throbber
EXHIBIT ,, 0
`WIT. Ktbnoti
`DATE --4 -
`KRAMM COURTREPOR rING
`
`UC San Diego
`UC San Diego Previously Published Works
`
`Title
`Comparison of binding characteristics and in vitro activities of three inhibitors of vascular
`endothelial growth factor A.
`
`Permalink
`https://escholarship.orci/uc/item/3408m1cv
`
`Journal
`Molecular pharmaceutics, 11(10)
`
`ISSN
`1543-8384
`
`Authors
`Yang, Jihong
`Wang, Xiangdan
`Fuh, Germaine
`et al.
`
`Publication Date
`2014-10-01
`
`DOI
`10.1021/mpsool6ov
`
`Peer reviewed
`
`eScholarshio.orq
`
`Powered by the California Digital Library
`University of California
`
`Mylan Exhibit 1106
`Mylan v. Regeneron, IPR2021-00880
`Page 1
`
`

`

`oarmaceutics
`
`pubs.acs.org/molecu la rpha rmaceutics
`
`Article
`
`Comparison of Binding Characteristics and In Vitro Activities of
`Three Inhibitors of Vascular Endothelial Growth Factor A
`Jihong Yang Xiangdan Wang Germaine Fuh, Lanlan Yu, Eric Wakshull, Mehraban Khosraviani,
`Eric S. Day, Barthélemy Demeule, Jun Liu, Steven J. Shire) Napoleone Ferrara,t and Sandeep Yadav*
`
`Genentech, Inc., 1 DNA Way, MS 56-2A, South San Francisco, California 94080, United States
`
`Ran,b,ziirnab
`
`Atibecctat
`
`U"w (UtUe
`
`Bev,ci,u mat
`
`A
`
`B
`
`C
`
`t
`
`ABSTRACT: The objectives of this study were to evaluate the relative binding and potencies of three inhibitors of vascular
`endothelial growth factor A (VEGF), used to treat neovascular age-related macular degeneration, and assess their relevance in the
`context of clinical outcome. Ranibizumab is a 48 kDa antigen binding fragment, which lacks a fragment crystallizable (Fc) region
`and is rapidly cleared from systemic circulation. Aflibercept, a 110 kDa fusion protein, and bevacizumab, a 150 kDa monoclonal
`antibody, each contain an Fc region. Binding affinities were determined using Biacore analysis. Competitive binding by
`sedimentation velocity analytical ultracentrifugation (SV-AUC) was used to support the binding affinities determined by Biacore
`of ranibizumab and aflibercept to VEGF. A bovine retinal microvascular endothelial cell (BREC) proliferation assay was used to
`measure potency. Biacore measurements were format dependent, especially for aflibercept, suggesting that biologically relevant,
`true affinities of recombinant VEGF (rhVEGF) and its inhibitors are yet to be determined. Despite this assay format dependency,
`ranibizumab appeared to be a very tight VEGF binder in all three formats. The results are also very comparable to those reported
`previously.` At equivalent molar ratios, ranibizumab was able to displace aflibercept from preformed aflibercept/VEGF
`complexes in solution as assessed by SV-AUC, whereas aflibercept was not able to significantly displace ranibizumab from
`preformed ranibizumab/VEGF complexes. Ranibizumab, aflibercept, and bevacizumab showed dose-dependent inhibition of
`BREC proliferation induced by 6 ng/mL VEGF, with average IC,,, values of 0.088 ± 0.032, 0.090 ± 0.009, and 0.500 ± 0.091
`nM, respectively. Similar results were obtained with 3 nglmL VEGF. In summary Biacore studies and SV-AUC solution studies
`show that aflibercept does not bind with higher affinity than ranibizumab to VEGF as recently reported,' and both inhibitors
`appeared to be equipotent with respect to their ability to inhibit VEGF function.
`KEYWORDS: rauibizuniab, aflibercept, bevacizu,nab, VEGF, affinity, analytical ultracentnfugation
`
`• INTRODUCTION
`
`important variables that govern the determination of affinity
`
`The determination of binding affinity of a therapeutic protein
`to a target is an integral part of pharmaceutical development. A
`widely used methodology for assessing tight interactions is
`based on surface plasmon resonance (SPR) such as Biacore.5
`One caveat in this technology is that it requires ligand
`immobilization to a surface and it has been shown that
`orientation, method of binding, and which ligand is bound are
`
`constants. 6,7
`
`Received: February 25, 2014
`Revised:
`August 22, 2014
`Accepted: August 27, 2014
`Published: August 27, 2014
`
`'rACS Publications
`
`02014 American Chemical Society
`
`3421
`
`dadolorg/10.1021/nipsooltoy I Moi. plwrmoceatic, 2014. 11.3421-3430
`
`Mylan Exhibit 1106
`Mylan v. Regeneron, IPR2021-00880
`Page 2
`
`

`

`Molecular Pharmaceutics
`
`Alternatively, analytical ultracentrifugation (AUC) can be
`used as an orthogonal free-solution technique that circumvents
`the potential artifacts of matrix/stationary phase or chemical
`modifications associated with SPR. AUC has been widely used
`in the biophysical characterization of proteins to determine
`weight-average molecular mass, sedimentation coefficient,
`frictional coefficient associated with molecular shape, bimo-
`lecular interactions involving reversible associations or complex
`formation, self-association of glycosylated and nonglycosylated
`proteins, and competitive binding of anti-IgE antibodies to IgE
`and also as an orthogonal technique to size exclusion
`chromatography to determine the presence of aggregates.' In
`addition to being able to detect the presence of protein
`aggregates, AUG analysis allows measurements directly in the
`formulation buffer or condition of interest, thereby avoiding
`common size exclusion HPLC limitations of protein-resin
`interactions and significant dilution in the elution buffer that
`can potentially alter the size distribution of the self-associates
`and aggregates, as highlighted in the above studies. One aspect
`of this work is to assess AUC as an orthogonal technique to
`SPR in evaluating the binding of therapeutic proteins,
`highlighting that caution must be exercised while relying on
`SPR results. In addition, the recent development of
`fluorescence optics in the analytical ultracentrifuge9'° com-
`bined with the use of fluorescently labeled material can provide
`definitive information about the type of complex formed.
`There have been several SPR studies and potency assess-
`ments of inhibitors of vascular endothelial growth factor A
`(VEGF),2'-'4"' 2 a key driver of the vascular leakage and
`neovascularization seen in intraocular vascular diseases
`including age-related macular degeneration (wet AMD), retinal
`vein occlusion (RVO), and diabetic macular edema (DME).'
`VEGF inhibitors such as ranibizumab, aflibercept, and
`bevacizumab are used intravitreally in patients with wet
`AMD, RVO, and DME. The US Food and Drug Admin-
`istration has approved ranibizumab for the treatment of wet
`AMD, RVO, and DME 13 and allibercept for the treatment of
`wet AMD and central RVO. 14 lievacizumab is used off-label. A
`recent SPR study concludes that aflibercept binds to VEGF
`with much higher affinity than ranibizumab.' Herein we report
`the results of the determination of affinity constants for binding
`of VEGF to ranibizumab, affibercept, and bevacizumab by SPR
`using different assay formats as well as the potency of inhibition
`of VEGF. In addition, a novel solution based competitive
`analytical ultracentrifuge method is used to support our
`conclusions.
`
`• MATERIALS AND METHODS
`
`Lucentis (ranibizumab) and Avastin (bevacizumab) were
`obtained from Genentech, Inc., South San Francisco, CA.
`Eylea (aflibercept; Regeneron, Inc., Tarrytown, NY) Was
`obtained commercially. Recombinant human VEGF 165
`(rhVEGF) was expressed and purified at Genentech. Anti-Fab
`antibody was obtained from GE Healthcare (Pittsburgh, PA),
`and protein A was obtained from Thermo Fisher Scientific,
`Pierce Protein Biology Products (Rockford, IL).
`SPR Binding Assays. Binding kinetics and affinities of
`inhibitors of rhVEGF were assessed using surface plasmon
`resonance technology on a Biacore T200 instrument (GE
`Healthcare, Pittsburgh, PA). A series of analyte concentrations
`were prepared in HES-EP running buffer (0.01 M HEPES, 0.15
`lvi NaCl, 3 mM EDTA, and 0.05% surfactant Polysorbate 20)
`and injected at a flow rate of 50 pL/min for 3 min over flow
`
`Article
`
`cells (FCs) of Series S CMS sensor chips immobilized with
`ligand molecules at various densities depending on assay
`formats.
`In format 1, rhVEGF was the ligand immobilized directly
`onto FCs at --'20 resonance units (RU) density, while the
`inhibitors were the analytes. The dissociation of inhibitors from
`the immobilized rhVEGF was allowed to proceed for S min for
`all samples except for the ranibizumab and bevacizumab
`samples with the highest concentration (200 nM), in which
`dissociation proceeded for 3 h or is mm, respectively. All
`experiments were carried out at 37 °C.
`In format 2, the inhibitors were the immobilized ligands and
`rhVEGF was the analyte in the mobile phase. The final ligand
`density was 22-45 RU. The dissociation of the analytes from
`the immobilized ligand was allowed to proceed for S min for all
`samples except for the ranibizumab sample with the highest
`concentration (200 nM), in which dissociation proceeded for 3
`h. The dissociation time for all bevacizumab samples was 20
`mm. All experiments were carried out at 37 °C.
`In format 3, the inhibitors were immobilized indirectly to the
`sensor chip using anti-human lgG Fab antibody or protein A as
`capturing molecules as previously reported.' Rigorous surface
`testing was conducted in the current study to evaluate the
`validity of the method for all inhibitors. The densities of the
`capture molecules were -11000 or 1000 RU for anti-human
`1gG Fab antibody and -S500 RU for protein A. The final
`ligand density used was -'28 RU for the indirectly captured
`ranibizumab, -'40 RU for bevacizumab, and -'50 RU for
`aflibercept. The dissociation of the analytes from the
`immobilized ligand was allowed to proceed for S min for
`ranibizumab samples except for the sample with the highest
`concentration (200 nM), in which dissociation proceeded for 3
`h. The dissociation time for all aflibercept and bevacizumab
`samples was 30 mm. Experiments were carried out at 37 °C or
`25 °C for this format.
`Sensorgrams of ranibizumab, allibercept, and bevacizunsab
`binding to rhVEGF using all three formats were analyzed to
`obtain kinetic data and affinities using Biacore T200 Evaluation
`Software (version 2.0.1; GE Healthcare). Because of the
`dimeric nature of rhVEGF and the presence of two potential
`binding sites in all inhibitors except ranibizumab, definitive
`monovalent binding affinities for rhVEGF and its inhibitors can
`be challenging to obtain. Very low immobilization densities
`were used to encourage monovalent binding, and the presence
`of such interactions were evaluated using a 1:1 Langmuir
`binding model. In all but two conditions tested, the 1:1 binding
`model was sufficient to describe interactions between rhVEGF
`and its inhibitors. The dissociation rate constant (kd) and
`association rate constant (Ic,) were obtained via kinetic fitting,
`and the equilibrium dissociation constant (K,,) was derived by
`taking the ratio of kd over k. calculated using the simplest 1:1
`binding model. Only in cases where allibercept was evaluated in
`formats I and 2 was the 1:1 binding model insufficient to
`describe interactions between the inhibitor and rhVEGF. In
`those cases a bivalent analyte binding model was used and the
`first equilibrium dissociation constant (K,,,), first dissociation
`rate constant (kdj, and first association rate constant (Ic,,) were
`reported.
`Competitive Binding Assessed by Sedimentation
`Velocity Analytical Ultracentrifugation (SV-AUC). Each
`molecule individually and preformed complexes between
`ranibizumab and VEGF and allibercept and VEGF were first
`evaluated to obtain their sedimentation coefficients. After this,
`
`3422
`
`d,.doI.o,g10102i/n,p500160v I Mel. Pharmoc.utirs 2014, 1,3421-3430
`
`Mylan Exhibit 1106
`Mylan v. Regeneron, IPR2021-00880
`Page 3
`
`

`

`Molecular Pharmaceutics
`
`Ranibizumab
`La
`La
`ts
`pM)
`67
`
`0.19
`
`0.67
`
`\\.
`
`tI
`
`
`
`04
`
`Aflibercept
`kal
`Ml
`(IC' 1/MO) )I1 I!')
`3.30
`1)0.1
`
`6,6
`)pM)
`0163
`
`Article
`
`Bevacizumab
`Si
`64
`60
`30' lAos)
`lu'
`two
`0,14
`219
`4410
`'.3
`
`A
`
`B
`
`C
`
`La
`110' I/Mt)
`
`64
`510' 1/')
`
`Op
`two
`
`64'
`La!
`210' I/Mi) 510' IfS)
`U4
`6906
`
`6<1
`(pM)
`6744
`
`La
`fiG' 1.44,)
`31.1
`
`Olt
`
`LaO
`
`rn' V.)
`
`32.9
`
`K,
`5pM)
`159
`
`La
`
`60
`
`La
`
`7.7
`
`La
`La
`510' l/MOS 110 - 1A)
`213
`203
`
`An114, ii 111 & L) Ab
`
`'
`
`rro, on A
`
`Figure 1. Binding of rhVEGF—anti-VEGF inhibitor molecules in Biacore assays. (A) rhVEGF is the ligand immobilized directly onto the flow cell
`(PC) while the inhibitors were the analytes injected over the PC at varying concentrations (format I). (B) VEGF inhibitors immobilized as ligand
`with VEGF in the mobile phase as analyse (format 2). (C) Inhibitors immobilized indirectly to the sensor chip with VEGF in the mobile phase as
`analyse (format 3). Note: limit of !Cj that can be accurately measured by the instrument is —S X io s'. To be conservative, a k4 < io s— was
`chosen.
`
`competition experiments were conducted using a preformed
`inhibitor/VEGF complex challenged with a different VEGF
`inhibitor to assess whether the previously reported —100-fold
`higher affinity of aflibercept to rhVEGF compared with
`ranibizumab4 is valid in free solution, i.e., no binding to a
`surface as in SPR measurements.
`Experiments were performed at room temperature in PBS,
`PH 7.2 (137 mM NaCl, 27 mM KCl, 8 mM Na1HPO 4, and 1.5
`mM KH 2PO4). Alexa Fluor 488 protein labeling kits were
`purchased from Molecular Probes (Eugene, OR). All chemicals
`used were reagent grade or higher. Alexa Fluor 488 labeled
`ranibizumab (denoted as ranibizumab*) was produced as
`recommended by the manufacturer.
`Sedimentation velocity experiments were performed in an
`Optima XL-I analytical ultracentrifuge equipped with absorb-
`ance optics, interference optics (Beckman Coulter, Fullerton,
`CA), and fluorescence optics (Aviv Biomedical) in centrifuge
`cells with 12 mm graphite-filled Epon centerpieces (Spin
`Analytical, Durham, NH) at 20 °C and rotor speed of 40000
`rpm. Quartz windows were used when using the absorbance
`optics, and the scans were acquired at a wavelength of 230 nm
`at 30 pm radial increments. When using the fluorescence optics,
`sapphire windows were used, and the data were acquired at 20
`pm radial increments averaging five revolutions per scan. The
`sedimentation boundaries were analyzed with SEDFIT, version
`11.3 and 11.72c.' 5 The resulting continuous, c(s), distribution
`with 70% confidence level was calculated after optimizing
`baseline, meniscus, and cell bottom positions by nonlinear
`regression. All s values obtained with the c(s) distribution in
`
`PBS were converted to s2p. with SEDNTERP (version 1.09)
`using the measured density and viscosity of PBS.
`Bovine Retinal Microvascular Endothelial Cell (BREC)
`Proliferation Assay. A BREC assay was used because bovine
`microvascular endothelial cells are well established as a cell type
`that is highly responsive to growth factors such as VEGF and
`bFGF.' 6 Unlike another cell line that has been used to assess
`potency, HUVEC, which is derived from a large vessel, BREC is
`a more physiologically relevant cell type to investigate
`angiogenesis.
`BREC proliferation assays were performed as previously
`described. 2 Cells were seeded in 96-well plates in low glucose
`DMEM (supplemented with 10% heat-inactivated calf serum, 2
`mM glutamine, and antibiotics) at a density of 500 cells/well.
`Ranibizumab and allibercept were tested from 0.004 to 10 nM,
`while bevacizumab was tested from 0.04 to 90 nM. Twenty
`minutes after addition of inhibitors, VEGF was added to a final
`concentration of 6 ng/mL (o.is nM) or 3 nglmL (0.075 nM).
`After 6 days, cell growth was assayed with the use of alamarBlue
`(BioSource). Fluorescence was measured at 530 nm excitation
`wavelength and 590 nm emission wavelength. 1C values were
`calculated using KaleidaGraph. For statistical analysis, one-way
`ANOVA was used, followed by the Tukey—Kramer HSD test
`comparing all pairs.
`
`• RESULTS
`Binding Affinities and Kinetics of VEGF Inhibitors
`Using SPR Technology. Format 1. rhVEGF, an antiparallel
`homodimer, was the immobilized ligand with inhibitors in the
`mobile phase as analyses (Figure IA). Binding of ranibizumab
`
`3423
`
`ddrA.orp/l0.10ll/rrp50Ol60, I Mat Pho,lnoreel)cs 2014,11,3421-3430
`
`Mylan Exhibit 1106
`Mylan v. Regeneron, IPR2021-00880
`Page 4
`
`

`

`Molecular Pharmaceutics
`
`Article
`
`followed a simple monovalent (1:1) analyte binding model as
`expected because the Fab molecule has only one VEGF binding
`site. This was clearly shown by the closeness of the fit to the
`experimental data (Figure IA). Allibercept and bevacizumab
`each contained two VEGF-binding sites. Using very low
`rhVEGF immobilization levels, binding of these two inhibitors
`to rhVEGF was encouraged to favor monovalent binding. 17,18
`This approach worked for bevacizumab since the 1:1 binding
`model sufficiently described the interactions between the
`inhibitor and rhVEGF (Figure 1A). However, in the case of
`aflibercept, even the lowest immobilization level of rhVEGF
`was insufficient to completely shift the interaction to
`monovalent binding, and attempts to fit the data with a 1:1
`Langmuir binding model failed (data not shown). Therefore, a
`bivalent analyte binding model was used considering each
`aflibercept molecule has two potential VEGF binding sites.
`Although the curve fits still deviated from the experimentally
`obtained results (Figure IA), the overall quality of the fit was
`much improved over that obtained from using a 1:1 binding
`model (data not shown). The challenge in fitting the aflibercept
`binding curves may be due to the global fit bivalent model that
`allows individual bulk effect correction to accommodate
`baseline drift,' 8 although other factors such as binding induced
`conformational change cannot be ruled out. Because the second
`step in the bivalent analyte binding model involves intra-
`molecular binding on a sensor chip without an increase in mass,
`only two-dimensional kinetics for the second step are obtained.
`The first step kinetics from a bivalent analyte binding model are
`most relevant in understanding the binding kinetics and
`- strength between an analyte and a ligand. Therefore, only
`first kinetic parameters (k46, k3,) and first KD, were shown for
`aflibercept binding to rhVEGF. Although the (first) association
`rate constants for all three inhibitors were similar, ranibizumab
`had a much slower dissociation rate constant (039 X la_s j'1)
`than aflibercept and bevacizumab (zso.z x io- and 21.9 )<
`lO' s', respectively). As a result, ranibizumab showed a lower
`value (67 pM) than aflibercept (9263 pM) or bevacizumab
`(4456 pM) (Figure IA, insets).
`Format 2. The inhibitors were the immobilized ligands with
`rhVEGF in mobile phase as the analyte (Figure IB). Since each
`rhVEGF has two potential binding sites for the immobilized
`inhibitor molecules, experimental conditions were again
`optimized to encourage monovalent interactions by using low
`ligand immobilization levels. Similar to format 1, the fits using
`the 1:1 binding model for both ranibizumab and bevacizumab
`showed reasonable agreement to the experimentally obtained
`results (Figure IB); for aflibercept, much discrepancy was once
`again observed between the experimental data and the fitted
`curves using the 1:1 binding model (data not shown).
`Therefore, the bivalent analyte binding model was used, and
`first kinetic parameters and dissociation equilibrium constant
`were summarized in Figure 1B, inset. Ranibizumab again
`dissociated much more slowly than the other two inhibitors,
`and a very conservative limit (i x i0 5 s') of it3 was used in
`order to confidently assess the upper limit of K0 value. Even
`with this conservative approach, ranibizumab showed a higher
`binding affinity than aflibercept and bevacizumab, with K0
`(K0,) values of <9.2, 4744, and 159 pM, respectively (Figure
`IB, insets). Comparing results obtained from format 2 and
`format I revealed some interesting observations: the (first)
`association rate constants it, for all three inhibitors were higher
`using format 2 than format I. While it is challenging to know
`exactly how different the it3 values were for ranibizumab, both
`
`formats showed very slow dissociation. The it3 (it31 ) values
`obtained from both formats were very similar for bevacizumab
`but not for aflibercept (Figure IA,B, insets).
`Format 3. The inhibitors were immobilized indirectly to the
`sensor chip using anti-human 1gG Fab antibody or protein A as
`capturing molecules following surface testing to evaluate the
`validity of the method for all VEGF inhibitors (Figure IC).
`While the levels of captured aflibercept and bevacizumab (not
`shown) stayed almost the same during the time needed for
`kinetic analysis (Figure 2A), a significant amount of
`
`520
`.2
`
`B
`
`rime, seconds
`
`1
`
`2000
`
`14.
`
`Time, seconds
`
`2
`
`4305
`
`5300
`
`0000
`
`I 2008
`
`12000
`
`14M
`
`Time, seconds
`
`Figure 2. Levels of aflibercept and ranibizumab captured by an anti-
`Fab antibody or protein A. (A) Level of aflibercept captured by protein
`A over 3 h in an indirect capturing format at 37°C. Level of protein A
`immobilized was approximately 5500 RU. Alhibercept was indirectly
`captured at approximately 50 RU. (B) Level of ranibizumab captured
`by anti-Fab antibody over 3 Ii in an indirect capturing format at 37 °C.
`Level of anti-Fab antibody immobilized on the sensor chip was
`approximately 11000 RU. Ranibizumab was indirectly captured at
`approximately 28 RU. (C) Level of ranibizumab captured by the anti-
`Fab antibody over 3 h in an indirect capturing format at 25 °C. Level
`of anti-Fab antibody immobilized on the sensor chip was
`approximately 1000 RU. Ranibizumab was indirectly captured at
`approximately 27 RU.
`
`ranibizumab dissociated from the anti-Fab antibody capture
`molecule: signal decreased nearly 100% when anti-Fab antibody
`was immobilized at 11000 RU at 37 °C (Figure 2B) and more
`than 5096 when anti-Fab antibody was immobilized at 1000 RU
`at 25 °C (Figure 2C), a condition reported in the literature.'
`These results indicated that this anti-Fab antibody is not
`suitable to capture ranibizumab for affinity measurements. We
`
`3424
`
`do.do&ocf30A011/mp500l60vI Md. Phom,oceol,c, 2014, Ii, 0411-3430
`
`Mylan Exhibit 1106
`Mylan v. Regeneron, IPR2021-00880
`Page 5
`
`

`

`Molecular Pharmaceutics
`
`Article
`
`3.7
`
`5.2
`
`(I
`Ii
`II
`II
`II
`II
`
`3.0,
`
`6.5
`
`A
`
`3
`
`2
`
`1
`
`0
`00 1.0 2.0 3.0 4.0 5.0 6.0 7.0 8.0 9.0 10.0 11.0 12.0
`Sedimentation coefficient (5,,j
`
`40 0
`
`4008
`
`6.1
`
`C
`
`3500
`
`3000
`
`2500
`
`Zy 2000
`
`1500
`
`1000
`
`500
`
`0.0
`
`2.0
`
`4.0
`
`6.0
`
`3.7
`
`0 0 1.0 2.0 3.0 4.0 5.0 6.0 7.0 8.0 9.0 10.0 ILO 12.0
`Sedimentation Coefficient (S,.)
`
`1
`
`5.7
`
`55 N
`
`j
`
`5.8
`
`D
`
`3.4
`4
`
`.
`
`.
`
`• 0
`0
`001.02.0304.05.06.0708.09,010.011.012.0 00 1.0 2.0 3.0 4.0 5.0 6.0 7.0 8.0 9.0 10.0 11.0 12.0
`Sedimentation Coefficient (S)
`Sedimentation Coefficient (S,)
`
`.4.
`
`Figure 3. Analytical ultracentrifugation analysis. (A) Continuous sedimentation coefficient distribution of VEGF (solid black trace), ranibizumab
`(dashed black trace), aflibercept (green trace), 2:1 ranibizumablVtcF complex (red trace), and 1:1 aflibercept/VEGF complex (blue trace) in PBS
`at a total protein concentration of 0.1 mg/mL. The 2:1 ranibizumab/VEGF complex and 1:1 aflibercept/VEGF complex were prepared on a molar
`basis. Ranibizumab, VEGF, and aflibercept were 99% monomeric and did not reveal the presence of aggregate missed by SE-HPLC. Aflibercept
`showed a small amount of higher order species around 7.1 5, which appears to be present in 1:1 aflibercept/VEGF complex, as well at 8.7 S. The
`higher order species were present at a low enough concentration to be considered insignificant. (B) Competition experiment: the sedimentation
`profile of a solution containing 1 mol equiv of ranibizumab (dotted line) or 2 mol equiv of ranibizumab (solid line) added to preformed 1:1
`aflibercept/VEGF complex. The addition of ranibizumab to the aflibercept/VEGF complex displaced aflibercept, which appears as a monomer at 5.3
`S. (C) The continuous sedimentation coefficient distribution for labeled ranibizumab* (inset) and 2 mol equiv of ranibizumab* (labeled) added to
`preformed 1:1 aflibercept/VEGF complex in PBS measured using the fluorescence detection system (FDS) optics. The total concentration of labeled
`ranibi zumab* was maintained at 200 nM. The preformed l:t aflibercept/VEGF complex in PBS was challenged with 2 mol equiv of labeled
`ranibizumab*. The sedimentation coefficient distribution clearly shows the formation of a ranibizumab/VEGF complex. (D) The sedimentation
`profile of various complexes where a preformed 2:1 ranibizumab/VEGF complex was titrated with different amounts of aflibercept in PBS. The total
`protein concentration was maintained at 0.1 mg/mt. A preformed 2:1 ranibizumab/VEGF complex was challenged with 0.5 mol equiv (red trace), 1
`mol equiv (blue trace), and 4 mol equiv (black trace) of aflibercept. Aflibercept was not able to materially displace ranibizumab from the
`ranibizuanab/VEGF complex.
`
`were able to obtain a stable level of captured ranibizumab by
`using an antibody specific to hulgG heavy and light chain
`(Jackson ImmunoResearch Laboratories, Inc., West Grove,
`PA). The results for ranibizumab were similar to those obtained
`in format 2, with an equilibrium dissociation constant of less
`than 13.1 pM using a conservative estimation of the k value
`(Figure IC, inset). In contrast, the results for aflibercept in this
`assay format were drastically different from the ones obtained
`in the first two assay formats: first of all, a simple 1:1 binding
`model was sufficient to describe the interactions between
`aflibercept and rhVEGF using format 3; in addition, the Ic, value
`was over 500- and 10-fold higher than k, 1s obtained from
`formats I and 2, respectively; conversely, the kd value was about
`10- and 200-fold smaller than the kd s's derived from formats I
`and 2, respectively. These kinetic differences resulted in K0
`
`(K01 ) values for aflibercept ranging from 1.83 to 9263 pM
`(Figure I, insets). The K0 of 1.83 pM for aflibercept is similar
`to what was reported by Papadopoulos et al .'4 although the
`experiment reported here was conducted at 37 °C rather than
`25 °C. The association rate constant for aflibercept as
`determined in this format, 1.63 X 108 M' s, is extremely
`fast and may have exceeded the instrument's limit for reliable
`measurement. The results for bevacizumab were similar to the
`ones in format 2 with similar Ic,, Ic4, and IC0 values, obtained
`using a 1:1 binding model (Figures IB,C, insets). The K0 for
`bevacizumab was 7&4 pM as determined using format 3.
`Comparing kinetics obtained from format 3, it is interesting to
`note that both ranibizumab and aflibercept have very high
`affinities in their binding to VEGF, although the main drivers
`
`3425
`
`dx dea.orgllo.loal/rnpstoleov I Moe Phe,mcceurica 2014, 11. a421-a4a0
`
`Mylan Exhibit 1106
`Mylan v. Regeneron, IPR2021-00880
`Page 6
`
`

`

`Molecular Pharmaceutics
`
`Article
`
`for those tight interactions are quite different since ranibizumab
`dissociated very slowly, while aflibercept associated very rapidly.
`Evaluation of Ranibizumab and Aflibercept Binding
`to VEGF Using Sedimentation Velocity Analytical Ultra-
`centrifugation (SV-AUC). Each molecule and preformed
`ranibizumab/VEGF and aflibercept/VEGF complex was first
`evaluated individually to obtain its sedimentation coefficient.
`The analytical ultracentrifugation data was fitted by the direct
`boundary model, c(s), maximizing the resolution and
`sensitivity. 1"9'2° The sedimentation coefficients of VEGF,
`ranibizumab, and aflibercept in PBS, corrected for the standard
`conditions of water at 20 °C
`were 3.0, 3.7, and 5.2 5,
`respectively (Figure 3A). The sedimentation coefficients of the
`2:1 ranibizumab/VEGF complex and 1:1 aflibercept/VEGF
`complex were 6.0 and 6.5 5, respectively (Figure 3A).
`A competition assay in which a preformed 1:1 complex of
`aflibercept/VEGF was challenged with 1 mot equiv of free
`ranibizumab (Figure 3B, dotted line) showed a major peak at
`6.2 5, a peak at 3.6 5, and a raised baseline between 4 and 6 S.
`The peak at 3.6 S is consistent with free ranibizumab (Figure
`3A). The main peak at 6.25 likely represents a mixture of 2:1
`complex of ranibizumabiVhGF (6.0 5) and 1:1 complex of
`aflibercept/VEGF (6.5 5). The raised baseline indicates that the
`mixture is complex and may also contain free aflibercept.
`Interestingly, the addition of 2 mot equiv of free ranibizumab to
`a 1:1 aflibercept/VEGF complex showed a more symmetrical
`peak at 6.2 S with a distinct shoulder at 5.3 S and a peak at 3.6 S
`(Figure 3B, solid line). The shoulder at 5.3 S is consistent with
`free aflibercept and that at 3.6 S with free ranibizumab (Figure
`3A), indicating that ranibizumab was able to displace aflibercept
`from the preformed aflibercept/VEGF complex.
`To verify that the peak at 6.2 5 contains significant levels of
`ranibizumab/VEGF complex, ranibizumab was labeled with the
`Alexa Fluor 488 dye (ranibizumab*) and the competition
`experiment was performed with detection using fluorescence
`optics (Figure 3C). The sedimentation coefficient
`= 3.7
`5) for ranibizumab* alone (inset, Figure 3C) confirmed that
`the labeling procedure did not change the sedimentation
`characteristics of ranibizumab. Additionally, the potency was
`also unaffected (data not shown). The addition of 2 mot equiv
`of ranibizumab to the preformed 1:1 aflibercept/VEGF
`complex clearly showed a ranibizumab*/VEGF complex peak
`at 6.1 S in addition to the free ranibizumab* peak at 3.75. Note
`that with fluorescence optics, only the fluorescently labeled
`species (i.e., ranibizumab* and ranibizumab* complex) are
`visible, and any free aflibercept or aflibercept associated
`complex will not be detected.
`The reverse competition experiment was performed where a
`preformed 2:1 ranibizumab/VEGF complex was challenged
`with 0.5, 1, and 4 mot equiv of free aflibercept (Figure 3D). In
`this case, no labeling was used and the detection was achieved
`with UV optics. The minor peak at 3.4 S is likely a small
`amount of free ranibizumab. The free ranibizuniab peak at 3.4 S
`remained constant with increasing amount of free aflibercept
`added to the preformed complex, indicating no large
`displacement of ranibizumab by aflibercept, even at a 4-fold
`molar excess of aflibercept. The slight difference observed in
`the sedimentation coefficient of free ranibizumab (3.4 vs 3.7 5
`from Figure 3A) is likely due to the known limits of c(s)
`analysis to accurately resolve minor peaks.2' With increasing
`molar concentrations of free aflibercept, the major peak shifted
`to a smaller sedimentation coefficient, that is, from 5.8 to 5.7 to
`5.6 S at 0.5, I, and 4 mot equiv of free aflibercept, respectively,
`
`indicating that the peak may consist of a mixture of 2:1
`ranibizumab/VEGF complex and increasing levels of free
`aflibercept that are not well resolved. The following mass
`balance equations predict this behavior:
`
`s20,(obs) = [A1o,w(A)] + 1p(RV)s20,11(RV)1
`
`A) + (av) = 1
`
`(A) = C(A)/ C, 0031
`
`(I)
`
`(2)
`
`(3)
`
`where F is the weight fraction, C is the concentration in mg/
`mL, and the subscripts (A) and (RV) indicate free aflibercept
`and ranibizumab/VEGF complex) respectively.
`Equation 1 assumes that the observed sedimentation
`coefficient is composed of the weight fraction of free aflibercept
`and ranibizumab/VEGF complex cosedimenting. Equation 2
`represents the assumption that free aflibercept and ranibizu-
`mab/VEGF complex account for all sedimenting species.
`Equation 3 defines the weight fraction of free aflibercept as
`the concentration of aflibercept added to the total protein
`concentration present.
`With C,001 = 0.1 mglmL and the added concentration of
`aflibercept, (A) and F( Rv) can be calculated from eqs 2 and 3.
`With the measured sedimentation coefficients from Figure 3k
`520,w(A) = 5.2 and s2, (,) = 6.0, th e obse rved sedimentation
`coefficient [s2 (obs)] can be calculated from eq I and
`compared with the experimentally determined sedimentation
`coefficients (Table i).
`
`Table 1
`
`co)
`0.030
`O.O4
`0.770
`
`' (caic)
`
`5.76
`5.63
`5.38
`
`s,,.. (obs)
`5.8
`5.7
`5.5
`
`The calculated sw,, values are in remarkably good agreement
`with the experimentally observed 520,w' corroborating the notion
`that the observed shift in the s-value of the major peak in Figure
`3D is likely a consequence of the main peak consisting of
`unresolved 2:1 ranibizumab/VEGF complex and free afliber-
`cept and that increasing amounts of free aflibercept added to
`the preformed ranibizumab/VEGF complex does not signifi-
`cantly disrupt this comp

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