throbber
Downloaded from
`
`symposium.cshlp.org
`
`
`
`Cold Spring Harbor Laboratory Press on November 9, 2017 - Published by
`
`VEGF Trap as a Novel Antiangiogenic Treatment Currently in
`Clinical Trials for Cancer and Eye Diseases, and VelociGene®-
`based Discovery of the Next Generation of Angiogenesis Targets
`
`J.S. RUDGE, G. THURSTON, S. DAVIS, N. PAPADOPOULOS, N. GALE,
`S.J. WIEGAND, AND G.D. YANCOPOULOS
`Regneron Pharmaceuticals, Inc., Tarrytown, New York 10591
`
`The concept that tumors can be controlled by directly targeting their vascular supply has finally come of age, because clini-
`cal trials using a humanized monoclonal antibody that blocks VEGF have demonstrated exciting efficacy in cancer patients,
`as well as in vascular eye diseases that can lead to blindness. However, data suggest that these current regimens may not pro-
`vide complete VEGF inhibition and, thus, that the maximum therapeutic potential of VEGF blockade has not yet been
`achieved. We describe the status of a very potent and high-affinity VEGF blocker, termed the VEGF Trap, that may provide
`the opportunity to maximize the potential of VEGF blockade in cancer as well as in vascular eye diseases. We also describe
`use of the VEGF Trap as a research tool, when coupled to high-throughput mouse genetics approaches such as VelociGene®
`that can be exploited in strategies to discover and validate the next generation of angiogenesis targets.
`
`The concept that tumors can be controlled by directly
`targeting their vascular supply has finally come of age.
`The first antiangiogenesis approach to be validated in
`human cancer patients involves blocking vascular en-
`dothelial growth factor (VEGF-A). In this regard, the
`most advanced clinical data have been generated with a
`humanized monoclonal antibody termed bevacizumab
`(Avastin) that directly binds and blocks all isoforms of
`VEGF-A (Ferrara et al. 2004). Despite the promising
`data achieved to date, dose-response studies suggest that
`higher doses of bevacizumab may provide even greater
`benefit (Yang et al. 2003; Yang 2004), implying that cur-
`rent bevacizumab regimens may not provide optimal
`VEGF inhibition and thus may not have yet demon-
`strated the maximum potential of VEGF blockade in
`cancer. In addition to the promise of anti-VEGF ap-
`proaches in cancer, blocking VEGF-A has also been im-
`pressive in maintaining and improving vision in wet age-
`related macular degeneration (AMD), a disease marked
`by leaky and proliferating vessels which distort the
`retina, and these data suggest that VEGF blockade may
`provide benefit in other eye diseases involving vascular
`leak and proliferation (Bergsland 2004). Efficacy in wet
`AMD has most notably been achieved using a modified
`fragment of
`the bevacizumab antibody,
`termed
`ranibizumab (Lucentis), delivered via monthly intraocu-
`lar injections (Brown et al. 2006; Heier et al. 2006).
`In this paper, we focus on the development and status
`of a novel VEGF-blocking agent, termed the VEGF Trap,
`that retains many of the advantages of a blocking anti-
`body but may offer further potential (Holash et al. 2002).
`The VEGF Trap consists of portions of VEGF receptors
`that have been fused to the constant region of an antibody,
`resulting in a fully human biologic with exceedingly high
`affinity that blocks not only all isoforms of VEGF-A, but
`also related VEGF family members such as placental
`
`growth factor (PlGF). The VEGF Trap also displays ex-
`tended pharmacological half-life, allowing long-term as
`well as very high affinity blockade. The VEGF Trap has
`performed impressively in extensive animal studies of
`cancer and eye diseases, and initial clinical trials appear
`promising. The VEGF Trap may provide the opportunity
`to explore the potential of more complete VEGF block-
`ade in cancer, as well as the opportunity for more com-
`plete blockade and even longer-interval dosing regimens
`in eye diseases. To conclude this paper, we describe how
`the VEGF Trap can be used as a research tool in efforts to
`discover and validate the next generation of targets in the
`field of angiogenesis.
`
`DISCOVERY OF VEGF AND ITS REQUISITE
`ROLES DURING NORMAL DEVELOPMENT
`AND IN DISEASE SETTINGS
`
`Initial studies by Dvorak and his colleagues (Senger et
`al. 1986; Dvorak et al. 1999) identified a protein in tumor
`ascites fluid that was capable of inducing vascular leak and
`permeability, which they termed vascular permeability fac-
`tor (VPF). Independent efforts by Ferrara and his col-
`leagues to identify secreted factors that could promote
`tumor angiogenesis led to the discovery of a protein in
`bovine pituitary follicular cell conditioned medium with
`mitogenic properties for endothelial cells which they
`termed vascular endothelial cell growth factor (VEGF)
`(Ferrara and Henzel 1989; Leung et al. 1989). Upon se-
`quencing and further studies, this VEGF protein was unex-
`pectedly found to correspond to the VPF previously iden-
`tified by the Dvorak lab. These findings set the stage for a
`concerted effort to define the role of VEGF/VPF (hereon
`VEGF) in cancer angiogenesis as well as other settings of
`vascular disease, which have led to the realization that both
`of its initially realized actions—i.e., promoting vascular
`
`Cold Spring Harbor Symposia on Quantitative Biology, Volume LXX. © 2005 Cold Spring Harbor Laboratory Press 0-87969-773-3.
`
`411
`
`0001
`
`CELLTRION - EXHIBIT 1052
`
`

`

`Downloaded from
`
`
`
`symposium.cshlp.org Cold Spring Harbor Laboratory Press on November 9, 2017 - Published by
`
`
`
`412
`
`RUDGE ET AL.
`
`permeability and vascular growth—appear critical to un-
`derstanding its roles during normal biology and in disease.
`Approximately two decades of intensive investigation
`by numerous laboratories has revealed a great deal about
`VEGF and its actions. It is now clear that VEGF is per-
`haps the most critical vascular regulator during normal
`development as well as in many disease states, and more-
`over, that its dosage must be exquisitely regulated in a
`spatial and temporal manner to avoid vascular disaster.
`Disruption of both VEGF alleles in developing mice,
`which ablates all VEGF production, results in complete
`failure to develop even a primordial vasculature, demon-
`strating that VEGF is absolutely essential for the earliest
`stages of blood vessel development. Still more remark-
`ably, disruption of even a single VEGF allele in develop-
`ing mice, which decreases VEGF levels by half, also re-
`sults in embryonic lethality due to severe vascular
`abnormalities (Carmeliet et al. 1996; Ferrara et al. 1996),
`demonstrating the need for exquisite regulation of VEGF
`levels to form normal vessels. Reciprocally, modest in-
`creases in VEGF levels during development also lead to
`vascular disaster and lethality (Miquerol et al. 2000).
`VEGF continues to be critical during early postnatal
`growth and development, as evidenced by the lethality
`and major growth disturbances caused by conditional dis-
`ruption of the VEGF gene or by administration of VEGF
`blockers (Ravindranath et al. 1992; Carmeliet et al. 1996;
`Ferrara et al. 1996, 1998; Gerber et al. 1999a; Ryan et al.
`1999; Fraser et al. 2000; Zimmermann et al. 2001; Haz-
`zard et al. 2002; Eremina et al. 2003). However, VEGF
`blockade in older animals is much less traumatic, affect-
`ing only those structures that continue to depend on on-
`going vascular remodeling, such as occurs in bone
`growth plates or during remodeling of the female repro-
`ductive organs (Ferrara et al. 1998; Gerber et al. 1999a,b).
`As discussed in greater detail below, vascular remodeling
`is absolutely required in a variety of pathological settings,
`such as during tumor growth, providing major therapeu-
`tic opportunities for VEGF blockade in the adult setting
`in which such blockade can be tolerated.
`
`VEGF ISOFORMS, VEGF FAMILY MEMBERS,
`AND VEGF RECEPTORS
`
`Further study of the gene encoding human VEGF re-
`vealed eight exons separated by seven introns, which re-
`sults in the generation of four isoforms of increasing
`size—VEGF121, VEGF165, VEGF189, and VEGF206 (sub-
`scripts refer to number of amino acids comprising the iso-
`form, with the VEGF isoforms varying in length at their
`carboxyl termini). The main purpose of these isoforms
`appears to relate to their bioavailability such that the 121
`isoform is diffusible, whereas the higher-molecular-
`weight isoforms remain bound to the extracellular matrix,
`requiring cleavage to be released (Houck et al. 1992; Park
`et al. 1993; Keyt et al. 1996).
`Because of the discovery of additional members of
`the VEGF family, VEGF is now often referred to as
`VEGF-A. Other members of the VEGF family were
`
`identified based on their homology with VEGF, as well
`as their ability to interact with a related set of cell-sur-
`face receptors (Eriksson and Alitalo 1999; see below).
`The first VEGF relative to be identified is PlGF, and un-
`til recently, little was known about its normal function
`(Maglione et al. 1991). Whereas mice lacking PlGF ap-
`pear to undergo normal vascular development, recent
`findings indicate that adult mice lacking PlGF exhibit
`deficiencies in certain models of adult vascular remod-
`eling, including in tumors and eye disease models, rais-
`ing the interesting possibility that the activity of PlGF
`may be limited to these settings and that blockade of
`PlGF may provide enhanced efficacy when combined
`with VEGF blockade (Persico et al. 1999; Carmeliet
`2000). Little is known about VEGF-B, and mice lacking
`VEGF-B are overtly healthy and fertile. VEGF-C and D
`seem to play more critical roles in the lymphatic vascu-
`lature than in the blood vasculature, showing specificity
`for a VEGF receptor (see below) expressed on this vas-
`culature; administration of both of these factors leads to
`lymphatic vessel hyperplasia (Joukov et al. 1996; Orlan-
`dini et al. 1996; Olofsson et al. 1999).
`Following rapidly on the heels of the discovery of
`VEGF came the identification of two closely related
`high-affinity receptors for VEGF—FLT1 (FMS-like ty-
`rosine kinase) now termed VEGFR1 (de Vries et al.
`1992), and KDR or Flk1, now termed VEGFR2 (Shibuya
`et al. 1990; Terman et al. 1992; Millauer et al. 1993).
`These high-affinity receptors share features of many
`other growth factor receptors, in that they contain an ex-
`tracellular domain which binds and is dimerized by lig-
`and, and a cytoplasmic tyrosine kinase domain that can be
`regulated upon binding of ligand to the extracellular do-
`main. VEGFR2 seems to be the receptor which mediates
`the major growth and permeability actions of VEGF,
`whereas VEGFR1 may have a negative role, either by
`acting as a decoy receptor or by suppressing signaling
`through VEGFR2. Thus, mice engineered to lack
`VEGFR2 fail to develop a vasculature and have very few
`endothelial cells (Shalaby et al. 1995), phenocopying
`mice lacking VEGF, whereas mice lacking VEGFR1
`seem to have excess formation of endothelial cells that
`abnormally coalesce into disorganized tubules (Fong et
`al. 1995). Mice engineered to express only a truncated
`form of VEGFR1, lacking its kinase domain, appear
`rather normal, consistent with the notion that the primary
`role of VEGFR1 may be that of a decoy receptor (Hirat-
`suka et al. 1998), and supporting only a minor role for the
`cytoplasmic kinase domain. The third member of this re-
`ceptor family, initially called Flt-4 and now termed
`VEGFR3, does not bind to VEGF-A nor PlGF, and in-
`stead binds to VEGF-C and VEGF-D and seems to medi-
`ate the actions of these latter two factors on the lymphatic
`vasculature (Taipale et al. 1999).
`In addition to these primary receptors, a number of po-
`tential accessory receptors for the VEGFs have been
`identified, although the requisite roles of these receptors
`in mediating VEGF responses have not been clearly elu-
`cidated. These potential accessory receptors include the
`neuropilins (Soker et al. 1998).
`
`0002
`
`

`

`Downloaded from
`
`symposium.cshlp.org
`
`
`
`Cold Spring Harbor Laboratory Press on November 9, 2017 - Published by
`
`VEGF TRAP AND THE NEXT ANGIOGENESIS TARGETS
`
`413
`
`ROLE OF VEGF IN TUMOR ANGIOGENESIS
`SUPPORTS CONCEPT OF BLOCKING VEGF AS
`AN ANTITUMOR STRATEGY
`
`One area of intense study after the discovery of VEGF
`was the analysis of VEGF expression levels in different
`tumor types using in situ hybridization. VEGF was found
`to be expressed in a number of different tumors such as
`renal, gastrointestinal, breast, ovarian, pancreatic, and
`lung, but the variability in expression across the tumor
`and between different tumor types made the simple cor-
`relation between VEGF and severity of the cancer impos-
`sible (Yoshiji et al. 1996; Sowter et al. 1997; Volm et al.
`1997; Ellis et al. 1998; Tomisawa et al. 1999). However,
`out of these studies came the interesting finding that one
`tumor type, renal cell carcinoma, had particularly high
`VEGF expression which correlated with inactivation of
`the von Hippel Lindau locus, resulting in loss of control
`of the tumor’s oxygen sensor, hypoxia-inducible factor
`(HIF) (Iliopoulos et al. 1996; Lonser et al. 2003). The up-
`regulation of VEGF in an attempt to reoxygenate the tu-
`mor through revascularization led to the belief that this
`tumor may either be highly sensitive to anti-VEGF ther-
`apy or highly refractory. Fortunately, the former seems to
`be the case (Yang et al. 2003).
`Concomitant with the analysis of human tumors for
`VEGF expression came the development of animal mod-
`els of cancer where the hypothesis that VEGF was re-
`quired for tumor vasculature, and thus tumor growth,
`could be tested. In 1993, 4 years after their discovery of
`VEGF, Ferrara and colleagues demonstrated that a mouse
`monoclonal antibody to human VEGF (A.4.6.1) could in-
`hibit the growth of several human tumor types in nude
`mice with inhibition ranging from 70% to more than 90%
`(Kim et al. 1993). Subsequent to this observation, a num-
`ber of laboratories using different strategies to inhibit
`VEGF signaling have shown to a greater or lesser extent
`that inhibition of VEGF can have a major impact on tumor
`growth in mice. In addition to numerous studies using the
`VEGF-blocking antibody, other strategies to block VEGF
`in tumor models included blocking antibodies targeting
`VEGFR2 (Prewett et al. 1999), soluble VEGF receptors
`acting as circulating decoys to capture VEGF and prevent-
`ing it from binding cell-surface receptors (Ferrara et al.
`1998; Gerber et al. 1999a,b; Liang et al. 2006), dominant-
`negative VEGF receptors expressed at high levels on tu-
`mor surfaces, small-molecule inhibitors of VEGF receptor
`kinases and other kinases (Smith et al. 2004), antisense
`oligonucleotides targeting VEGF, and VEGF siRNA
`(Grunweller and Hartmann 2005; Lu et al. 2005).
`As the number of studies increased comparing the dif-
`ferent modes of inhibiting VEGF, it became apparent that
`blocking tumor-derived VEGF without blocking stromal
`VEGF was not as efficacious, implicating stromal VEGF
`as a crucial player in tumor growth and angiogenesis.
`Thus, antibodies such as A.4.6.1 which only block human
`VEGF did not fare as well in blocking human tumor
`growth in immunocompromised mice as reagents block-
`ing both tumor and host stroma-derived VEGF (Gerber et
`al. 2000; Liang et al. 2006).
`
`DEVELOPMENT OF VEGF TRAP
`
`The clinical promise of initial anti-VEGF approaches
`highlighted the need to optimize blockade of this path-
`way. Early studies indicated that one of the most effec-
`tive ways to block the VEGF signaling pathway is to
`prevent VEGF from binding to its endogenous receptors
`by administering soluble decoy receptors (Ferrara et al.
`1998; Gerber et al. 1999b). In particular, a soluble decoy
`receptor created by fusing the first three immunoglobu-
`lin-like (Ig) domains of VEGFR1 to the constant region
`(Fc portion) of human IgG1 resulted in a forced homo-
`dimer that acted as a very high affinity blocking reagent
`with 5–20 picomolar binding affinity for VEGF, and in
`tumor experiments this VEGFR1-Fc reagent was effica-
`cious at approximately 500-fold lower concentration
`than a similar VEGFR2-Fc construct (Kuo et al. 2001).
`Despite its high affinity, the VEGFR1-Fc was not a fea-
`sible clinical candidate because of its poor pharmaco-
`kinetic profile; in rodent studies, this protein had to be
`administered frequently and at very high doses to
`achieve efficacious levels. In addition, this agent ap-
`peared to have nonspecific toxicity effects that did not
`seem to be accounted for by its blocking of VEGF (Kuo
`et al. 2001). We decided to exploit our Trap technology
`platform (Economides et al. 2003), which involves
`defining and fusing minimal binding units from differ-
`ent receptor components to generate chimeric fusion
`proteins that act as high-affinity soluble blockers, in an
`attempt to create a potent and well-behaved Trap for
`VEGF. The result was a chimeric fusion protein con-
`taining a modified domain 2 of VEGFR1 and the third
`Ig domain of VEGFR2 fused to the Fc region of human
`IgG1, resulting in a fully human protein that we term
`VEGF Trap (Holash et al. 2002). This reagent has the
`advantage of being fully human and thus potentially
`non-immunogenic, as well as being substantially
`smaller than previous fusion proteins and antibodies,
`raising the possibility that it might allow improved tis-
`sue and tumor penetration. In addition, this VEGF Trap
`had greatly improved pharmacological bioavailability
`as compared to the initial VEGFR1-Fc reagent, exhibit-
`ing about a 300-fold increase in the maximum concen-
`tration achieved in the circulation (i.e., Cmax), as well as
`about a 1000-fold increase in total circulation exposure
`(i.e., AUC) (Holash et al. 2002). Importantly, the affin-
`ity of VEGF Trap binding to both mouse and human
`VEGF isoforms (0.58 pM, 0.46 pM) was superior to that
`of the parental VEGFR1-Fc (~ 5–20 pM) (Holash et al.
`2002). In addition, the VEGF Trap also bound PlGF
`with high affinity (1.8 pM).
`To determine whether the improved pharmacological
`bioavailabity and high-affinity binding of VEGF Trap
`translated into superior performance in vivo, we first
`used a short-term and quantitative in vivo model of
`VEGF activity in which a single dose of VEGF induces
`a stereotypic reduction in blood pressure. In this acute
`assay model, we found that equivalent doses of VEGF
`Trap were indeed far superior to that of the parental
`VEGFR1-Fc (Holash et al. 2002).
`
`0003
`
`

`

`Downloaded from
`
`symposium.cshlp.org
`
`
`
`Cold Spring Harbor Laboratory Press on November 9, 2017 - Published by
`
`414
`
`RUDGE ET AL.
`
`VEGF TRAP EXHIBITS IMPRESSIVE
`EFFICACY IN PRECLINICAL TUMOR MODELS
`
`On the basis of the above evidence suggesting that the
`VEGF Trap was a potent VEGF blocker that was effica-
`cious in vivo, we moved to evaluate the VEGF Trap in tu-
`mor models. Initial studies confirmed the remarkable ef-
`ficacy of the VEGF Trap. Not only did the VEGF Trap
`blunt tumor growth in the early models, but it could com-
`pletely block tumor angiogenesis in many cases, resulting
`in completely avascular tumors (Holash et al. 2002).
`These initial studies inspired exploration of the VEGF
`Trap in multiple tumor models, in the laboratories of
`many different investigators, with impressive results in
`almost every case. In addition to its activity in multiple
`subcutaneous models of melanoma, glioma, and rhab-
`domyosarcoma tumors (Holash et al. 2002), the VEGF
`Trap has been shown to work in multiple pancreatic can-
`cer models (Fukasawa and Korc 2004), Wilms’ tumor
`(Huang et al. 2003), Ewing’s sarcoma (Dalal et al. 2005),
`glioblastoma (Wachsberger et al. 2005), and models of
`ovarian cancer as well as associated malignant ascites
`(Hu et al. 2005).
`In addition to the above published studies, recent un-
`published temporal studies indicate that vascular regres-
`sion can be seen in most tumors within hours of VEGF
`Trap treatment, resulting in marked and widespread hy-
`poxia within the tumors. In addition, transcription profil-
`ing studies during these temporal studies have revealed a
`set of endothelial-specific genes that are rapidly and pro-
`foundly regulated in response to VEGF Trap treatment.
`Further studies on some of these genes have led to their
`identification as potential targets for new antiangiogene-
`sis therapies (see below).
`In summary, animal tumor studies have indicated that
`treatment with VEGF Trap effectively inhibited tumor
`growth of a wide variety of murine, rat, and human tumor
`cell lines implanted either subcutaneously or orthotopi-
`cally in mice. VEGF Trap treatment inhibited the growth
`of tumors representing a variety of tumor types, including
`melanoma, glioma, rhabdomyosarcoma, ovarian, pancre-
`atic, renal, and mammary tumor tissue, with a broad ther-
`apeutic index. Growth of small established tumors was
`also inhibited. Histological analysis indicated that treat-
`ment with VEGF Trap resulted in the formation of largely
`avascular and necrotic tumors, demonstrating that tumor-
`induced angiogenesis was blocked. VEGF Trap was also
`active in blocking tumor growth in similar animal tumor
`models in combination with paclitaxel, docetaxel, or ra-
`diation, and was synergistic with 5-fluorouracil. VEGF
`Trap as a single agent and in combination with paclitaxel
`also prevented the formation of ascites in mouse tumor
`models (Byrne et al. 2003; Hu et al. 2005).
`
`VEGF TRAP IN CLINICAL
`TRIALS FOR CANCER
`
`The above results in animal tumor models supported
`the exploration of the VEGF Trap in human studies. Ini-
`tial clinical studies are promising (Dupont et al. 2005;
`
`Mulay et al. 2006; Rixe et al. 2006) in that the VEGF
`Trap as a single agent has resulted in objective radio-
`graphic responses in several advanced cancer patients
`suffering from multiply treated chemotherapy-refractory
`disease, as well as long-term stable disease in patients.
`Similar data are being generated in patients treated with
`VEGF Trap in combination with various chemotherapeu-
`tic agents. The VEGF Trap is now entering an assortment
`of additional exploratory as well as potentially pivotal ef-
`ficacy studies, both as a single agent and in combination
`with chemotherapy.
`
`VEGF TRAP EXHIBITS IMPRESSIVE
`EFFICACY IN PRECLINICAL MODELS OF
`VASCULAR EYE DISEASES
`
`In addition to the role for VEGF in tumor angiogenesis,
`a variety of studies have indicated that VEGF may play a
`key pathological role in vascular eye diseases, in particu-
`lar in diabetic edema and retinopathy settings, and in age-
`related macular degeneration (AMD), which are leading
`causes of vision loss and blindness. In these diseases, ex-
`cess VEGF is thought to result in vascular leak that con-
`tributes to abnormal swelling of the retina and resulting
`vision impairment, as well as in the abnormal growth of
`choroidal and retinal vessels that can destroy normal reti-
`nal architecture. Consistent with these possibilities, the
`VEGF Trap has demonstrated impressive efficacy in an
`assortment of animal models of these eye diseases.
`Preclinical studies in rodents have shown that VEGF
`Trap can inhibit choroidal (Saishin et al. 2003) and
`corneal (Wiegand et al. 2003) neovascularization, as well
`as suppress vascular leak into the retina (Qaum et al.
`2001), and that the VEGF Trap can also promote the sur-
`vival of corneal transplants by inhibiting associated neo-
`vascularization (Cursiefen et al. 2004). In addition, in a
`primate model of AMD, in which choroidal neovascular
`lesions and vascular leak are induced by using a laser to
`create small lesions in the retinas of adult cynomolgus
`macaques, both systemically and intravitreally delivered
`VEGF Trap not only prevented development of vascular
`leak and neovascular membranes when administered
`prior to laser lesion, but also induced regression when ad-
`ministered after lesions had developed (Wiegand et al.
`2005). These preclinical results support a role for VEGF
`blockade, and in particular for local delivery of the VEGF
`Trap, in multiple vascular eye diseases ranging from
`AMD and diabetic eye diseases to corneal injury and
`transplantation.
`
`VEGF TRAP IN CLINICAL TRIALS FOR
`VASCULAR EYE DISEASES
`
`The above results in animal models have supported the
`exploration of the VEGF Trap in human studies of vascu-
`lar eye diseases. Initial clinical studies in human patients
`suffering from both AMD and diabetic edema and
`retinopathy appear quite promising, with evidence in
`early trials that the VEGF Trap can rapidly and impres-
`sively decrease retinal swelling, and that these changes
`
`0004
`
`

`

`Downloaded from
`
`
`
`symposium.cshlp.org Cold Spring Harbor Laboratory Press on November 9, 2017 - Published by
`
`
`
`VEGF TRAP AND THE NEXT ANGIOGENESIS TARGETS
`
`415
`
`can be associated with improvement in visual acuity
`(Nguygen et al. 2006; Shah et al. 2006). The VEGF Trap
`is now entering more advanced clinical trials in vascular
`eye diseases.
`
`In addition, the success with this screen in terms of yield-
`ing Dll4 as an exciting new antiangiogenesis target has led
`us to rigorously pursue several additional similarly identi-
`fied targets.
`
`THE NEXT GENERATION OF ANGIOGENESIS
`TARGETS: ANGIOPOIETINS AND Dll4
`
`Despite the promise of anti-VEGF approaches in gen-
`eral, and that of the VEGF Trap in particular, it is clear
`both from animal studies and from the emerging human
`trials that tumors display varying degrees of responsivity
`to VEGF blockade. Whereas some tumors might show
`marked regression and/or very long term stabilization,
`other tumors can continue to grow even in the face of anti-
`VEGF treatments. The realization that some tumors can
`be relatively resistant to anti-VEGF approaches raises the
`need for additional antiangiogenesis approaches that
`might be useful in such settings. Toward this end, as noted
`above, we performed transcriptional profiling screens to
`identify endothelial-specific targets that are markedly reg-
`ulated either by VEGF blockade or by excess VEGF activ-
`ity, reasoning that such targets might prove interesting as
`new antiangiogenesis targets. Confirming the potential of
`such a screen, one target that was “rediscovered” via such
`screens was Angiopoietin-2. We had previously indepen-
`dently identified the Angiopoietins as key new angiogenic
`regulators that seemed to work in tandem with the VEGFs
`(Davis et al. 1996; Suri et al. 1996; Maisonpierre et al.
`1997; Valenzuela et al. 1999; Yancopoulos et al. 2000;
`Gale et al. 2002), and moreover, obtained substantial data
`that Angiopoietin-2 in particular was specifically induced
`in tumor vasculature and that it was important for tumor
`angiogenesis (Holash et al. 1999); a recent study employ-
`ing Angiopoietin-2-blocking antibodies confirmed no-
`table antitumor effects (Oliner et al. 2004). On the basis of
`the confidence in these transcriptional profiling screens
`engendered by the reidentification of Angiopoietin-2, we
`explored additional potential targets identified by the
`screens. Among these targets we have reported the identi-
`fication of Delta-like ligand 4 (Dll4) (a ligand for the
`Notch family of receptors) as a gene that is markedly and
`specifically induced in tumor vasculature (Gale et al.
`2004). Moreover, Dll4 is strikingly up-regulated in
`VEGF-overexpressing tumors and down-regulated in tu-
`mors by VEGF blockade. Using VelociGene® technol-
`ogy, which provides a high-throughput approach to create
`mouse mutants for genes of interest (Valenzuela et al.
`2003), we found that mice lacking Dll4 exhibit profound
`vascular defects early in development (Gale et al. 2004).
`Remarkably, and as previously seen only for VEGF (see
`above), deletion of even just one of the two Dll4 alleles in
`developing embryos resulted in embryonic lethality due to
`vascular defects (Gale et al. 2004). All this evidence for a
`critical role for Dll4 in normal as well as tumor angiogen-
`esis provided a rationale to develop blockers for Dll4. Re-
`cent testing in tumor models indicates that Dll4 may in-
`deed prove to be an important new antiangiogenesis
`target, either alone or in combination with the VEGF Trap,
`or in settings of relative resistance to anti-VEGF therapies.
`
`SUMMARY
`
`In summary, using our Trap technology platform
`(Economides et al. 2003), we have created the VEGF
`Trap, a very potent VEGF blocking agent with excellent
`pharmacological properties. This drug has proven to be
`highly efficacious in a number of diverse preclinical
`models. It dramatically inhibits the growth of a variety of
`types of tumors and can even cause frank tumor regres-
`sion in some settings. In other preclinical cancer models,
`we have found that combination of VEGF Trap with a cy-
`totoxic agent can result in potency far greater than that of
`either single agent. Furthermore, the VEGF Trap is also
`very effective in animal models of vascular eye diseases.
`The impressive efficacy in preclinical models of cancer
`and eye diseases provided a rationale for advancement of
`the VEGF Trap into clinical trials, where it is producing
`promising initial results in both cancer and eye diseases.
`In addition to its potential therapeutic value in cancer
`and vascular eye diseases, the VEGF Trap is also an in-
`valuable research tool. Transcription profiling screens us-
`ing VEGF Trap have allowed a number of strategies de-
`signed to identify new antiangiogenesis targets. It is
`hoped that these strategies are helping to identify the next
`generation of antiangiogenesis targets, which may work
`either alone or in combination with the VEGF Trap, or in
`settings of relative resistance to anti-VEGF therapies.
`
`ACKNOWLEDGMENTS
`
`The authors gratefully acknowledge the substantial
`contributions of our colleagues at Regeneron, in particu-
`lar, Jocelyn Holash, Susan D. Croll, Lillian Ho, Michelle
`Russell, Patricia Boland, Ray Leidich, Donna Hylton,
`Shelly Jiang, Sarah Nandor, Alexander Adler, Hua Jiang,
`Elena Burova, Irene Noguera, Ella Ioffe, Tammy Huang,
`Czeslaw Radziejewski, Calvin Lin, Jingtai Cao, Kevin
`Bailey, James Fandl, Tom Daly, Eric Furfine, Jesse
`Cedarbaum, and Neil Stahl. In addition, we acknowledge
`our collaborators on VEGF Trap studies, including Jes-
`sica Kandel, Darrell Yamashiro, Robert Jaffe, Donald
`McDonald, Murray Korc, Phyllis Wachsberger, Adam
`Dicker, Tony Adamis, C. Cursiefen, J.W. Streilein, and
`Peter Campochiaro. We regret if we have omitted key
`contributors.
`
`REFERENCES
`Bergsland E.K. 2004. Update on clinical trials targeting vascular
`endothelial growth factor in cancer. Am. J. Health Syst.
`Pharm. 1: 61 (21 suppl. 5): S12–20. Review.
`Brown D.M. et al. IOVS 2006. 47: ARVO E-Abstract 2963.
`Byrne A.T., Ross L., Holash J., Nakanishi M., Hu L., Hofmann
`J.I., Yancopoulos G.D., and Jaffe R.B. 2003. Vascular en-
`dothelial growth factor-trap decreases tumor burden, inhibits
`ascites, and causes dramatic vascular remodeling in an ovar-
`ian cancer model. Clin. Cancer Res. 9: 5721.
`
`0005
`
`

`

`Downloaded from
`
`symposium.cshlp.org
`
`
`
`Cold Spring Harbor Laboratory Press on November 9, 2017 - Published by
`
`416
`
`RUDGE ET AL.
`
`Carmeliet P. 2000. Mechanisms of angiogenesis and arteriogen-
`esis. Nat. Med. 6: 389.
`Carmeliet P., Ferreira V., Breier G., Pollefeyt S., Kieckens L.,
`Gertsenstein M., Fahrig M., Vandenhoeck A., Harpal K.,
`Eberhardt C., Declercq C., Pawling J., Moons L., Collen D.,
`Risau W., and Nagy A. 1996. Abnormal blood vessel devel-
`opment and lethality in embryos lacking a single VEGF allele.
`Nature 380: 435.
`Cursiefen C., Cao J., Chen L., Liu Y., Maruyama K., Jackson D.,
`Kruse F.E., Wiegand S.J., Dana M.R., and Streilein J.W.
`2004. Inhibition of hemangiogenesis and lymphangiogenesis
`after normal-risk corneal transplantation by neutralizing
`VEGF promotes graft survival. Invest. Ophthalmol. Vis. Sci.
`45: 2666.
`Dalal S., Berry A.M., Cullinane C.J., Mangham D.C., Grimer R.,
`Lewis I.J., Johnston C., Laurence V., and Burchill S.A. 2005.
`Vascular endothelial growth factor: A therapeutic target for
`tumors of the Ewing’s sarcoma family. Clin. Cancer Res. 11:
`2364.
`Davis S., Aldrich T.H., Jones P.F., Acheson A., Compton D.L.,
`Jain V., Ryan T.E., Bruno J., Radziejewski C., Maisonpierre
`P.C., and Yancopoulos G.D. 1996. Isolation of angiopoietin-
`1, a ligand for the TIE2 receptor, by secretion-trap expression
`cloning. Cell 87: 1161.
`de Vries C., Escobedo J.A., Ueno H., Houck K., Ferrara N., and
`Williams L.T. 1992. The fms-like tyrosine kinase, a receptor
`for vascular endothelial growth factor. Science 255: 989.
`Dupont J., Rothenberg M.L., Spriggs D.R., Cedarbaum J.M.,
`Furfine E.S., Cohen D.P., Dancy I., Lee H., Cooper W., and
`Lockhart A.C. 2005. Safety and pharmacokinetics of intra-
`venous VEGF Trap in a phase I clinical trial of patients with
`advanced solid tumors. Proc. Am. Soc. Clin. Oncol. 23: 3029.
`Dvorak H.F., Nagy J.A., Feng D., Brown L.F., and Dvorak A.M.
`1999. Vascular permeability factor/vascular endothelial
`growth factor and the significance of microvascular hyperper-
`meability in angiogenesis. Curr. Top. Microbiol. Immunol.
`237: 97.
`Economides A.N., Carpenter L.R., Rudge J.S., Wong V.

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