throbber
MEDICALSCIENCES
`
`VEGF-Trap: A VEGF blocker with potent
`antitumor effects
`
`Jocelyn Holash*, Sam Davis, Nick Papadopoulos, Susan D. Croll, Lillian Ho, Michelle Russell, Patricia Boland, Ray Leidich,
`Donna Hylton, Elena Burova, Ella Ioffe, Tammy Huang, Czeslaw Radziejewski, Kevin Bailey, James P. Fandl, Tom Daly,
`Stanley J. Wiegand, George D. Yancopoulos, and John S. Rudge
`
`Regeneron Pharmaceuticals, Incorporated, 777 Old Saw Mill River Road, Tarrytown, NY 10591
`
`Communicated by P. Roy Vagelos, Merck & Co., Inc., Bedminster, NJ, July 2, 2002 (received for review April 19, 2002)
`
`Vascular endothelial growth factor (VEGF) plays a critical role
`during normal embryonic angiogenesis and also in the pathological
`angiogenesis that occurs in a number of diseases, including cancer.
`Initial attempts to block VEGF by using a humanized monoclonal
`antibody are beginning to show promise in human cancer patients,
`underscoring the importance of optimizing VEGF blockade. Previ-
`ous studies have found that one of the most effective ways to block
`the VEGF-signaling pathway is to prevent VEGF from binding to its
`normal receptors by administering decoy-soluble receptors. The
`highest-affinity VEGF blocker described to date is a soluble decoy
`receptor created by fusing the first three Ig domains of VEGF
`receptor 1 to an Ig constant region; however, this fusion protein
`has very poor in vivo pharmacokinetic properties. By determining
`the requirements to maintain high affinity while extending in vivo
`half life, we were able to engineer a very potent high-affinity VEGF
`blocker that has markedly enhanced pharmacokinetic properties.
`This VEGF-Trap effectively suppresses tumor growth and vascular-
`ization in vivo, resulting in stunted and almost completely avas-
`cular tumors. VEGF-Trap-mediated blockade may be superior to
`that achieved by other agents, such as monoclonal antibodies
`targeted against the VEGF receptor.
`
`The sprouting of new blood vessels, termed angiogenesis, is
`
`required to support growth in the embryo and young animal,
`as well as to allow for repair and remodeling processes in the
`adult. However, aberrant angiogenesis is also associated with a
`number of pathological conditions and diseases, including cancer
`(1, 2). Tumors, like many normal tissues, use the vasculature as
`a means to obtain oxygen and nutrients and to remove waste
`products. Although tumors can in part grow by coopting existing
`host vessels (3–6), most tumors also induce new vessel formation,
`suggesting that this neovascularization is required for their
`growth (1, 2, 7). Consequently, much effort has been directed
`toward discovering antiangiogenic agents and evaluating them as
`cancer therapeutics. Perhaps the best characterized and most
`highly validated antiangiogenic approach involves targeting the
`vascular endothelial growth factor (VEGF) pathway (1, 8–11).
`Based on numerous animal studies, the VEGF pathway is the
`only well-defined signaling pathway known to be required for
`normal development of the vasculature as well as for the
`pathologic angiogenesis that accompanies cancer and other
`disease states (8–10).
`The VEGF pathway is initiated when VEGF binds to its
`receptors on endothelial cells. The two best characterized VEGF
`receptors are termed VEGF receptor 1 (VEGFR1) and VEGF
`receptor 2 (VEGFR2). VEGFR1 and VEGFR2 are highly
`related transmembrane tyrosine kinases that use their ectodo-
`mains to bind VEGF; this binding in turn activates the intrinsic
`tyrosine kinase activity of their cytodomains, initiating intracel-
`lular signaling. Interestingly, although VEGFR1 binds to VEGF
`with substantially higher affinity, most of the biologic effects of
`VEGF seem to be mediated via VEGFR2. In animals, blockade
`of the VEGF pathway has been achieved by many different
`means, including blocking antibodies targeted against VEGF
`(12–14) or its receptors (15), soluble decoy receptors that
`
`prevent VEGF from binding to its normal receptors (16–20), as
`well as small molecule inhibitors of the tyrosine kinase activity
`of the VEGFRs (21–23). Recently, a study that compared the
`efficacy of VEGF blockade to other ‘‘antiangiogenic’’ strategies
`established that this approach is superior to many others (ref.
`11). Consistent with predictions from animal studies, blockade
`of VEGF using a humanized monoclonal antibody has emerged
`as the first and thus far only antiangiogenesis approach reporting
`promising results in human cancer patients, based on prelimi-
`nary reports from early clinical trials.† The hope is that anti-
`VEGF approaches can be generalized to many different types of
`cancer, as well as to other diseases in which pathologic angio-
`genesis contributes, such as diabetic retinopathy and psoriasis.
`The clinical promise of initial anti-VEGF approaches high-
`lights the need to optimize blockade of this pathway. Previous
`studies have found that one of the most effective ways to block
`the VEGF signaling pathway is to prevent VEGF from binding
`to its normal receptors by administering decoy VEGF receptors
`(11, 16, 17, 24). The highest-affinity VEGF blocker described to
`date is a soluble decoy receptor created by fusing the first three
`Ig domains of VEGFR1 to the constant region (Fc portion) of
`human IgG1, resulting in a forced homodimer that has picomolar
`binding affinity (16, 17). In tumor experiments, this VEGFR1-Fc
`reagent is efficacious at approximately 500-fold lower concen-
`tration than a similar VEGFR-2 construct (11). Despite its high
`affinity, the VEGFR1-Fc is not a feasible clinical candidate
`because of its poor pharmacokinetic profile; in rodent studies,
`this protein has to be administered frequently and at very high
`levels to achieve efficacious levels (16, 17, 24). In addition, the
`VEGFR1-Fc exhibits certain toxicological side effects that are
`not seen with the VEGFR2-Fc (11). These effects appear to be
`due to nonmechanism-based and nonspecific properties of this
`agent (see Discussion). By determining the requirements to
`maintain high affinity while extending in vivo half life, we were
`able to engineer a very potent high-affinity VEGF blocker that
`has prolonged in vivo pharmacokinetics and pharmacodynamics,
`lacks nonspecific toxicities, and can effectively suppress the
`growth and vascularization of a number of different types of
`tumors in vivo.
`
`Materials and Methods
`Engineering VEGF-Traps. The parental VEGF-Trap was created by
`fusing the first three Ig domains of VEGFR1 to the constant
`region (Fc) of human IgG1. VEGF-Trap⌬B1 was created by
`removing a highly basic 10-aa stretch from the third Ig domain
`of the parental VEGF-Trap. VEGF-Trap⌬B2 was created by
`removing the entire first Ig domain from VEGF-Trap⌬B1.
`VEGF-TrapR1R2 was created by fusing the second Ig domain
`
`Abbreviations: VEGF, vascular endothelial growth factor; VEGFR1, VEGF receptor 1;
`VEGFR2, VEGF receptor 2; AUC, area under the curve.
`
`*To whom reprint requests should be addressed. E-mail: jocelyn.holash@regeneron.com.
`†Yang, J., Haworth, L., Steinberg, S., Rosenberg, S. & Novotny, W. (2002) Am. Soc. Clin.
`Oncol. (abstr. 15).
`
`www.pnas.org兾cgi兾doi兾10.1073兾pnas.172398299
`
`PNAS 兩 August 20, 2002 兩 vol. 99 兩 no. 17 兩 11393–11398
`
`CELLTRION - EXHIBIT 1004
`
`

`

`of VEGFR1 with the third Ig domain of VEGFR2. All of the
`VEGF-Trap variants were produced and purified from Chinese
`hamster ovary cells.
`Pharmacokinetic Analysis of VEGF-Traps. BALB兾c mice (25–30 g)
`were injected s.c. with 4 mg兾kg of the various Traps and bled at
`1, 2, 4, 6, 24, 48, 72, and 144 hr after injection. Levels of all
`VEGF-Traps were measured by an ELISA by using human
`VEGF165 to capture and an antibody to the human Fc region as
`the reporter.
`
`Extracellular Matrix (ECM)-Binding Assay. ECM-coated plates
`(Becton Dickinson no. 35–4607) were incubated with varying
`concentrations of VEGF-Traps for 1 hr at room temperature.
`They were washed and incubated with alkaline phosphatase-
`conjugated anti-human Fc antibody (Promega, 1:4,000 in PBS ⫹
`10% BCS) for 1 hr at room temperature. Plates were washed four
`times with PBS ⫹ 0.1%Triton-X 100 and reagent buffer added
`for color development. Plates were read at 405–570 nm.
`
`VEGF-Trap-Binding Assay. Binding affinities of VEGF-Traps were
`measured by using a specific and sensitive ELISA (R&D Systems
`kit no. DVE00) for detecting free (unbound) human VEGF in
`mixtures of the VEGF-Traps (ranging in concentration from 0.1
`to 160 pM) with human VEGF165 (at 10 pM), incubated over-
`night at room temperature.
`
`Human Umbilical Vein Endothelial Cell Phosphorylation Assay. Con-
`fluent monolayers of human umbilical vein endothelial cells [Vec
`Technologies (Rensselaer, NY) passage no. 5] were serum-starved
`for 2 hr and then challenged for 5 min with vehicle or 40 ng兾ml of
`human VEGF165, alone or preincubated with VEGF-Traps at
`1.5-fold molar excess. Cells were then lysed, immunoprecipitated by
`using a VEGFR2-specific antibody, and immunoblotted with
`a phosphotyrosine-specific antibody (Upstate Biotechnology,
`4G10 mAb).
`
`VEGF-Induced Proliferation Assay. Cells that proliferate in response
`to VEGF were generated by stably transfecting NIH 3T3 cells
`with a VEGFR2兾TrkB chimeric receptor (in which the cytodo-
`main of VEGFR2 was replaced with that of TrkB, a receptor for
`brain-derived neurotrophic factor that effectively drives prolif-
`eration in these cells). Five thousand cells were plated per well
`of a 96-well plate, allowed to settle for 2 hr, incubated for 1 hr
`with VEGF-Trap variants (titrated from 40 nM to 20 pM), then
`challenged for 72 hr with human VEGF165 at a concentration of
`1.56 nM, followed by addition of [3-(4,5 dimethylthiazol-2-yl)-
`5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium,
`innersai and spectrophotometric analysis at 450兾570 nm.
`
`Acute Hypotension. Male Wistar–Kyoto rats (180–240 g) from
`Taconic Laboratories were maintained on a 12:12 light兾dark cycle
`(lights on 0600) with food and water available ad libitum. Before
`challenge with VEGF, animals were pretreated with VEGF-Traps
`or PBS as indicated, anesthetized with 1.5–2% isoflurane in oxygen,
`and the left femoral artery catheterized for direct measurement of
`systolic blood pressure through a blood pressure transducer (IITC,
`Woodland Hills, CA) into a chart recorder (Linseis, Princeton
`Junction, NJ). Animals were then injected in the right jugular vein
`with a 200-␮l bolus containing 10 ␮g of recombinant human
`VEGF165. Systolic blood pressure was measured before VEGF
`injection and every minute thereafter for 20 min. Blood pressures
`were normalized to baseline preinjection and analyzed by using
`mixed factorial ANOVAs (see supporting information on the
`PNAS web site, www.pnas.org).
`Tumor Growth Experiments. C6 glioma cells (1.0 ⫻ 106 cells兾mouse)
`and A673 rhabdomyosarcoma cells (2.0 ⫻ 106 cells兾mouse) were
`
`Engineering of VEGF-Traps with improved pharmacokinetics. (A)
`Fig. 1.
`Schematics of full-length VEGFR1 (red) and VEGFR2 (blue) are provided,
`indicating their seven Ig domains, transmembrane regions (black bars), and
`kinase domains (ovals). The parental VEGF-Trap contains the first three Ig
`domains of VEGFR1 (including the highly basic 10-aa stretch in Ig3, blue box)
`fused to the Fc portion of human IgG1. VEGF-Trap⌬B1 is identical to the
`parental VEGF-Trap, except that the basic stretch in Ig3 has been removed.
`VEGF-Trap⌬B2 is the same construct as ⌬B1, except that the first Ig domain has
`been removed. VEGF-TrapR1R2 possesses the second Ig domain of VEGFR1 and
`the third Ig domain of VEGFR2 fused to the Fc portion of human IgG1. (B) The
`four indicated VEGF-Traps were assayed in vitro for their capacity to bind to
`extracellular matrix, with only the parental VEGF-Trap and VEGF-Trap⌬B1
`demonstrating binding. (C) Pharmacokinetic analysis of the VEGF-Traps re-
`veals that the parental VEGF-Trap has the poorest profile, whereas VEGF-
`TrapR1R2 showed the best profile.
`
`obtained from American Type Culture Collection, and B16F10.9
`melanoma cells (5.0 ⫻ 105 cells兾mouse) were a generous gift from
`Charles Lin (Duke University, Durham, NC). Cells were suspended
`in serum-free medium and implanted s.c. on the shaved right flank
`of male C.B-17 SCID mice at the indicated concentrations. After
`tumor cell implantation and twice weekly thereafter for the dura-
`tion of the experiment, mice received a s.c. injection (at the nape of
`the neck) of vehicle (PBS ⫹ 0.5% glycerol), VEGF-Trap, or DC101
`(from American Type Culture Collection). After 2–3.0 weeks,
`animals were killed and tumors were measured ex vivo with calipers
`(tumor volume ⫽ length ⫻ width ⫻ height). For immunohisto-
`chemistry studies, mice were perfused with 4% paraformaldehyde,
`and tissue was processed as previously described (25).
`
`Results
`Reengineering Parental VEGF-Trap to Improve Its Pharmacokinetic
`Profile. On the basis of the previously reported high affinity of a
`soluble decoy receptor in which VEGFR1 is fused to the Fc
`portion of human IgG1 (16, 17), we produced this fusion protein
`to study its properties (see parental VEGF-Trap, Fig. 1A). Single
`s.c. injections of parental VEGF-Trap (4 mg兾kg) into mice were
`
`11394 兩 www.pnas.org兾cgi兾doi兾10.1073兾pnas.172398299
`
`Holash et al.
`
`

`

`MEDICALSCIENCES
`
`Binding affinity and inhibitory properties of VEGF-Traps. (A) Affini-
`Fig. 2.
`ties of indicated VEGF-Traps for VEGF, as determined by using a binding assay
`that measures unbound VEGF (ordinate) after incubation of 10 pM of human
`VEGF165 with varying concentrations of VEGF-Traps (abscissa). (B) Inhibition of
`VEGF-induced phosphorylation of VEGFR2 in human umbilical vein endothe-
`lial cell phosphorylations using indicated VEGF-Traps at 1.5-fold molar excess,
`as revealed with immunoblotting assay. (C) Inhibition of VEGF-induced pro-
`liferation of fibroblasts containing a chimeric VEGFR2兾TrkB receptor, using
`varying concentrations of VEGF-Traps in the presence of 1.56 nM of VEGF.
`
`analyses show that VEGF-TrapR1R2 has a kD of⬇ 1–10 pM for
`VEGF121 and approximately 45 pM for placental growth factor 2
`(not shown); other VEGF isoforms and relatives have not been
`analyzed.
`To determine whether Trap binding of VEGF could potently and
`effectively block the ability of VEGF to activate its receptor, VEGF
`and Traps were added to cultured endothelial cells, and the effects
`on VEGFR2 phosphorylation were examined, revealing that both
`parental VEGF-Trap as well as VEGF-TrapR1R2 can completely
`block VEGF-induced VEGFR2 phosphorylation when added at a
`1.5-fold molar excess compared with the added VEGF, consistent
`with very high-affinity binding to VEGF (Fig. 2B). Finally, to assess
`whether these Traps would also be effective in cell-based prolifer-
`ation assays, we engineered a cell line containing a chimeric
`VEGFR2 receptor that mediates a very strong proliferative re-
`sponse to VEGF and found that both parental VEGF-Trap and
`VEGF-TrapR1R2 potently blocked VEGF-induced proliferation in
`3-day growth assays in these cells, with an IC50 at approximately an
`equimolar concentration of Trap with the added VEGF, once again
`consistent with very high-affinity binding of the Traps for VEGF
`(Fig. 2C).
`
`VEGF-TrapR1R2 Provides Long-Term Blockade of Exogenously Admin-
`istered VEGF-Induced Acute Hypotension. The above studies indi-
`cated that VEGF-TrapR1R2 was at least as impressive a blocker
`of VEGF as the parental version, but that it had far superior
`pharmacokinetic properties. To initially explore whether these
`attributes translated into superior pharmacodynamic perfor-
`mance, we compared these reagents by using an acute readout
`of VEGF responsiveness in vivo. Administration of a single bolus
`dose (10 ␮g) of recombinant VEGF165 to rats results in acute
`hypotension, with a drop of about 40% from baseline systolic
`blood pressure; this drop is maximal at 5 min and slowly rectifies
`to normal by about 30 min (Fig. 3A). To compare the pharma-
`codynamic efficacy of the VEGF-Traps in blocking this acute
`response, we preadministered the parental VEGF-Trap or
`
`performed to confirm that it indeed displayed poor pharmaco-
`kinetic properties, with a maximal concentration (Cmax) of only
`0.05 ␮g兾ml and total ‘‘area under the curve concentration’’
`(AUC) of 0.04 ␮g ⫻ days兾ml (Fig. 1C). We postulated that these
`poor pharmacokinetic properties might be due to the high
`positive charge of this protein (pI 9.4), which in turn may result
`in its deposition at the site of s.c. injection because of nonspecific
`adhesion to highly negatively charged proteoglycans that com-
`prise the extracellular matrix. To test this hypothesis, we next
`engineered several variants of the parental VEGF-Trap with
`reduced positive charges. On review of the charge density in the
`parental molecule, we noted a highly basic stretch of 10 amino
`acids in the third Ig domain of VEGFR1 (see blue box in Fig.
`1 A). To reduce the charge, this region was excised, resulting in
`a decrease in the pI of this VEGF-Trap (termed VEGF-Trap⌬B1;
`see Fig. 1 A) from 9.4 to 9.1. It was also noted that the first Ig
`domain of VEGFR1 had a basic pI, and we thus decided to test
`removal of this domain as well as the above-noted basic region,
`resulting in a protein termed VEGF-Trap⌬B2 (Fig. 1 A), with a
`further reduced pI of 8.9. Finally, because the third Ig domain of
`VEGFR2 has a lower pI than the corresponding domain of
`VEGFR1, we simply switched these domains to make a Trap in
`which the second Ig domain of VEGFR1 is directly fused to the
`third Ig domain of VEGFR2; this trap was termed VEGF-
`TrapR1R2 (Fig. 1 A) and had a pI of 8.82. Previous structural
`analyses indicated that VEGFR1 might make greater use of its
`second Ig domain in contacting VEGF, whereas VEGFR2
`instead makes greater use of its third Ig domain (26), raising the
`interesting and useful possibility that VEGF-TrapR1R2 might
`actually bind more tightly to VEGF than the parental versions.
`Combining the distinct binding regions of two different recep-
`tors to create a higher-affinity interactor has previously been
`used in the creation of a series of interleukin and cytokine
`blockers also termed Traps (A. Economides, L. Rocco Carpen-
`ter, J.S.R., V. Wong, E. Koehler-Stec, C. Hartnett, E. Pyles, T.D.,
`M. Young, J.P.F., Frank Lee, Scott Carver, Jennifer McNay,
`K.B., S. Ramakanth, R. Hatabarat, C.R., T.H., G.D.Y., and
`N. Stahl, unpublished results). Using a simple extracellular
`matrix-binding assay, we then confirmed the hypothesis that
`decreasing the positive charge of the VEGF-Traps would result
`in decreased adhesion to extracellular matrix (Fig. 1B). Binding
`to extracellular matrix in this assay was directly related to the pI
`of the Traps, with both VEGF-TrapR1R2 and VEGF-Trap⌬B2
`displaying negligible binding in this assay.
`On the basis of the above results, we next tested these various
`VEGF-Traps in vivo for their pharmacokinetic behavior. Their in
`vivo behavior followed the theoretical charge predictions as well as
`the in vitro adhesion properties. Every reduction in pI was accom-
`panied by a corresponding improvement in Cmax and AUC: VEGF-
`Trap⌬B1 had a Cmax of 1.3 ␮g兾ml and an AUC of 1.36 ␮g ⫻ days兾ml;
`VEGF-Trap⌬B2 had a Cmax of 2.65 ␮g兾ml and an AUC of 5.42 ␮g ⫻
`days兾ml; whereas VEGF-TrapR1R2 revealed the best profile with a
`Cmax of 16 ␮g兾ml and an AUC of 36.28 ␮g ⫻ days兾ml (Fig. 1C).
`Thus, VEGF-TrapR1R2 had an AUC that was almost 1,000-fold
`higher than that of the parental VEGF-Trap, raising the possibility
`that it might be a far superior pharmacologic agent, assuming it
`retained its ability to bind and block VEGF.
`
`Comparison of Parental VEGF-Trap with VEGF-TrapR1R2 in Binding,
`Phosphorylation, and Cell Proliferation Assays inVitro. Because of the
`superior pharmacokinetic properties of VEGF-TrapR1R2, we next
`compared this Trap to its parent for its ability to bind and block
`VEGF in vitro. To determine binding affinity of the Traps for
`VEGF, equilibrium binding assays were performed in which dif-
`ferent concentrations of the Traps were incubated with VEGF165,
`and the amount of unbound VEGF165 was measured, revealing that
`parental VEGF-Trap displays a kD of ⬇5 pM, whereas VEGF-
`TrapR1R2 has a binding affinity of about 1 pM (Fig. 2A). Preliminary
`
`Holash et al.
`
`PNAS 兩 August 20, 2002 兩 vol. 99 兩 no. 17 兩 11395
`
`

`

`Using blockade of VEGF-induced acute hypotension to pharmaco-
`Fig. 3.
`dynamically compare VEGF-Traps. (A) When rats were treated with VEGF-Traps at
`25 mg兾kg at 1 day before VEGF challenge, VEGF-TrapR1R2 (n ⫽ 8) completely
`blocked VEGF-induced hypotension, whereas PBS (n ⫽ 6) and parental VEGF-Trap
`(n ⫽ 6) were ineffective. ANOVA shows treatment effect, P ⬍ 0.007. (B) At a 5-fold
`lower dose (5 mg兾kg), VEGF-TrapR1R2 was still effective at 1 day (n ⫽ 4) or 3 days
`(n ⫽ 3) before the VEGF challenge. ANOVA shows treatment effect, P ⬍ 0.03.
`
`VEGF-TrapR1R2 at 25 mg兾kg, 24 hr before VEGF administra-
`tion (Fig. 3A). Consistent with what would be expected from the
`above pharmacokinetic studies, this dose of VEGF-TrapR1R2
`completely blocked VEGF-induced hypotension, whereas the
`parental VEGF-Trap had no discernable effect. Thus, although
`the parental VEGF-Trap and its VEGF-TrapR1R2 derivative are
`quite comparable in vitro (see above), the VEGF-TrapR1R2
`performs much better in vivo, presumably because of its dra-
`matically enhanced pharmacokinetic profile.
`To further characterize the length of time in which VEGF-
`TrapR1R2 remained efficacious, we waited 1, 3, and 7 days after
`injection of the Trap at 5 mg兾kg before inducing hypotension. At
`this dose, VEGF-TrapR1R2 was completely effective in blocking
`VEGF-induced acute hypotension at 1 and 3 days after a single
`bolus (Fig. 3B) but was not significantly different from controls
`at 7 days (data not shown).
`
`VEGF-TrapRiR2 Dramatically Blocks Tumor Growth inVivo. Altogether,
`the above pharmacokinetic and pharmacodynamic studies indi-
`cated that VEGF-TrapR1R2 has the potential to be a long-term
`and potent pharmacologic blocker of VEGF-mediated activities
`in vivo, far superior to that of parental VEGF-Trap. To begin to
`explore the value of VEGF-TrapR1R2 as an anticancer thera-
`peutic and to compare it to other effective agents targeting the
`VEGF pathway, we evaluated its ability to block the growth of
`a variety of tumor cell lines in s.c. tumor models in mice. Tumor
`cells were derived from diverse tissue origins and different
`species (mouse B16F10.9 melanoma, human A673 rhabdomyo-
`sarcoma, and rat C6 glioma). After implantation of tumor cells,
`mice were allowed a brief recovery period and then received s.c.
`injections of VEGF-TrapR1R2 (25 mg兾kg) or vehicle twice weekly
`for the duration of the experiment (2–3.0 weeks), after which the
`
`Fig. 4. VEGF-TrapR1R2 dramatically inhibits the s.c. growth and vascularity of
`implanted tumors from diverse tissues and species. (A) VEGF-Trap R1R2 sub-
`stantially blocked the growth of the indicated s.c. implanted tumors, at the
`indicated doses twice weekly for 2 weeks (C6 and B16F10.9) or 3.0 weeks
`(A673). Error bars represent standard error of mean, n ⫽ five mice兾treatment
`group. The differences between control tumor volumes and VEGF-TrapR1R2–
`treated tumor volumes were analyzed by using Student’s t tests and found to
`be significant at the following levels: B16F10 P ⫽ 0.01; A673 P ⫽ 0.06; C6 P ⬍
`0.0001. (B–D) Histological analysis reveals that VEGF-TrapR1R2 can effectively
`block blood vessel growth in these implanted tumors. Sections of C6 tumors
`stained with antibodies to platelet– endothelial cell adhesion molecule reveal
`that vehicle-treated animals had large tumors that were highly vascularized
`(B), whereas animals treated with 25 mg兾kg VEGF-TrapR1R2 (C) had tumors that
`were largely avascular with large areas of necrosis (N). Viable tumor appeared
`to be vascularized because of cooption of preexisting host vessels (white
`arrowheads) associated with hypodermal musculature (M) and dermis. Treat-
`ment with 2.5 mg兾kg VEGF-TrapR1R2 greatly stunted tumor growth (C) and
`resulted in large necrotic regions (N), although small pockets of vessels were
`occasionally apparent (black arrows). (Bar ⫽ 100 ␮m.)
`
`animals were killed and tumors excised and measured. VEGF-
`TrapR1R2 significantly inhibited the growth of all three types of
`tumors (Fig. 4A). In the study using C6 glioma cells, a 10-fold
`lower dose of VEGF-TrapR1R2 (2.5 mg兾kg) was tested and found
`to be equally effective at inhibiting tumor growth.
`To evaluate the effects of VEGF-TrapR1R2 on tumor-associated
`angiogenesis, the tumors from the above studies were sectioned and
`immunostained with antibodies to platelet–endothelial cell adhe-
`sion molecule, so that the vasculature could be visualized (Fig. 4
`B–D). This analysis revealed that the higher dose of VEGF-
`TrapR1R2 almost completely blocked tumor-associated angiogene-
`sis, with the stunted tumors being largely avascular, save for regions
`in which preexisting host vessels appeared to be coopted by
`surrounding tumor (see open arrowheads, Fig. 4C). The lower dose
`of VEGF-TrapR1R2, which was quite comparable at inhibiting
`tumor growth (see above), appeared to be slightly less effective at
`completely blocking tumor-associated angiogenesis, allowing for
`small pockets of tumor-associated vessels in otherwise avascular
`tumors (see black arrowheads in Fig. 4D). In contrast to the
`VEGF-Trap-treated tumors, control tumors in vehicle-treated mice
`not only were much larger (see above) but also had a very high
`vascular density (Fig. 4B).
`
`11396 兩 www.pnas.org兾cgi兾doi兾10.1073兾pnas.172398299
`
`Holash et al.
`
`

`

`MEDICALSCIENCES
`
`against a wide assortment of cancers. In addition, pathological
`angiogenesis seems to contribute to a number of non-neoplastic
`diseases, such as diabetic retinopathy (31) and psoriasis (32),
`extending the potential utility of anti-VEGF therapeutics. All this
`promise highlights the need to optimize anti-VEGF approaches.
`Herein we describe the engineering of an anti-VEGF agent, termed
`VEGF-TrapR1R2. VEGF-TrapR1R2 is a derivative of perhaps the
`most potent VEGF binder known, VEGFR1. Soluble forms of
`VEGFR1 suffer from poor pharmacokinetic properties, which
`seem to correlate with their nonspecific interactions with extracel-
`lular matrix. VEGF-TrapR1R2 was engineered to have minimal
`interactions with extracellular matrix, and this property apparently
`accounts for its satisfying pharmacokinetic profile. The combina-
`tion of high-affinity and improved pharmacokinetics apparently
`contributes toward making VEGF-TrapR1R2 one of the most, if not
`the most, potent and efficacious VEGF blocker available. An
`additional advantage is that VEGF-TrapR1R2 is composed of en-
`tirely human sequences, hopefully minimizing the possibility that it
`might prove immunogenic in human patients. Despite its wholly
`human nature, VEGF-TrapR1R2 binds all species of VEGF tested,
`from human to chicken VEGF (not shown), making it a very
`versatile reagent that can be used in almost any experimental
`animal models.
`A recent study comparing numerous antiangiogenesis ap-
`proaches concluded that anti-VEGF approaches were the most
`efficacious (11). The particular anti-VEGF agent used for these
`studies was essentially equivalent to our parental VEGF-Trap
`but was delivered in an adenoviral system in which it was highly
`expressed in the livers of infected animals. In contrast to other
`anti-VEGF approaches that seem to be well-tolerated, the
`adenovirally delivered VEGF-Trap caused severe liver toxicity
`and ascites, raising the possibility that it might have some unique
`mechanism-based side effects compared with other anti-VEGF
`approaches. To explore this possibility, we made adenoviral
`versions of both the parental VEGF-Trap as well as VEGF-
`TrapR1R2 and found that, whereas adenoviral delivery of paren-
`tal VEGF-Trap reproduces the previously reported toxicities
`(11), adenoviral delivery of VEGF-TrapR1R2 did not cause these
`side effects even though much higher levels were achieved in the
`circulation. Our conclusion is that the nonspecific interactions of
`the parental VEGF-Trap with extracellular matrix contribute to
`its increased toxicity after adenoviral administration, and that
`comparable toxicity is not noted with adenoviral administration
`of the engineered VEGF-TrapR1R2.
`In addition to the anticancer findings reported here, recent
`studies have shown that various versions of the VEGF-Trap can
`efficaciously treat a cancer-associated condition in mice similar
`to liver peliosis (33), as well as noncancer-associated disease
`models, such as of diabetic retinopathy (34–36) and psoriasis
`(Y.-P. Xia, M. Detmar, G.D.Y., and J.S.R., unpublished results).
`The accompanying manuscript (29) compares the efficacy of the
`VEGF-Trap to that of several other VEGF blockers, including
`a humanized monoclonal antibody to VEGF, in a model of
`kidney cancer. Among the several VEGF blockers tested, the
`VEGF-Trap shows the best overall efficacy. In this manuscript,
`we compare the efficacy of the VEGF-Trap to that of a
`monoclonal antibody to VEGFR2 in cancer models and find that
`far lower circulating levels of VEGF-TrapR1R2 are required for
`similar efficacy. Tumors treated with highest doses of the
`VEGF-Trap are not only stunted but also strikingly avascular.
`Our description of a VEGF blocker with such superior blocking and
`pharmacologic properties seems to demand that it be tested in
`human patients suffering from diseases involving neoangiogenesis.
`Toward this end, the safety of the VEGF-Trap has recently been
`confirmed in toxicological studies in cynomologus monkeys (data
`not shown). Consequently, the VEGF-Trap is currently in human
`clinical trials for several different types of cancer.
`
`VEGF-TrapR1R2 blocks tumor growth (of subcutaneously implanted
`Fig. 5.
`B16F10.9 cells) at far lower concentrations than DC101, a monoclonal antibody
`directed to VEGFR2. Mice were treated twice weekly with the indicated dose of
`VEGF-TrapR1R2, DC101, or vehicle. After 2.5 weeks, mice were killed, and tumors
`were excised and measured. Individual tumor volumes are shown (colored bars),
`as are average tumor volumes for each treatment (black bars) ⫾ SEM, n ⫽ six
`mice兾treatment group. Differences between treatment groups were analyzed by
`using a one-way ANOVA followed by Fisher’s protected least significant differ-
`ence test. Average volume of tumors in all treatment groups is significantly
`smaller than control tumor volume (P ⬍ 0.01). Differences in tumor volume
`between the high-dose VEGF-Trap, low-dose VEGF-Trap, and high-dose DC101
`treatment groups are not significantly different, but they are significantly dif-
`ferent from those of the low-dose DC101 treatment group (P ⬍ 0.02).
`
`VEGF-TrapR1R2 Compares Favorably with Antibodies Targeting VEGFR2.
`After establishing that VEGF-TrapR1R2 was effective at blocking
`s.c. tumor growth, we undertook studies to compare its efficacy with
`other known VEGF blockers. One particularly effective and well-
`characterized blocker is a monoclonal antibody, termed DC101,
`that targets VEGFR2 (15). When equimolar doses of VEGF-
`TrapR1R2 and DC101 were compared in the B16F10 melanoma
`model, it was apparent that much higher doses of DC101 are
`required to inhibit tumor growth (Fig. 5). Furthermore, because
`antibodies have longer circulation times in mice than simple Fc
`fusion proteins, the highly efficacious dose of DC101 accumulates
`to approximately 60-fold higher serum levels than that of the
`equally efficacious low dose of VEGF-Trap: circulating levels of
`DC101 in animals treated with the 40-mg兾kg dose were 2,442 ⫾ 272
`␮g兾ml, in contrast to the circulating levels of VEGF-Trap in
`animals treated with 3.2 mg兾kg, which were 40 ⫾ 8 ␮g兾ml. Thus,
`circulating levels of VEGF-Trap that were approximately 60-fold
`lower than those of DC101 were equally efficacious in inhibiting
`tumor growth. Importantly, the favorable allometric scaling of Fc
`fusion proteins relative to antibodies (27, 28) suggests that in
`humans the circulation time for the VEGF-Trap will be much more
`comparable to that of antibodies, which in turn suggests that in
`humans the difference in efficacious doses would be further mag-
`nified and may be as great as 60-fold.
`As described in an accompanying manuscript (29), when used
`at the same dose, VEGF-Trap shows efficacy equal to or better
`than a monoclonal antibody to VEGF (30). As noted above,
`because Fc fusion proteins have much shorter circulating half-
`lives than antibodies in mice, but comparable half-lives in
`humans, the finding that the VEGF-TrapR1R2 is at least as potent
`as the monoclonal antibody in mice suggests that the efficacious
`dose of VEGF-Trap will be much lower than that of the
`monoclonal antibody in humans.
`
`Discussion
`Validation of VEGF as an important new target in the war
`against cancer comes from pioneering clinical studies using a
`humanized monoclonal antibody that binds and blocks VEGF.†
`Because anti-VEGF approaches act by blocking tumor-a

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