with the results of knockout mice

    experiments with integrin-deficient mice. Inhibition of

    integrin function with monoclonal antibodies prevented

    angiogenesis in animal models. Conversely, in knockout

    mice experiments, beta 3-negative mice contained nor-

    mal brain and intestinal blood vessels, suggesting thatbeta 3 is not essential for angiogenesis. Only 20% of alpha

    v integrin knockout mice survive to term, but these

    animals die within a few hours due to brain and intestinal

    blood vessel abnormalities and hemorrhages, indicating

    that the alpha v unit is important for angiogenesis. One

    possibility is that other adhesion molecules in the knock-

    out mouse model,such as alpha v beta 5,may takeover in

    the absence of alpha v integrins. However, this has not

    been confirmed. Another possibility is that monoclonal

    antibodies also directly inhibit the effect of other inte-

    grins.

    Migration and proliferation of cells is followed byformation of tubes that have a patent lumen. E-selectin,

    a glycoprotein that mediates endothelial cell-cell con-

    tacts, is important for lumen formation.

    Remodeling and Stabilization of BloodVesselsThe final steps in the process involve remodeling and

    stabilization of the blood vessels by recruitment of mes-

    enchymal cells and differentiation to pericytes. Platelet-

    derived growth factor (PDGF), which exists in three

    isoforms, plays a role in pericyte recruitment. PDGF-B-

    and PDGF receptor-deficient mice do not have pericytes

    ophthalmology angiogenesis

    NeoReviews Vol.4 No.12 December 2003 e359

  • 8/10/2019 Angiogenesis and Antiangiogenesis in the Neonate Relevance to Retinopathy of Prematurity

    5/8

    in their blood vessels and develop microaneurysms, thus

    suggesting that endothelial cells of the vascular sprouts

    are unable to recruit local mural cells in the absence of

    PDGF. Transforming growth factor-beta (TGF-beta) is

    one of a large family of growth factors that is implicated

    in cellular growth and differentiation. TGF-beta-

    deficient embryos have delayed-to-absent vasculogenesis

    as well as inadequate contact between endothelial and

    mesothelial cells. It is speculated that TGF-beta acts at

    multiple steps to stabilize blood vessel formation via

    several actions that include inhibition of endothelial cell

    proliferation and migration as well as induction of peri-

    cyte differentiation. TGF also can alter the integrin pro-

    files and stimulate basement membrane production.

    Angiopoietins and Tyrosine Kinase ReceptorsIn addition to the receptors for VEGF, the tyrosine

    kinase immunoglobulin and epidermal growth factor-

    like extracellular domains (Tie) receptors, Tie1 and Tie2,

    which are expressed by endothelial cells, are another

    group of tyrosine kinase receptors that play a critical role

    in embryonic angiogenesis. Both Tie1 and Tie2 are

    required for structural integrity of endothelial cells; mice

    deficient in Tie1 die during the perinatal period due to

    respiratory difficulties. Tie2-deficient mouse embryos do

    not survive beyond embryogenesis day 9.5 to 10.5 and

    have prominent abnormalities in the vasculature.The angiopoietins are ligands for Tie2. Angiopoietin-1

    (Ang-1) is associated with developing blood vessels. This

    activating ligand induces tyrosine phosphorylation of

    Tie2 in endothelial cells, sprouting of capillaries, and

    survival of endothelial cells. Disruption of the Ang-1

    gene in mouse embryos produces a phenotype that is very

    similar to Tie2-deficient mice. These animals develop a

    vasculature but have disordered remodeling of blood

    vessels and lack periendothelial supporting cells. In dia-

    betic retinopathy, it is possible that Ang-1 could be used

    in the future as an agent to prevents leakiness of blood

    vessels. Ang-2, a natural antagonist of Ang-1, is a desta-bilizing factor that promotes differentiation of endothe-

    lial cells by blocking Ang-1 activation of Tie2. In normal

    mice, retinal vascularization occurs postnatally in con-

    junction with regression of the hyaloid vessels encasing

    the lens. Ang-2-deficient mice do not develop retinal

    vascularization or regression of hyaloid vessels, thus sup-

    porting the role of Ang-2 as a destabilizing agent. Ang-2

    mRNA expression is increased in the inner nuclear layer

    and ganglion cell layer in the oxygen-induced mouse

    model of retinopathy, indicating that it may play a role in

    ROP.

    Tie1 signal transduction has been difficult to study

    because no ligand has been identified for this receptor. It

    is speculated that Tie1 may be involved in ligand-

    independent signaling or may form a heterodimer with

    Tie2 and modulate Tie2 signaling.

    AntiangiogenesisResearch in the early 1970s showed that formation of

    new blood vessels was important for tumor growth and

    progression. Since then, antiangiogenic therapy aimed at

    prevention of endothelial cell migration and induction of

    apoptosis has been tried as a treatment for cancer. Several

    early trials showed no benefit. However, an increased

    understanding of the angiogenic processes and factors

    involved has led to newer strategies that target either:

    1) proangiogenic factors, their receptors, or downstream

    signaling molecules; 2) increased expression of endoge-

    nous inhibitors or administration of exogenous inhibi-

    tors; or 3) the new blood vessels directly. One of the

    challenges is the designing of appropriate assays to mon-

    itor the effect of the antiangiogenic agents.

    Several inhibitors of tumor angiogenesis have been

    described (Table 2). Some are naturally occurring mole-

    cules, such as angiostatin, endostatin, platelet factor-4,

    thrombospondin-1, interferon-alpha, troponin I, and

    tissue inhibitors of metalloproteinases (TIMP).

    Antiangiogenic therapy has been extended to diseases

    causing intraocular neovascularization, particularly dia-betic retinopathy. Exogenous inhibitors have been tested

    most commonly in the mouse model of retinopathy.

    Studies in animals have shown that alpha v integrin

    antagonist peptides and antibodies produce a reduction

    in retinal neovascularization as well as inhibition of an-

    giogenesis in tumors and in arthritis. The humanized

    Table 2.Endogenous Inhibitors ofAngiogenesis

    StatinsAngiostatin (inhibits EC proliferation and migration

    by binding ATPase and annexin II on endothelialcells)

    Endostatin (inhibits EC proliferation by binding tointegrin)

    Platelet factor-4 (blocks binding of FGF to EC) Thrombospondin (modulates EC proliferation and

    migration) Interferon-alpha (downregulates action of VEGF) TIMP family (inhibition of metalloproteinases)

    EC endothelial cell, FGF fibroblast growth factor, VEGF vascular endothelial growth factor, TIMP tissue inhibitors ofmetalloproteinases

    ophthalmology angiogenesis

    e360 NeoReviews Vol.4 No.12 December 2003

  • 8/10/2019 Angiogenesis and Antiangiogenesis in the Neonate Relevance to Retinopathy of Prematurity

    6/8

    form of this antibody is being tested in clinical trials.

    Retinal neovascularization was inhibited in transgenic

    mice expressing a growth hormone antagonist gene, and

    the degree of inhibition was proportional to serum levels

    of growth hormone as well as insulinlike growth factor-1

    (IGF-1). This inhibition was decreased by administration

    of exogenous IGF-1, suggesting a potential therapy for

    intraocular neovascularization. Transgenic mice deficient

    for the endothelial nitric oxide synthase (eNOS) gene

    have decreased hyperoxia-induced attenuation of the

    retinal vasculature compared with normal controls,

    which suggests a possible therapeutic role for inhibitors

    of eNOS activity in the future. Angiotensin II also has

    been shown to play a possible role in induction of neo-

    vascularization in the oxygen-induced mouse model of

    retinopathy. Administration of perindopril, an

    angiotensin-converting enzyme inhibitor, to mice de-

    creased the number of endothelial cells in the retinas of

    treated versus untreated animals.

    Increased understanding of endothelial cell physiol-

    ogy and identification of inhibitors has led to an exten-

    sion of antiangiogenesis research from the bench to the

    bedside. This exciting, novel approach is being tested in

    several clinical trials for diabetic retinopathy that is in

    phase II /III. Some of the antiangiogenic strategies that

    are undergoing testing in clinical trials for ocular disease

    include VEGF-A inhibitors such as anti-VEGF aptamer,humanized anti-VEGF Fab monoclonal antibody (rhu-

    Fab), thalidomide, angiostatic steroids, protein kinase

    C-beta inhibitor (LY333531), growth hormone antago-

    nists such as long-acting somatostatin, and integrin an-

    tagonists. Treatment of ocular angiogenesis depends on

    adequate transfer of the drug to the back of the eye,

    where pathologic neovascularization typically occurs.

    This can be achieved by using eye drops or intravitreal

    injections. Gene transfer to the eye using a safe viral

    vector that constitutively expresses an antiangiogenic

    protein or by using a liposome expression vector is an-

    other promising technology that is being tested in clini-cal trials in patients who have myocardial ischemia or

    limb ischemia.

    Possible Role of Antiangiogenesis in ROPTo date, there are no trials of antiangiogenic agents in

    ROP, and several unique features of ROP must be ad-

    dressed before they can be undertaken. First, the blood

    vessels in ROP are immature, and unlike mature blood

    vessels, they are VEGF-dependent. Second, unlike in

    neovascularizing diseases affecting adults, a large part of

    the peripheral retina in infants is avascular. Therefore,

    antiangiogenic agents must be selective for abnormal

    vessels. Third, because neovascularization typically oc-

    curs at34 to36 weeks gestation, therapy can be targeted

    to a particular duration. With VEGF levels decreased in

    the first phase of ROP and increased in the second phase,

    two different strategies potentially can be used in this

    population. The first is administration of VEGF or a

    VEGF receptor agonist to rescue the vessels during the

    hyperoxic phase. The second is to antagonize selectively

    VEGF or downstream signaling molecules in abnormal

    vessels seen in the second phase of retinopathy. One of

    the concerns with the first approach is that VEGF also

    can stimulate abnormal vessel proliferation and increased

    permeability in ROP. Recent work by Shih and associates

    has shown that selective use of placental growth factor I

    (PGFI), a selective agonist for VEGF receptor-1, pro-

    tects the retinal vessels from hyperoxia-induced obliter-

    ation without any concomitant increase in retinal neovas-

    cularization during normal retinal vascular development.

    A similar vasoprotective effect of PGFI was shown in

    oxygen-damaged retinal vessels. The exact mechanism

    underlying this selective effect on pathologic angiogen-

    esis while sparing normal blood vessel growth is un-

    known. The signal transduction pathway resulting in

    vasoprotection by VEGF or PGFI is yet to be identified.

    Phosphorylation of the survival kinase Akt1 and inhibi-

    tion of apoptosis along with increased expression of

    antiapoptotic genes has been shown to be important.

    Use of Supplemental Oxygen for Preventionof Threshold ROPPhelps and colleagues employed the kitten model of

    oxygen-induced retinopathy to show that use of supple-

    mental oxygen (28%) in animals recovering from expo-

    sure to 80% oxygen decreased the degree of neovascular-

    ization compared with animals recovering in normoxia.

    This effect possibly is mediated by a decrease in VEGF

    levels. The Supplemental Therapeutic Oxygen for Pre-

    threshold ROP (STOP-ROP) was a randomized trial

    conducted to test this hypothesis. Infants who had pre-threshold ROP and were unable to maintain an oxygen

    saturation of 94% in room air were randomized to either

    the control (O2saturations of 88% to 94%) or the treat-

    ment group (O2saturations of 96% to 99%). No statisti-

    cally significant difference was noted in the rate of con-

    version to threshold ROP between the two groups,

    indicating the lack of an adverse effect of oxygen in active

    prethreshold ROP. However, infants in the supplemen-

    tal group had a much higher incidence of adverse pulmo-

    nary events, such as pneumonia and exacerbation of

    pulmonary disease. Although the current data do not

    support the use of supplemental oxygen for prethreshold

    ophthalmology angiogenesis

    NeoReviews Vol.4 No.12 December 2003 e361

  • 8/10/2019 Angiogenesis and Antiangiogenesis in the Neonate Relevance to Retinopathy of Prematurity

    7/8

    ROP, it may have a role in selected cases of ROP. A

    subgroup analysis suggested a benefit among infants who

    did not have plus disease. In addition, infants who were

    excluded from the study by virtue of maintaining O2saturations less than 94% in room air (ie, those who had

    less severe pulmonary disease) had a much lower inci-

    dence of progression to threshold ROP than those en-

    rolled in the STOP-ROP study. Further studies are re-

    quired to define more clearly the effect of oxygen on

    retinal neovascularization in preterm infants.

    ConclusionSeveral exciting new developments have expanded un-

    derstanding of the mechanisms underlying angiogenesis.Continued identification and characterization of molec-

    ular factors that regulate blood vessel growth hold con-

    siderable promise for the future of antiangiogenesis as a

    therapeutic approach for ROP.

    Suggested ReadingAshton N, Ward B, Serpell G. Effect of oxygen on developing

    retinal vessels with particular reference to the problem of retro-lentalfibroplasia.Br J Ophthalmol. 1954;38:397332

    Carmeliet P, Ferreira V, Breier G, et al. Abnormal blood vessel

    development and lethality in embryos lacking a single VEGF

    allele.Nature. 1996;380:435 439Chan-Ling T, Gock B, Stone J. The effect of oxygen on vasoforma-

    tive cell division. Evidence that physiological hypoxia is thestimulus for normal retinal vasculogenesis. Invest OphthalmolVis Sci. 1995;36:12011214

    Ferrara N, Alitalo K. Clinical applications of angiogenic growthfactors and their inhibitors.Nat Med. 1999;5:13591364

    Isner JM, Asahara T. Angiogenesis and vasculogenesis as therapeu-tic strategies for postnatal neovascularization. J Clin Invest.1999;103:12311236

    Kerbel R, Folkman J. Clinical translation of angiogenesis inhibitors.Nat Rev Cancer.2002;2:727739

    Patz A. The role of oxygen in retrolental fibroplasia. Pediatrics.1957;19:504524

    Shih SC, Ju M, Liu N, Smith LE. Selectivestimulation of VEGFR-1prevents oxygen-induced retinal vascular degeneration in reti-nopathy of prematurity.J Clin Invest. 2003;112:5057

    Shweiki D, Itin A, Soffer D, Keshet E. Vascular endothelial growthfactor induced by hypoxia may mediate hypoxia-initiated angio-genesis.Nature. 1992;359:843 845

    Smith LE, Kopchick JJ, Chen W, et al. Essential role of growthhormone in ischemia-induced retinal neovascularization. Sci-ence. 1997;276:17061709

    Supplemental Therapeutic Oxygen for Prethreshold Retinopathy ofPrematurity (STOP-ROP), a randomized, controlled trial. I:primary outcomes.Pediatrics.2000;105:295310

    Witmer AN, Vrensen GF, Van Noorden CJ, Schlingemann RO.Vascular endothelial growth factors and angiogenesis in eyedisease.Prog Retin Eye Res. 2003;22:129

    Yancopoulos GD, Davis S, Gale NW, Rudge JS, Wiegand SJ,

    Holash J. Vascular-specific growth factors and blood vessel

    formation.Nature. 2000;407:242248

    Supplemental ReadingAiello LP, Avery RL, Arrigg PG, et al. Vascular endothelial growth

    factor in ocularfluid of patients with diabetic retinopathy and

    other retinal disorders. N Engl J Med. 1994;331:14801487

    Aiello LP, Pierce EA, Foley ED, et al. Suppression of retinal

    neovascularization in vivo by inhibition of vascular endothelial

    growth factor (VEGF) using soluble VEGF-receptor chimeric

    proteins.Proc Natl Acad Sci U S A. 1995;92:1045710461

    Ashton N, Gaymore C, Pedler C. Studies in developing retinal

    vessels. V. Mechanism of vaso-obliteration. Br J Ophthalmol.

    1957;41:449 460Beck L,DAmore PA Jr. Vascular development: cellular and molec-

    ular regulation.FASEB J. 1997;11:365373

    Brooks SE, Gu X, Samuel S, et al. Reduced severity of oxygen-

    induced retinopathy in eNOS-deficient mice. Invest Ophthalmol

    Vis Sci. 2001;42:222228

    Dollery CT, Bulpitt CJ, Kohner EM. Oxygen supply to the retina

    from the retinal and choroidal circulations at normal and in-

    creased arterial oxygen tensions. Invest Ophthalmol. 1969;8:

    588594

    Eliceiri BP, Cheresh DA. The role of alpha v integrins during

    angiogenesis: insights into potential mechanisms of action and

    clinical development.J Clin Invest. 1999;103:12271230

    Ferrara N. Vascular endothelial growth factor: molecular and bio-

    logical aspects.Curr Top Microbiol Immunol. 1999;237:130

    Forsythe JA, Jiang J, Iyer NV, et al. Activation of vascular endothe-

    lial growth factor gene transcription by hypoxia-inducible factor

    1.Mol Cell Biol. 1996;16:4604 4613

    Keck PJ, Hauser SD, Krivi G, et al. Vascular permeability factor, an

    endothelial cell mitogen related to PDGF. Science. 1989;246:

    1309 1312

    Lindahl P, Johansson BR, Leveen P, Betsholtz C. Pericyte loss and

    microaneurysm formation in PDGF-B-deficient mice. Science.

    1997;277:242245

    Miller JW, Adamis AP, Shima DT, et al. Vascular endothelial

    growth factor/vascular permeability factor is temporally and

    spatially correlated with ocular angiogenesis in a primate model.

    Am J Pathol. 1994;145:574584

    Phelps DL, Rosenbaum AL. Effects of marginal hypoxemia on

    recovery from oxygen-induced retinopathy in the kitten model.Pediatrics. 1984;73:1 6

    Robinson GS, Pierce EA, Rook SL, Foley E, Webb R, Smith LE.

    Oligodeoxynucleotides inhibit retinal neovascularization in a

    murine model of proliferative retinopathy.Proc Natl Acad Sci

    U S A. 1996;93:4851 4856

    Sato TN, Tozawa Y, Deutsch U, et al. Distinct roles of the receptor

    tyrosine kinases TIE-1 and TIE-2 in blood vessel formation.

    Nature. 1995;376:7074

    Shalaby F, Rossant J, Yamaguchi TP, et al. Failure of blood-island

    formation and vasculogenesis in Flk-1-deficient mice.Nature.

    1995;376:62 66

    Smith LE, Wesolowski E, McLellan A, et al. Oxygen-induced

    retinopathy in the mouse. Invest Ophthalmol Vis Sci.1994;35:

    101111

    ophthalmology angiogenesis

    e362 NeoReviews Vol.4 No.12 December 2003

  • 8/10/2019 Angiogenesis and Antiangiogenesis in the Neonate Relevance to Retinopathy of Prematurity

    8/8

    Stetler-Stevenson WG. Matrixmetalloproteinases in angiogenesis: a

    moving target for therapeutic intervention.J Clin Invest. 1999;

    103:12371241Stone J, Itin A,AlonT, Peer J, Gnessin H, Chan-Ling T, Keshet A.

    Development of retinal vasculature is mediated by hypoxia-

    induced vascular endothelial growth factor (VEGF) expression

    by neuroglia.J Neurosci. 1995;15:4738 4747

    Yoshida A, Yoshida S, Khalil AK, Ishibashi T, Inomata H. Role ofNF-kappaB-mediated interleukin-8 expression in intraocular neo-

    vascularization.Invest Ophthalmol Vis Sci. 1998;39:10971106

    NeoReviews Quiz

    8. Vascularization of the retina during fetal development includes vasculogenesis (the de novo formation ofcapillaries from endothelial cells) followed by angiogenesis (the formation of blood vessels from existingblood vessels). Vascular endothelial growth factor (VEGF) plays a key role in retinal vascularization, bothduring normal development and in the pathogenesis of retinopathy of prematurity. Of the following, themost accurate statement regarding retinal vascularization is that:

    A. Angiogenesis begins around 16 weeks of gestational age.B. Consumption of oxygen is much higher in the retina in the dark than in the light.C. Vascularization of the retina is complete by 36 weeks of gestational age.D. Vasculogenesis is dependent on hypoxia-induced VEGF.E. VEGF is produced by endothelial cells in the retina.

    9. Angiogenesis is a complex process that involves several angiogenic factors that have specific putativeroles. Of the following, the angiogenic factor mostinvolved in the recruitment of pericytes in remodelingand stabilization of blood vessels is:

    A. Fibroblast growth factor.B. Interleukin-8.C. Platelet-derived growth factor.

    D. Tumor necrosis factor-alpha.E. Vascular endothelial growth factor.

    10. Retinopathy of prematurity is characterized by two phases: an initial phase of vasoconstriction and vaso-obliteration of developing retinal blood vessels and a subsequent phase of hypoxia-induced abnormalneovascularization. An angiogenic factor that protects against both vaso-obliteration andneovascularization would be useful in the treatment of retinopathy of prematurity. Of the following, theangiogenic factor that has the mostpromise as a potential treatment of retinopathy of prematurity is:

    A. Fibroblast growth factor.B. Placental growth factor.C. Platelet-derived growth factor.D. Transforming growth factor.E. Vascular endothelial growth factor.

    ophthalmology angiogenesis

    NeoReviews Vol.4 No.12 December 2003 e363