throbber
'a
`
`Liu et at BMC Cancer (2015)15:170
`DOI 10.1186/512885-015-1140-1
`
`EXRIBff o2f
`
`K,4A(M COURTPpURrrnG
`
`BMC
`Cancer
`
`RESEARCH ARTICLE
`
`Open Access
`
`A novel engineered VEGF blocker with an
`excellent pharmacokinetic profile and robust
`anti-tumor activity
`
`Lily Liu', Haijia Vu 1, Xin Huartg 2, Hongzhi Tan', Song Li 2, Van Luo 1, Li Zhang3, Sumei Jiang t, Huifeng Jia 3,
`Yao Xiong 3, Ruliang Zhang4, Vi Huang 3, Charles C Ch U5,6, 1 and Wenzhi Tian"
`
`Abstract
`
`Background: Relatively poor penetration and retention in tumor tissue has been documented for large molecule
`drugs including therapeutic antibodies and recombinant immunoglobulin constant region (Fc)-fusion proteins due
`to their large size, positive charge, and strong target binding affinity. Therefore, when designing a large molecular
`drug candidate, smaller size, neutral charge, and optimal affinity should be considered.
`Methods: We engineered a recombinant protein by molecular engineering the second domain of VEGFR1 and a
`few flanking residues fused with the Fc fragment of human IgG 1, which we named HB-002.1. This recombinant
`protein was extensively characterized both in vitro and in vivo for its target-binding and target-blocking activities,
`pharmacokinetic profile, angiogenesis inhibition activity, and anti-tumor therapeutic efficacy.
`Results: HB-002.1 has a molecular weight of —80 kDa, isoelectric point of —6.7, and an optimal target binding
`affinity of <1 nM. The pharmacokinetic profile was excellent with a half-life of 5 days, maximal concentration of
`20.27 pg/mI, and area under the curve of 81.46 pg days/ml. When tested in a transgenic zebrafish embryonic
`angiogenesis model, dramatic inhibition in angiogenesis was exhibited by a markedly reduced number of subintestinal
`vessels. When tested for anti-tumor efficacy, HB-002,1 was confirmed in two xenograft tumor models I and
`Colo-205) to have a robust tumor killing activity, showing a percentage of inhibition over go% at the dose of
`20 mg/kg. Most promisingly, HB-002.1 showed a superior therapeutic efficacy compared to bevacizumab in the
`A54g xenograft model (tumor inhibition: 84.7% for HB-002.1 versus 67.6% for bevacizumab, P <0.0001).
`Conclusions: 1-18-002.1 is a strong angiogenesis inhibitor that has the potential to be a novel promising drug for
`angiogenesis-related diseases such as tumor neoplasms and age-related macular degeneration.
`
`Keywords: VEGF inhibitor, VEGFR1, Recombinant Fc-fusion protein, Anti-tumor therapy, Angiogenesis
`
`Background
`Targeted tumor therapy is the focus of recent intense
`drug development by the pharmaceutical industry with
`the primary interests centered on antibody drugs [1].
`However antibody and/or recombinant protein drugs
`with molecular weights (MWs) of over 100 kDa usually
`have relatively poor tumor penetration and retention
`capacity for which the molecular size, charge, as well as
`target binding affinity play important roles [2]. There are
`
`* Correspondence: tiant10602@huabobio.com
`Department or cell Biology, Huabo Biopharm Cc Ltd., shanghai 201203,
`china
`Full list of author information is available at the end of the article
`
`several barriers to large molecule transport in solid tu-
`mors due to disordered vasculature, tissue structure, as
`well as extracellular matrix (ECM). These factors, which
`impact penetration and retention of large molecule
`drugs, have to be considered when designing new mo-
`lecular constructs.
`Angiogenesis, the process by which the existing vascular
`network expands to form new blood vessels, is mainly me-
`diated by vascular endothelial growth factor (VEGF),
`which upon binding with VEGF receptor (VEGFR), can in-
`duce phosphorylation of the receptors expressed in the
`blood vessel endothelial cells [1], thus leading to prolifera-
`tion of the endothelial cells and the development of the
`
`(ii) BioMed Central
`
`e 2015 Liu et at, licensee BlaMed Central. This is an Open Access article distributed under the terms of the Creative Commons
`Attribution License (httpi/creativecommons org/licenses/by/2.0), which permits unrestricted use, distribution, and
`reproduction in any medium, provided the original work is properly credited. The Creative Commons Public Domain
`Dedication waiver thrtpi/c,eativecomnlons.org/publicdomairvzero/t.00 applies to the data made available in this article,
`unless otherwise stated
`
`Mylan Exhibit 1121
`Mylan v. Regeneron, IPR2021-00880
`Page 1
`
`

`

`Liu et al. BMC Cancer (2015)15:170
`
`Page 2 of 14
`
`vascular system. Under pathological conditions, VEGF-A
`and other members of the VEGF family including placen-
`tal growth factor (PIGF) are upregulated [3-6]. Among the
`factors contributing to angiogenesis, VEGF-A is the main
`ligand driving angiogenesis, making it an important target
`for drug development.
`Several drugs targeting VEGF have been approved for
`use in the treatment of cancer [7] as well as for wet age-
`related macular degeneration (AMD) [8]. Bevacizumab is
`a humanized antibody targeting VEGF-A and was ap-
`proved under the trade name of Avastin in 2004 for the
`treatment of metastatic colon cancer [9-11] as well as sev-
`eral other solid tumors including lung cancers [12,13),
`glioblastoma [14,15], renal cancers [16], and ovarian can-
`cers [17-19]. The main mechanism by which bevacizumab
`exerts anti-tumor activity is by preventing VEGF-A from
`binding with its receptors, thus resulting in inhibition of
`new blood vessel growth in tumor tissues. Bevacizumab is
`a humanized IgGi with over 90% of human and less than
`10% of murine components [20]. The recommended dose
`for bevacizumab is 5 mg/kg every 2 weeks, even though it
`could be detected in serum for 12 weeks [21]. Bevacizu-
`mab is the first VEGF blocker proven to improve survival
`by 30% in patients with metastatic colorectal cancer
`[22]. However due to target limitation (only targeting
`VEGF-A) as well as relatively poor tissue penetration
`because of its large size, the overall impact of bevacizu-
`mab in prolonging survival was very limited [22,23],
`with 5-year survival generally between 5% and 8% [23],
`suggesting that VEGF-A blockade alone may not be
`good enough to completely prevent tumor angiogenesis
`and corresponding tumor growth.
`Aflibercept (originally called VEGF-Trap) was approved
`in August of 2012 under the trade name of Zaltrap for the
`treatment of metastatic colon cancer, and the same mol-
`ecule was approved in November of 2011 under the trade
`name of Eylea for the treatment of AMD. Aflibercept is a
`recombinant fusion protein consisting of the second im-
`munoglobulin (Ig) domain of VEGFR1 and the third Ig
`domain of VEGFR2, fused to the immunoglobulin con-
`stant region (Fc) portion of human IgGi [24]. Unlike bev-
`acizumab, aflibercept exhibits affinity for all isoforms of
`VEGF and P1GF [25] and exerts robust antivascular ef-
`fects by rapid regression of existing tumor vessels [26],
`normalization of surviving mature vessels [27], and in-
`hibition of new tumor vessel growth [28). The anti-tumor
`efficacy of aflibercept has been confirmed in several solid
`tumor models, all demonstrating effective tumor inhib-
`ition [29]. Aflibercept has a MW of 110 kDa and has a
`half-life in plasma of 4-5 days [24]. The clinical benefits
`for aflibercept treatment of metastatic colon cancer pa-
`tients are similar to bevacizumab [30].
`It has been documented that the VEGF-binding affin-
`ity of VEGFR1 is 10 fold higher than that of VEGFR2
`
`[31] and the second Ig domain of VEGFR1 is critical for
`VEGF binding [32]. We reasoned that a recombinant
`protein composed of only the second domain (D2) of
`VEGFR1 might retain sufficient VEGF binding, but also
`have better bioavailability and penetration properties
`due to its smaller size as compared to the previously de-
`scribed current generation of drugs that block VEGF.
`We therefore designed an expression vector that
`expressed a recombinant protein consisting of the D2
`portion of VEGFR1 fused with the Fc portion of human
`IgGi. This protein was extensively characterized for its
`target-binding affinity, angiogenesis inhibition, and phar-
`macokinetic (PK) profile, as well as for its anti-tumor ef-
`ficacy in several xenograft tumor models.
`
`Methods
`Engineering of recombinant proteins
`HB-002.1 is a recombinant protein consisting of two com-
`ponents: one is the D2 domain of human VEGFR1 (Flt!)
`(P134-T226) plus 5 (S129-R133) and 2 (N227, T228)
`amino acids of upstream and downstream flanking se-
`quence respectively, and the second is the Fc fragment of
`human IgG1. To construct the HB-002.1 expression vec-
`tor, 57 nucleotides encoding the signal peptide of mouse
`IgGi heavy chain were added to the 5' end of VEGFR1-
`D2, a Kozak sequence was added to the 5' end of the
`signal peptide sequence, and cloning sites, Hindu! and
`EcoRl, were added to the 5' and 3' ends of the resulting
`sequence, respectively. This designed D2 expression cassette
`sequence was synthesized (GenScript) and subcloned
`into the Hindlll and EcoRI sites of the pHB-Fc vector
`(Generay, ID: X9913T).
`The recombinant Fltl[2]-Fc protein contains the
`VEGFR1-D2 domain (P134-T226) without the addition
`of flanking region amino acids, plus the Fc fragment of
`human IgGi.
`All recombinant proteins were expressed and purified
`from Chinese hamster ovary (CHO) cells (Cat# CCL-61,
`ATCC). 5 jig of each protein were loaded on 10% SDS-
`PAGE gels under reducing as well as non-reducing con-
`ditions. Gels were stained with 0.3% Coomassie Brilliant
`Blue R-250 and destained with 20% methanol.
`
`Western blotting and digestion of proteins with
`N-glycosidase F
`To validate the identity of the purified protein, Western
`blotting analysis was performed [33]. Briefly, different
`amounts of the purified protein (1, 0.5, 0.25 Vg) were sepa-
`rated by electrophoresis in 4-12% Bis-Tris protein gels, and
`then transferred to a polyvinylidene difluoride membrane.
`The membrane was probed using antibodies specific either
`for Fc fragment (horseradish peroxidase (HRP)-conjugated
`rabbit F(ab')2 anti-human lgG, Fc-fragment specific (Immu-
`noResearch Lab) or HRP*Polyclonal Rabbit Anti-Human
`
`Mylan Exhibit 1121
`Mylan v. Regeneron, IPR2021-00880
`Page 2
`
`

`

`Liu etal. BMC Cancer (2015)15:170
`
`Page 3 of 14
`
`IgG (Fc) (Cat#C030222, Cellway-Lab, Luoyang, China)), or
`for human VEGFR1 (Cat# 10136-RPO2, Sino Biological Inc)
`followed by incubation with secondary antibody (HRP-con-
`jugated Affinipure F(ab')2 Fragment Goat Anti Rabbit
`IgGi, F(ab')2 Fragment Specific (ImmunoResearch Lab)).
`Specific bands were visualized via the ECL kit according to
`the manufacturer's instructions (Amersham).
`To analyze the impact of glycosylation on protein activ-
`ity, HB-002.1 protein (Lot#20130521, 3.62 mg/ml) diluted
`to 0.5 mg/ml in 100 mM of ammonium bicarbonate was
`incubated with N-glycosidase F (Cat#11365193001, Sigma)
`(5 Unit/10 Vg protein) at 37°C for 18 hours. Digested and
`non-digested proteins were analyzed in 12% SDS-PAGE
`under reducing and non-reducing conditions. In parallel,
`the digested protein was also assayed for target binding ac-
`tivity, which was compared to that of the parental protein.
`
`Target-binding assay
`Target binding affinity of HB-002.1 was measured by
`ELISA in Falcon 96-Well ELISA Micro Plates coated
`overnight at room temperature with VEGF ligands or
`PIGF (R&D Systems) in PBS (100 ng per well). Coated
`plates were blocked with 3% dry fat milk in PBS-T buffer
`(PBS containing 0.05% Tween-20) and then 100 p1 of
`serially diluted I-IB-002.1 or bevacizumab (Lot#:N3526,
`Roche) or hlgG-Fc (Cat#:10702-HNAH, Sino Biological
`Inc) (from 5 nM to 0.0024 nM) were transferred into the
`plates. After incubation at room temperature for 1 hour,
`plates were washed S times with PBS-T solution, and
`then incubated with HRP-conjugated Fc-specific anti-
`body (Cat#C030222, Celiway-Lab, Luoyang, China) at
`room temperature for 1 hour. Plates were washed 5 times
`with PBS-T buffer and then developed with 100 p1 of
`HRP-substrate solution for up to 5 minutes. The reaction
`was stopped with 1 N H2SO4, and the absorbance at 450
`nM was determined in a standard plate reader.
`To determine the kinetic target binding affinity of HB-
`002.1, varying amounts of VEGF-A were mixed with 0.5
`nM of HB-002.1, Flt1[2]-Fc, hlgG-Fc or bevacizumab and
`then incubated for 2 hours at room temperature. The mix-
`tures were transferred to VEGF-A coated plates and in-
`cubated for 1 hour at room temperature, the non-bound
`proteins in solution were washed away, and the amounts
`of I-IB-002.1, Flt1 [21-Fc, hlgG-Fc or bevacizumab bound
`to the plates were measured by HRP-conjugated rabbit
`anti-human IgG-Fc antibody. The kinetic binding affinities
`were analyzed according to the amounts of free VEGF
`blocker in the mixtures.
`
`VEGFR2 phosphorylation assay
`4 ml of human umbilical vein endothelial cells (HUVECs)
`(Cat#HUVEC-004, ALLCELLS) in complete HUVEC-
`adapted medium (Cat#H-004, ALLCELLS) were incubated
`in 6 cm dishes at 37°C, 5% CO2 for 24 hours, cells were
`
`starved for 2 hours and then challenged for 15 minutes
`with either medium alone, or VEGF-A (20 ngiml) only, or
`VEGF-A pre-incubated with varying amounts of HB-
`002.1. Cells were washed twice with cold PBS and then
`dissolved in 200 p1 of lysis buffer (50 mM Tris, pH 7.4, 1%
`sodium deoxycholate, 1% Triton X-100, 0.1% SDS, 1 mM
`EDTA, pH 8.0, 150 mM NaCi). After centrifugation and
`quantitation, equal amounts of supernatant from each
`sample were subjected to Western blotting analysis using
`antibodies specific either for total VEGFR2 (Cat# 2479,
`Cell Signaling Technology) or for VEGFR2 phosphotyro-
`sine (Cat# 3770S, Cell Signaling Technology).
`
`VEGF-Induced HUVEC proliferation and tube formation
`assay
`I-UJVEC proliferation in response to VEGF-A and the
`impact of HB-002.1 on cell proliferation was measured
`using CCK-8 kits (Cat# CKO4-11, DOJINDO Laborator-
`ies) following the manufacturer's instructions. Briefly,
`2000 HUVECs per well were plated in a 96-well plate,
`which was incubated at 37°C for 2 hours. 100 1il of re-
`agent solution containing 20 ng/ml of VEGF-A and
`varying amounts of HB-002.1, bevacizumab or hlgG-Fc
`were transferred to the plate. Cells were cultured for
`72 hours at 37°C, and then CCK-8 was added to these
`cultures, which were incubated for 4 additional hours
`followed by spectrophotometric analysis at 450 nm.
`The VEGF-induced tube formation assay was conducted
`as previously described [34]. Briefly, 50 p1 of HUVECs at 3
`x l05lml in culture medium were mixed with 50 0 of cul-
`ture medium containing 20 ng/ml of VEGF-A plus 1000
`nM HB-002.1 protein, bevacizumab or control human
`IgG. The mixtures were added to 96-well plates contain-
`ing 50 p1 of solidified MatrigeL Plates were incubated in a
`cell culture incubator at 37°C for 24 hours. Tube forma-
`tion was observed using an inverted phase contrast micro-
`scope (Eclipse TS100, Nikon). Images were captured with
`a CCD color camera (KP-D2OAU, Hitachi) attached to the
`microscope using 40x magnification plus 1.5x amplifica-
`tion by the CCD camera. The tube length in three differ-
`ent fields was measured using Image-Pro Plus software
`(Version 6.0, Media Cybernetics).
`
`Angiogenesis analysis
`The impact of HB-002.1 on angiogenesis was investigated
`using a transgenic zebrafish embryonic angiogenesis model
`[35]. Briefly, the tested protein or control drugs were
`microinjected into the common cardinal vein of zebraflsh
`at 48 hours post-fertilization (hpf). The subintestinal vessels
`(SIVs) were visualized under a Multi-Purpose Zoom Micro-
`scope (Nikon AZ100), and the area of the SIVs at 72 hpf
`was measured as mean f]uoresence intensity (MFI) using
`NIS-Elements D imaging software. The percentage of
`angiogenesis inhibition was calculated as (MR of vehicle
`
`Mylan Exhibit 1121
`Mylan v. Regeneron, IPR2021-00880
`Page 3
`
`

`

`Liu etal. BMC Cancer (2015)15:170
`
`Page 4 of 14
`
`treated S1Vs - MFI of drug treated SIVs)IMFl of vehicle
`treated SlVs x 100.
`
`Pharmacokinetic analysis
`16 BALBIc mice (female, age of 4-5 weeks, body weight
`of 18-20 g) received a subcutaneous (s.c.) injection of
`50 Vg HB-002.1 protein (-.2.5 mg/kg mouse) and bled at
`1, 2, 4, 6, 24, 48, 72, and 144 hours after injection. Levels
`of J-IB-002.1 in the plasma were measured by ELISA
`assay using human VEGF16S (R&D Systems) as capture
`protein and l-IRP-anti-human Fc (Jackson ImmunoRe-
`search Lab) as the detection antibody.
`
`In vivo efficacy study
`Mouse xenograft tumor models using human Colo-205
`and A549 cancer cells were applied to the investigation
`of the in vivo efficacy of l-(B-002.1. Cells purchased from
`ATCC were resuspended in serum-free medium. BALB/c
`nude mice were ordered from Shanghai SLAC Laboratory
`Animal Co. Ltd. The animals were specific pathogen free
`and approximately 4 - 5 weeks old upon arrival at Phar-
`maLegacy Laboratories. The procedures that were applied
`to animals in this protocol had been approved by Pharma-
`Legacy Laboratories JACUC before the execution of the
`study. Approximately 5 x 10' cells in 200 p1 of serum-free
`medium/matrigel (50:50 v/v) were injected s.c. in the right
`flank of each of the 70 mice for each model under
`anesthesia by 3 - 4% isoflurane. When the average tumor
`volume reached 100 - 200 mm3, 50 mice bearing tumors
`of suitable size were randomized into 5 groups (10 mice
`per group) according to tumor volume and body weight.
`Mice were treated with two different doses (5 mg/kg,
`20 mg/kg) of HB 002.1 or control drugs by intraperitoneal
`(i.p.) injections twice weekly for four weeks except for
`doxorubicin which was given only in one injection. Tumor
`volume and body weight were measured twice a week
`until the termination of the study. Tumor growth inhib-
`ition (TGI%) = (1-(change in mean treated tumor volume/
`change in mean control untreated tumor volume)) x 100.
`Tumor weight measured at time of mice sacrifice.
`
`Histology analysis
`Tumors were harvested and sectioned at the end of the ex-
`periments. Tumor sections were subsequently dewaxed
`and rehydrated. After quenching endogenous peroxidase
`activity, sections were immunohistochemically stained with
`respective antibody. Stained sections were dehydrated in
`alcohol and xylene, and then mounted. The procedure for
`hematoxylin and eosin (H&E) staining of tumor sections
`was as follows: dewaxing in xylene, gradient ethanol dehy-
`dration, hematoxylin staining, rinsing with tap water, coun-
`terstaining with eosin, rinsing with ethanol, gradient
`ethanol dehydration, and vitrification with xylene. Immu-
`nohistochemical staining was performed using antibodies
`
`specific for CD31 (Cat#: ab9498, Abeam) followed by goat
`anti-mouse secondary antibody (Cat#: K1T5002, Fuzhou
`Maixim) and goat anti-rabbit secondary antibody (Cat#:
`K1T5005, Fuzhou Maixim), respectively. The microvessel
`density was quantified by the visual approximation tech-
`nique, which involved manual counting vessels in three
`different microscope fields at IN magnification. The hist-
`ology results were analyzed by a pathologist on a single-
`blind basis. For tumor necrosis evaluation on H&E stained
`slides, homogenous staining in pink or pale color without
`cellular profiles/outline were considered necrotic cells,
`while cellular profiles/outlines with dark blue nuclei were
`considered healthy cells.
`
`Statistics
`Statistical software used for data analysis and presenta-
`tion was SAS 9.3 (SAS Institute), Prism 5 (GraphPad
`Software), and Excel 11 (Microsoft). Binding curves were
`calculated and presented using Prism 5 nonlinear reg-
`ression least squares fit sigmoidal dose-response variable
`slope (also known as four-parameter dose-response)
`curves. Comparisons between different treatment groups
`in HUVEC proliferation was performed using a two-way
`analysis of variance (ANOVA), which included the main
`effects of treatment group and loglO concentration, as
`well as the treatment group x loglO concentration inter-
`action. Upon finding a significant interaction effect, sep-
`arate one-way ANOVA comparisons were carried out at
`each concentration. If a significant difference was found,
`then Tukey's multiple comparisons were used. Compari-
`sons between different treatment groups in tube forma-
`tion by one-way ANOVA provided a F-test with a small
`P value (P = 0.0015) supporting subsequent Tukey's mul-
`tiple comparison test. Comparisons between control (ve-
`hicle-treated) and different treatment groups for inhibition
`of zebrafish angiogenesis were made by Dunnett's multiple
`comparison test. In vivo tumor volumes and weights were
`expressed as mean ± standard error of the mean or geo-
`metric mean with 95% confidence interval. Comparisons
`between different in vivo treatments and control PBS
`treated mice for changes in tumor weights were made by
`Mann-Whitney two-tailed test. For tumor volume, re-
`peated measures (RM) ANOVA with a mixed models ap-
`proach was used to determine if the treatment groups
`behaved differently across time (Le. the "group x time"
`interaction). A logio transformation of tumor volume was
`used to satisfy the required underlying assumptions of this
`statistical model. Since graphical analysis and theoretical
`considerations suggest that tumor volume grows logarith-
`mically, such that its rate of growth decreases over time, a
`log10transformation was applied to day (specifically, log 10of
`Day +1), and included as a linear main effect, as well as in
`the interaction term with group. The model contained one
`repeated "within subjects" factor of time, a "between
`
`Mylan Exhibit 1121
`Mylan v. Regeneron, IPR2021-00880
`Page 4
`
`

`

`Liu of at BMC Cancer (2015) 15:170
`
`Pages of 14
`
`animals' factor of treatment group, and the group x time
`interaction. Both group and time were considered fixed ef-
`fects in each of the RM ANOVA models, as necessarily
`was, the group x time interaction. Upon finding a signifi-
`cant difference, interest only focused on the comparison of
`the treatment groups to control (PBS), but not amongst
`each other. To calculate the statistical significance of treat-
`ments on TGI%, we calculated the ratio of tumor volume
`at Day 35 relative to Day 0 for each mouse, followed by a
`log transformation of this ratio to achieve normality (log
`Day35/Dayo), which is analytically equivalent to looking at
`percent change in tumor volume, but is more suited to
`conventional analysis. ANOVA was then used to compare
`the mean log ratios with the Student-Newman-Keuls test
`to make multiple comparisons. P < 0.05 was considered
`significant. For CD31 staining of tumor sections, only
`group descriptive statistics were calculated. No inferential
`statistical comparisons were performed since the sample
`size was so small (n = 3).
`
`Results
`
`Engineering and production of HB-002.1
`It has been documented that the second Ig-like domain
`(D2) of human VEGFR1 (Fltl[2]) is critical to VEGF
`binding [32], however the purified Fltl[2] fused with Pc
`
`did not bind to VEGF at all, and neither did truncated
`protein containing the first 2 domains (Flt1[1,2]) or that
`containing domains 2 and 3 (rltl[2,3]) [32]. Only protein
`containing domains 1-3 had fill VEGF binding activity
`comparable to that of the whole extracellular portion of
`wild type VEGFR1. This phenomenon was confirmed as
`well by Barleon et al [36), revealing the requirement of
`VEGF binding for the first three Ig-like domains, Based on
`these studies, we designed the HB-002.1 protein in which S
`flanking amino acids (S129-R133) at the N-terminal and 2
`amino acids (N227, T228) at the C-terminal of the D2 do-
`main were included with D2 (Figure 1A). The D2 domain-
`only (Fltl[2]-Fc) was also expressed as a control for VEGF
`binding assay.
`The 1-13-002.1 and Flt1[2)-Fc proteins were produced
`in CHO cells upon transfection with the corresponding
`construct. The secreted proteins were purified and
`resolved in 10% SDS-PAGE gels showing MWs of HB-
`002.1 and Flt1[2]-Fc at -110 kDa in non-reducing
`conditions, and -45 kDa in reducing conditions (Figure 1B),
`both relatively larger than the calculated MW, which
`is most likely due to glycosylation since there are
`two N-linked glycosylation sites in the D2 domain.
`Bevacizumab resolved in the correct MW positions
`(Figure 1B).
`
`A.
`
`Elti (2)-Fc
`
`HBOO2.1
`
`B.
`
`LB
`
`VEGFR1-02
`
`hlgol-Fc
`
`N-Ftank (saa)
`
`OFrank (2aa)
`it
`
`C.
`
`1
`
`D.
`
`NR
`
`P
`
`I 2 3 i,oa4 5 6
`
`rrtgo-Fc
`
`hr gO 'Fc
`
`P
`
`NR
`
`Load (Ito):
`
`0.5 0.25 aDa i 0.5 0.25
`
`htgG-Fc
`
`htgS-Fe
`
`Load (eg)
`
`P 0.2
`
`NF
`I 0.5 5 kDa 1 0.5 0.25
`
`Blotting with Fe-specific Ab
`
`Blotting with VEOFR1-specific Ab
`
`4
`
`I. 4: HB-002.l;
`2, 5: Bevacizumab
`3 6: Flt1(2)-Fc
`
`Ito
`
`=5—
`
`10
`05
`43
`
`34
`25
`
`Figure 1 Engineering and production of HB-002.1. (A) Diagram of HB-002,1 engineered structure, Illustration on top represents Fiti 121-Fc
`consisting of the 02 domain only fused with the Fc portion of human gG1. HB-002.1 contains the 02 domain plus sand 2 amino adds at the
`Sand 3' flanking region respectively. Signal peptide derived from the heavy chain of mouse IgGi ([5) was included for both constructs. (B)
`SOS-PAGE gel analysis. Three proteins were included in the analysis: HB-002.1 (Lane 1,4); Bevacizumab (Lane 2,5); Flt1l2l-Fc (Lane 3, 6). 5 pg of
`each protein were loaded under reducing and non-reducing conditions. (C-D), Western blot analysis. 0.25, 0.5, and 1 pg of HE 002.1 protein and
`1 pg of hlgG-Fc were resolved on 10% SOS-PAGE gels under reducing (RI or non reducing (NB) conditions, transferred to a polinylidene difluoride
`membrane, and probed with Fc-specific (C) or VEGFRI-speciftc (0) antibody (Ab). For comparison, protein MW size markers are shown in kDa.
`
`Mylan Exhibit 1121
`Mylan v. Regeneron, IPR2021-00880
`Page 5
`
`

`

`Mu et at. OMC Cancer (2015)15:170
`
`Pages of 14
`
`To confirm the identity of the proteins, Western blotting
`was performed using antibodies specific for Fc (Figure 1C)
`or VEGFR1 (Figure 1D), showing specific bands for each
`specified portion of the protein at different protein loading
`amounts.
`
`HB-002.l has strong binding affinity to VEGF-A
`HB-002.1 was first analyzed for its binding affinity to
`VEGF-A and compared with that of Fltl[2]-Fc and beva-
`cizumab. The data showed that HB-002.1 had a high af-
`finity with a half maximal effective concentration (ECSO)
`
`of 24 pM, which was 3-fold higher than that of bevacizu-
`mab (EC5O=72 pM) (Figure 2A). As expected, Flt1[2]-Fc
`only had a minimal binding activity to VEGF-A, con-
`firming a binding requirement for the flanking sequence.
`Binding activity of 1-18-002.1 to VEGF-B and PIGF was
`also investigated by ELISA, showing a modest binding to
`VEGF-B (Figure 28) but low binding to PIGF (Figure 2C).
`To determine the target-binding kinetics of HB-002.1,
`equilibrium binding assays were performed in which
`varying amounts of VEGF-A were mixed with 0.5 nM of
`l-18-002.1 or bevacizumab, and the unbound HB-002.1
`
`A.
`
`VEGF-A
`
`-
`
`• FIt1(2)-Fc
`1-15-002.1
`Bevacizurnab
`hlgG-Fc
`
`A
`
`D.
`
`0.6
`
`• 1-16-002.1
`Bevacizurnab
`
`A
`
`3-
`
`0
`
`In n
`0
`0
`
`0
`0.001
`
`0.01
`
`'n- "r rn,.,
`0.1
`1
`10
`conc. nM
`
`0.0
`0.01
`
`0.1
`
`I
`VEGE-165 (nM)
`
`10
`
`100
`
`B.
`
`VEGF-B
`
`E.
`
`Non-reducing
`
`Reducing
`
`• rhVEGFR1-Fc
`• HB-002.1
`hlgG-Fc
`
`11 00
`10
`1
`Protein cone. (nM)
`
`1000
`
`C.
`
`PIGF
`
`0.4
`
`0.31
`
`0.2
`
`0.1 -j
`
`0
`0
`
`• rhvEGFR1-Fc
`• HB-002.1
`htgG-Fe
`
`F.
`
`0
`In
`
`0
`
`D ND
`
`M 7 4:MT
`
`D ND
`
`- w
`
`S
`
`a
`
`-t Digested
`-.- Non-cgested
`-.- hlgG-Fc
`
`01
`
`1000
`
`0.01
`
`0.1
`
`100
`10
`1
`1 10
`Protein conc. (nM)
`Protein conc.(nM)
`Figure 2 Target binding activity of HB-002.1.Target binding activity of intact as well as deglycosylated HB-002.1 was analyzed by ELISA (A) Binding
`to VEGF-A was compared to bevacizurnab and Fft1(2). hlgG-Fc was used as negative control. (8-C) Binding to VEGF-B (8) and PIGF (C) was compared
`with rhVEGFR1-Fc (0) Kinetic binding affinity was measured by equilibrium binding assays that measures unbound HB-t2.1 or bevacizumab after
`incubation of 0.5 nM of HB-002.1 or bevacizumab with varying amounts of VEGF-165. (E) HB-002.1 deglycosylated by treatment with N-glycosidase F
`(D) or not deglycosylated (ND) was separated by SDS-PAGE under reducing or non-reducing conditions and visualized by staining with coomassie
`Brilliant Blue. (F) VEGF-A binding affinity was compared between intact (non-digested) and deglycosylated (digested) HB-oo2.1.
`
`100
`
`iobo
`
`Mylan Exhibit 1121
`Mylan v. Regeneron, IPR2021-00880
`Page 6
`
`

`

`Liu et aL BMC Cancer (2015) 15:170
`
`Page 7 of 14
`
`or bevacizumab was measured by ELISA using VEGF-A
`coated plate, revealing that HB-002.1 displays an equilib-
`rium dissociation constant (KD) of 180 pM, whereas bev-
`acizumab has a KD of 890 pM (Figure 2D).
`Since two different-sized bands were observed both in
`SDS-PAGE gels and in Western blots, we wondered if
`this was due to Winked glycosylation and if this gly-
`cosylation might have an impact on VEGF binding. To
`address these questions, HB-002.1 protein was digested
`with N-glycosidase F and then resolved in 10% SDS-
`PAGE gels, which showed a single band under reducing
`conditions and a smaller size single band under non-
`reducing conditions when compared to that of non-
`digested parental protein (Figure 2E). This confirmed
`our hypothesis that the doublet bands were due to N-
`linked glycosylation. The digested protein retained simi-
`lar VEGF-binding activity to that of parental protein
`(Figure 2F), indicating glycosylation is not essential for
`high affinity binding, which is consistent with the report
`by Barleon et al [36].
`
`HB-002.1 dose-dependently inhibited VEGF-induced
`VEGFR2 phosphorylation, HUVEC proliferation and tube
`formation
`Due to the strong VEGF binding affinity, we anticipated
`that FIB-002.1 must also have strong blocking activity
`against VEGF-induced VEGFR2 phosphorylation as well
`as the resulting cell proliferation and tube formation. As
`shown in Figure 3A, while strong phosphorylation was
`observed with VEGF addition and VEGF plus hlgG, the
`induced phosphorylation was sequentially diminished
`following addition of sequentially increasing amounts of
`HB-002.1, which is comparable to that of bevacizumab
`showing a dose-dependent inhibition of VEGFR2 phos-
`phorylation (Tyr951/1175) in 1-EUVECs [37].
`Comparisons between different treatment groups in
`HUVEC proliferation (Figure 3B) by two-way analysis of
`variance (ANOVA) provided a F-test with a small P value
`(P <0.0004) supporting subsequent evaluation for differ-
`ences among treatment groups. Significant inhibition, as
`compared to hlgG-Fc, in VEGF-induced HUVEC proli-
`feration was observed in a dose-dependent manner for
`I-EB-002.1 (P < 0.05 at all except lowest dose), which was
`comparable to that of bevacizumab (P <0.05 at all doses)
`(Figure 3B). The same phenomenon was also observed for
`VEGF-induced tube formation (Figure 3C, D), for which
`HB-002.1 had a significant and comparable inhibition to
`that of bevacizumab (P <0.05) as compared to hlgG, sug-
`gesting a strong blocking activity of HB-002.1 in VEGF-
`mediated cell biological activity.
`
`HB-002.1 dose-dependently Inhibited in vivo angiogenesis
`Using a transgenic zebrafish embryonic angiogenesis model
`[35], the impact of HB-002.1 on in viva angiogenesis was
`
`investigated and showed a dramatic reduction in number
`of SIVs. While 7-8 SIVs were usually observed in zebrafish
`at 72 hpf (Figure 4A), a decreased number of SIVs was ob-
`served when treated with HB..002.1 (Figure 43). The level
`of inhibition versus vehicle group reached 7.5 (±3.5) %
`(P>0.05), 15.2 (±3.3) % (P<0.01), and 21.4 (±2.4) %
`(P <0.001) for 1-13-002.1 at the doses of 4.4, 14.7, 44 ng. re-
`spectively. Because bevacizumab specifically binds human
`VEGF and its activity against zebrafish VEGF is not known,
`a broad spectrum angiogenesis inhibitor, endostatin,
`known to inhibit angiogenesis in this model [38], was used
`as a positive control showing a 9.7 (±2.8) % and 20.1 (±2.6)
`% inhibition at the dose of 44 and 100 ng respectively
`(Figure 4B). These results suggest a strong angiogenesis
`inhibition activity for HB-002.1.
`
`HB-002.1 has an excellent pharmacokinetic profile
`HB-002.1 has a much smaller molecular mass than
`current VEGF inhibitors, bevacizumab and aflibercept
`(-80 vs. -160 and -110 kDa, respectively), thus it might
`have a shorter half-life and worse PK profile compared
`to these drugs. To address these questions, 25 mg/kg of
`HB-002.1 were injected s.c. into mice (n = 16) and plasma
`taken at different time points post-injection were analyzed
`for 1-13-002.1 levels by ELISA. The results indicated that
`HB-002.1

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