throbber
{CANCER RESEARCH57, 4593-4599, October 15, 1997]
`
`Humanization of an Anti-Vascular Endothelial Growth Factor Monoclonal
`
`Antibody for the Therapy of Solid Tumors and Other Disorders
`
`Leonard G.Presta, Helen Chen, Shane J. O’Connor, Vanessa Chisholm, Y. Gloria Meng, Lynne Krummen,
`Marjorie Winkler, and Napoleone Ferrara’
`Departments of Immunology, Process Sciences, Molecular Biology, Bioanalytical Technology and Cardiovascular Research, Genentech,
`94080
`
`Inc., South San Francisco, California
`
`ABSTRACT
`
`Vascular endothelial growth factor (VEGF) is a major mediator of
`angiogenesis associated with tumors and other pathological conditions,
`including proliferative diabetic retinopathy and age-related macular
`degeneration. The murine anti-human VEGF monoclonal antibody
`(muMAb VEGF) A.4.6.1 has been shown to potently suppress angio-
`genesis and growth in a variety of human tumorcells lines transplanted
`in nude mice and also to inhibit neovascularization in a primate model
`of ischemic retinal disease. In this report, we describe the humaniza-
`tion of muMAbVEGFA.4.6.1. by site-directed mutagenesis of a human
`framework. Not only the residues involved in the six complementarity-
`determiningregions but also several framework residues were changed
`from humanto murine. Humanized anti-VEGF F(ab) and IgG1 vari-
`ants bind VEGF with affinity very similar to that of the original
`murine antibody. Furthermore, recombinant humanized MAb VEGF
`inhibits VEGF-induced proliferation of endothelial cells in vitro and
`tumorgrowthin vivo with potency andefficacy very similar to those of
`muMAb VEGFA.4.6.1. Therefore, recombinant humanized MAb
`VEGFis suitable to test the hypothesis that inhibition of VEGF-
`induced angiogenesis is a valid strategy for the treatment of solid
`tumors and other disorders in humans.
`
`INTRODUCTION
`
`It is now well established that angiogenesis is implicated in the
`pathogenesis of a variety of disorders. These include solid tumors,
`intraocular neovascular syndromessuchas proliferative retinopathies
`or AMD,? rheumatoidarthritis, and psoriasis (1, 2, 3). In the case of
`solid tumors, the neovascularization allows the tumorcells to acquire
`a growth advantage and proliferative autonomy compared to the
`normal cells. Accordingly, a correlation has been observed between
`density of microvessels in tumor sections and patient survival
`in
`breast canceras well as in several other tumors (4-6).
`The search for positive regulators of angiogenesis has yielded
`several candidates, including acidic fibroblast growth factor (FGF),
`bFGF, transforming growth factor a, transforming growth factor B,
`hepatocyte growth factor, tumor necrosis factor-a, angiogenin, inter-
`leukin 8, and others (1, 2). However, in spite of extensive research,
`there is still uncertainty as to their role as endogenous mediators of
`angiogenesis. The negative regulators thus far identified include
`thrombospondin (7), the M, 16,000 NH,-terminal fragmentof prolac-
`tin (8), angiostatin (9), and endostatin (10).
`Work doneoverthe last several years has established the key role
`of VEGFin the regulation of normal and abnormal angiogenesis(11).
`The finding that the loss of even a single VEGFallele results in
`
`embryonic lethality points to an irreplaceable role played by this
`factor in the development and differentiation of the vascular system
`(11). Also, VEGF has been shownto be a key mediator of neovas-
`cularization associated with tumors and intraocular disorders (11).
`The VEGF mRNAis overexpressed by the majority of human tumors
`examined (12-16). In addition, the concentration of VEGF in eye
`fluids is highly correlated to the presence of active proliferation of
`blood vessels in patients with diabetic and other ischemia-related
`retinopathies (17). Furthermore, recent studies have demonstrated the
`localization of VEGFin choroidal neovascular membranesin patients
`affected by AMD(18).
`The muMAbVEGFA.4.6.1 (19) has been used extensively to
`test the hypothesis that VEGFis a mediator of pathological angio-
`genesis in vivo. This high affinity MAb is able to recognize all
`VEGFisoforms (19) and has been shown to inhibit potently and
`reproducibly the growth of a variety of human tumorcell lines in
`nude mice (11, 20-23). Moreover, intraocular administration of
`muMAbVEGFA.4.6.1 resulted in virtually complete inhibition of
`iris neovascularization secondary to retinal ischemia in a primate
`model (24).
`A majorlimitation in the use of murine antibodies in human therapy
`is the anti-globulin response (25, 26). Even chimeric molecules, where
`the variable (V) domains of rodent antibodies are fused to human
`constant(C) regions,arestill capable ofeliciting a significant immune
`response (27). A powerful approach to overcomethese limitations in
`the clinical use of monoclonal antibodies is “humanization” of the
`murine antibody. This approach was pioneered by Joneset al. (28)
`and Riechmanetal. (29), whofirst transplanted the CDRsof a murine
`antibody into human V domainsantibody.
`In the present article, we report on the humanization of muMAb
`VEGFA.4.6.1. Our strategy wasto transfer the six CDRs, as defined
`by Kabat et al. (30), from muMAb VEGF A.4.6.1 to a consensus
`human framework used in previous humanizations (31-33). Seven
`framework residues in the humanized variable heavy (VH) domain
`and one framework residue in the humanized variable light (VL)
`domain were changed from human to murine to achieve binding
`equivalent to muMAb VEGFA.4.6.1. This humanized MAbissuit-
`able for clinical trials to test the hypothesis that inhibition of VEGF
`action is an effective strategy for the treatment of cancer and other
`disorders in humans.
`
`MATERIALS AND METHODS
`
`Received 5/27/97; accepted 8/16/97.
`The costs of publication ofthis article were defrayed in part by the payment ofpage
`charges. This article must therefore be hereby marked advertisement in accordance with
`18 U.S.C. Section 1734 solely to indicate this fact.
`'To whom requests for reprints should be addressed, at Department of Cardio-
`vascular Research, Genentech,
`Inc., 460 Point San Bruno Boulevard, South San
`Francisco, CA 94080. Phone: (415) 225-2968; Fax: (415) 225-6327; E-mail: Ferrara.
`Napoleone@gene.com.
`? The abbreviations used are: AMD, age-related macular degeneration; bFGF, basic
`fibroblast growth factor, VEGF, vascular endothelial growth factor; MAb, monoclonal
`antibody; muMAb, murine MAb; rhuMAb,recombinant humanized MAb; CDR,comple-
`mentarity-determining region.
`
`Cloning of Murine Mab A.4.6.1 and Construction of Mouse-Human
`Chimeric Fab. Total RNA wasisolated from hybridomacells producing
`the anti-VEGF MAb A.4.6.1 using RNAsol (Tel-Test) and reverse-tran-
`scribed to cDNA using Oligo-dT primer and the SuperScript II system (Life
`Technologies, Inc., Gaithersburg, MD). Degenerate oligonucleotide primer
`pools, based of the NH,-terminal amino acid sequences of the light and
`heavy chains of the antibody, were synthesized and used as forward
`primers. Reverse primers were based on framework 4 sequences obtained
`from murine light chain subgroup «V and heavy chain subgroupII (30).
`After PCR amplification, DNA fragments were ligated to a TA cloning
`vector (Invitrogen, San Diego, CA). Eight clones each of the light and
`Pfizer v. Genentech
`4593
`Pfizer v. Genentech
`IPR2017-01488
`IPR2017-01488
`Genentech Exhibit 2021
`Genentech Exhibit 2021
`
`

`

`HUMANIZATION OF AN ANTI-VEGF MONOCLONAL ANTIBODY
`
`heavy chains were sequenced. Oneclone with a consensus sequencefor the
`light chain VL domain and one with a consensus sequence for the heavy
`chain VH domain were subcloned, respectively, into the pEMX1 vector
`containing the human CL and CH1 domains(31), thus generating a mouse-
`human chimeric F(ab). This chimeric F(ab) consisted of the entire murine
`A.4.6.1 VH domain fused to a human CH1 domain at amino acid SerH1 13,
`and the entire murine A.4.6.1 VL domain fused to a human CL domain at
`
`centrifugation in a 1-liter centrifuge bottle at 3000 X g, and the supernatant
`was removed. After freezing for 1 h, the pellet was resuspended in 25 ml
`of cold 10 mM Tris, 1 mm EDTA,and 20% sucrose, pH 8.0. Two hundred
`fifty ml of 0.1 M benzamidine (Sigma Chemical Co., St. Louis, MO) was
`addedto inhibit proteolysis. After gentle stirring on ice for 3 h, the sample
`wascentrifuged at 40,000 < g for 15 min. The supernatant wasthen applied
`to a protein G-Sepharose CL-4B (Pharmacia Biotech, Inc., Uppsala, Swe-
`amino acid LysL107. Expression and purification of the chimeric F(ab)
`den) column (0.5-ml bed volume) equilibrated with 10 mm Tris-1 mm
`were identical to those of the humanized F(ab)s. The chimeric F(ab) was
`EDTA,pH 7.5. The column was washed with 10 ml of 10 mM Tris-1 mm
`used as the standard in the binding assays.
`EDTA,pH7.5, and eluted with 3 ml of 0.3 M glycine, pH 3.0,into 1.25 ml
`Computer Graphics Models of Murine and Humanized F(ab)s. Se-
`of 1 M Tris, pH 8.0. The F(ab) was then buffer exchanged into PBS using
`quencesof the VL and VH domains(Fig. 1) were used to construct a computer
`a Centricon-30 (Amicon, Beverly, MA) and concentratedto a final volume
`graphics model of the murine A.4.6.1 VL-VH domains. This model was used
`of 0.5 ml. SDS-PAGEgels ofall F(ab)s were run to ascertain purity, and
`to determine which framework residues should be incorporated into the hu-
`the molecular weight of each variant was verified by electrospray mass
`spectrometry.
`manized antibody. A model of the humanized F(ab) was also constructed to
`verify correct selection of murine framework residues. Construction of models
`Construction, Expression, and Purification of Chimeric and Human-
`was performed as described previously (32, 33).
`ized IgG Variants. For the generation of human IgG1 variants of chimeric
`Construction of Humanized F(ab)s. The plasmid pEMX1 used for mu-
`(chIgG1) and humanized (rhuMAb VEGF) A.4.6.1, the appropriate murine or
`humanized VL and VH (F(ab)-12; Table 1) domains were subcloned into
`tagenesis and expression of F(ab)s in Escherichia coli has been described
`separate, previously described pRK vectors (35). The DNA coding for the
`previously (31). Briefly, the plasmid contains a DNA fragment encoding a
`consensus human « subgroupIlight chain (VL«I-CL) and a consensus human
`entire light and the entire heavy chain of each variant was verified by
`subgroupIII heavy chain (VHIII-CH1) and an alkaline phosphatase promoter.
`dideoxynucleotide sequencing.
`The use of the consensus sequences for VL and VH has been described
`For transient expression of variants, heavy and light chain plasmids were
`previously (32).
`cotransfected into human 293cells (36) using a high efficiency procedure (37).
`the first F(ab) variant of humanized A.4.6.1, F(ab)-1,
`To construct
`Media were changed to serum free and harvested daily for up to 5 days.
`site-directed mutagenesis (34) was performed on a deoxyuridine-containing
`Antibodies were purified from the pooled supernatants using protein A-
`template of pEMX1. The six CDRs were changed to the murine A.4.6.1
`Sepharose CL-4B (Pharmacia). The eluted antibody was buffer exchanged into
`sequence; the residues included in each CDR were from the sequence-based
`PBS using a Centricon-30 (Amicon), concentrated to 0.5 ml, sterile filtered
`using a Millex-GV (Millipore, Bedford, MA), and stored at 4°C.
`CDRdefinitions (30). F(ab)-1, therefore, consisted of a complete human
`framework (VL « subgroup I and VH subgroupIII) with the six complete
`For stable expression of the final humanized IgG1 variant (rhuMAb
`murine CDR sequences. Plasmids for all other F(ab) variants were con-
`VEGF), Chinese hamster ovary (CHO) cells were transfected with dicis-
`structed from the plasmid template of F(ab)-1. Plasmids were transformed
`tronic vectors designed to coexpress both heavy and light chains (38).
`Plasmids were introduced into DP12 cells, a proprietary derivative of the
`into E. coli strain XL-1 Blue (Stratagene, San Diego, CA) for preparation
`of double- and single-stranded DNA.For each variant, DNA coding for
`CHO-K1 DUX B11 cell line developed by L. Chasin (Columbia University,
`light and heavy chains was completely sequenced using the dideoxynucle-
`New York, NY), via lipofection and selected for growth in glycine/
`otide method (Sequenase; U.S. Biochemical Corp., Cleveland, OH). Plas-
`hypoxanthine/thymidine (GHT)-free medium (39). Approximately 20 un-
`mids were transformed into E. coli strain 16C9, a derivative of MM294,
`amplified clones were randomly chosen and reseeded into 96-wellplates.
`plated onto Luria broth plates containing 50 yg/ml carbenicillin, and a
`Relative specific productivity of each colony was monitored using an
`ELISAto quantitate the full-length human IgG accumulated in each well
`single colony selected for protein expression. The single colony was grown
`in 5 ml of Luria broth-100 ug/ml carbenicillin for 5—8 h at 37°C. The 5-ml
`after 3 days and a fluorescent dye, Calcien AM,as a surrogate marker of
`viable cell number per well. Based on these data, several unamplified
`culture was added to 500 ml of AP5-50 g/ml carbenicillin and allowed to
`clones were chosen for further amplification in the presence of increasing
`grow for 20hin a 4-liter baffled shake flask at 30°C. AP5 media consists
`of 1.5 g of glucose, 11.0 g of Hycase SF, 0.6 g of yeast extract (certified),
`concentrations of methotrexate. Individual clones surviving at 10, 50, and
`0.19 g of MgSO, (anhydrous), 1.07 g of NH,Cl, 3.73 g of KCl, 1.2 g of
`100 nm methotrexate were chosen and transferred to 96-well plates for
`NaCl, 120 ml of 1 M triethanolamine, pH 7.4, to | liter of water and then
`productivity screening. One clone, which reproducibly exhibited high spe-
`sterile filtered through a 0.1-mm Sealkeenfilter. Cells were harvested by
`cific productivity, was expanded in T-flasks and usedto inoculate a spinner
`
`Table 1 Binding of humanized anti-VEGF F(ab) variants to VEGF?
`
`ECS0 F(ab)-X
`
`Variant
`chim-F(ab)
`F(ab)-1
`F(ab)-2
`
`F(ab)-3
`
`F(ab)-4
`
`Fcab)-5
`F(ab)-6
`F(ab)-7
`F(ab)-8
`
`Template
`Chimeric F(ab)
`Human FR
`
`Changes?
`
`F(ab)-1
`
`F(ab)-4
`F(ab)-5
`F(ab)-S
`F(ab)-5
`
`Purpose
`1.0
`Straight CDR swap
`Chimera light chain
`F(ab)-1 heavy chain
`F(ab)-1 light chain
`Chimera heavy chain
`CDR-H2 conformation
`ArgH71Leu
`Framework
`AspH73Asn
`VL-VHinterface
`LeuL46Val
`CDR-H1 conformation
`LeuH78Ala
`CDR-H2 conformation
`TleH69Phe
`CDR-H2conformation
`TleH69Phe
`CDR-H1 conformation
`LeuH78Ala
`>150
`CDR-H2conformation
`_GlyH49Ala
`F(ab)-8
`F(ab)-9
`6.4
`Framework
`AsnH76Ser
`F(ab)-8
`F(ab)-10
`3.3
`Framework
`LysH75Ala
`F(ab)-10
`F(ab)-11
`1.6
`CDR-H3conformation
`ArgH94Lys
`F(ab)-10
`F(ab)-12
`* Anti-VEGF F(ab)variants were incubated with biotinylated VEGFand then transferred to ELISA plates coated with KDR-IgG (40).
`> Murine residues are underlined; residue numbers are according to Kabat etal. (30).
`© Mean and SDare the average ofthe ratios calculated for each of the independent assays; the ECs, for chimeric F(ab) was 0.049 + 0.013 mg/ml (1.0 nm).
`4594
`
`Mean
`
`>1350
`>145
`
`2.6
`
`>295
`
`80.9
`36.4
`45.2
`9.6
`
`ECs chimeric F(ab)°
`SD
`
`0.1
`
`6.5
`4.2
`23
`0.9
`
`L2
`0.4
`0.6
`
`N
`
`2
`3
`
`2
`
`3
`
`2
`2
`2
`4
`
`2
`4
`2
`4
`
`

`

`HUMANIZATION OF AN ANTI-VEGF MONOCLONAL ANTIBODY
`
`RESULTS
`
`antibiotics (growth medium), essentially as described previously (42). For
`culture. After several passages, the suspension-adapted cells were used to
`mitogenic assays, endothelial cells were seeded at a density of 6 X 10°
`inoculate production cultures in GHT-containing, serum-free media sup-
`cells/well in 6-well plates in growth medium. Either muMAb VEGFA.4.6.1 or
`plemented with various hormonesand protein hydrolysates. Harvested cell
`rhuMAb VEGFwas then addedat concentrations ranging between | and 5000
`culture fluid containing rhuMAb VEGFwaspurified using protein A-
`Sepharose CL-4B. The purity after this step was ~99%. Subsequent
`ng/ml. After 2-3 h, purified E. coli-expressed rhWEGF,,. was addedtoafinal
`concentration of 3 ng/ml. For specificity control, each antibody was added to
`purification to homogeneity was carried out using an ion exchange chro-
`endothelial cells at the concentration of 5000 ng/ml, either alone or in the
`matographystep. The endotoxin content of the final purified antibody was
`<0.10 eu/mg.
`presence of 2 ng/ml bFGF. After 5 or 6 days, cells were dissociated by
`exposure to trypsin, and duplicate wells were counted in a Coulter counter
`F(ab) and IgG Quantitation. For quantitating F(ab) molecules, ELISA
`(Coulter Electronics, Hialeah, FL). The variation from the mean did not exceed
`plates were coated with 2 g/ml of goat anti-human IgG Fab (Organon
`Teknika, Durham, NC) in 50 mmcarbonate buffer, pH 9.6, at 4°C overnight
`10%. Data were analyzed by a four-parameter curvefitting program (Kalei-
`daGraph).
`and blocked with PBS-0.5% BSA (blocking buffer) at room temperature for
`In Vivo Tumor Studies. Human A673 rhabdomyosarcomacells (Amer-
`1 h. Standards (0.78-50 ng/ml human F(ab)] were purchased from Chemicon
`ican Type Culture Collection; CRL 1598) were cultured as described
`(Temecula, CA). Serial dilutions of samples in PBS-0.5% BSA-0.05% poly-
`previously in DMEM/F12 supplemented with 10% fetal bovine serum, 2
`sorbate 20 (assay buffer) were incubated on the plates for 2h. Bound F(ab) was
`mMglutamine, and antibiotics (20, 22). Female BALB/c nude mice, 6-10
`detected using horseradish peroxidase-labeled goat anti-human IgG F(ab)
`weeksold, were injected s.c. with 2 < 10° tumorcells in the dorsal area in
`(Organon Teknika), followed by 3,3’,5,5'-tetramethylbenzidine (Kirkegaard &
`a volume of 200 ul. Animals were then treated with muMAb VEGF
`Perry Laboratories, Gaithersburg, MD) as the substrate. Plates were washed
`A.4.6.1, rhuMAb VEGF,or a control murine MAbdirected against the
`between steps. Absorbance was read at 450 nm on a V,,,, plate reader
`gp120 protein. Both anti-VEGF MAbs were administered at the doses of
`(Molecular Devices, Menlo Park, CA). The standard curve was fit using a
`0.5 and 5 mg/kg; the control MAb wasgiven at the dose of 5 mg/kg. Each
`four-parameter nonlinear
`regression curve-fitting program developed at
`Genentech. Data points that fell in the range of the standard curve were used
`MAbwasadministered twice weekly i.p. in a volume of 100 yl, starting
`24 h after tumorcell inoculation. Each group consisted of 10 mice. Tumor
`for calculating the F(ab) concentrations of samples.
`size was determined at weekly intervals. Four weeks after tumor cell
`The concentration of full-length antibody was determined using goat anti-
`inoculation, animals were euthanized, and the tumors were removed and
`human IgG Fc (Cappel, Westchester, PA) for capture and horseradish perox-
`weighed. Statistical analysis was performed by ANOVA.
`idase-labeled goat anti-human Fc (Cappel) for detection. Human IgG1 (Chemi-
`con) was used as standard.
`VEGFBinding Assays. For measuring the VEGFbinding activity of
`F(ab)s, ELISA plates were coated with 2 ug/ml rabbit F(ab’), to human
`IgG Fe (Jackson ImmunoResearch, West Grove, PA) and blocked with
`blocking buffer (described above). Diluted conditioned medium containing
`3 ng/ml of KDR-IgG (40)in blocking buffer were incubated on the plate for
`I h. Standards [6.9—440 ng/ml chimeric F(ab)] and 2-fold serial dilutions
`of samples were incubated with 2 nm biotinylated VEGFfor 1 h in tubes.
`The solutions from the tubes were then transferred to the ELISA plates and
`incubated for 1 h. After washing, biotinylated VEGF bound to KDR was
`detected using horseradish peroxidase-labeled streptavidin (Zymed, South
`San Francisco, CA or Sigma)followed by 3,3',5,5’-tetramethylbenzidine as
`the substrate. Titration curves were fit with a four-parameter nonlinear
`regression curve-fitting program (KaleidaGraph; Synergy Software, Read-
`ing, PA). Concentrations of F(ab) variants corresponding to the midpoint
`absorbanceofthe titration curve of the standard were calculated and then
`
`Humanization. The consensus sequence for the human heavy
`chain subgroupIII and the light chain subgroup « I were used as the
`framework for the humanization (Ref. 30; Fig. 1). This framework has
`been successfully used in the humanization of other murine antibodies
`(31, 32, 43, 44). All humanized variants were initially made and
`screened for binding as F(ab)s expressed in E. coli. Typical yields
`from 500-ml shake flasks were 0.1—0.4 mg F(ab).
`Twodefinitions of CDR residues have been proposed. Oneis based
`on sequence hypervariability (30) and the other on crystal structures
`of F(ab)-antigen complexes (45). The sequence-based CDRs are
`larger than the structure-based CDRs, and the two definitions are in
`agreement except for CDR-H1; CDR-H1 includes residues H31—H35
`according to the sequence-based definition, and residues H26—H32
`according to the structure-based definition (light chain residue num-
`bers are prefixed with L; heavy chain residue numbers are prefixed
`BlAcore Biosensor Assays. VEGFbinding ofthe humanized and chimeric
`with H). We,therefore, defined CDR-H1 as a combination ofthe two,
`F(ab)s were compared using a BlAcore biosensor (41). Concentrations of
`i.e., including residues H26-H35. The other CDRswere defined using
`F(ab)s were determined by quantitative amino acid analysis. VEGF was
`the sequence-based definition (30).
`coupled to a CM-5 biosensor chip through primary amine groups according to
`The chimeric F(ab) wasusedas the standard in the binding assays.
`manufacturer's instructions (Pharmacia). Off-rate kinetics were measured by
`In the initial variant, F(ab)-1, the CDR residues were transferred from
`saturating the chip with F(ab) (35 jl of 2 um F(ab)at a flowrate of 20 l/min]
`the murine antibody to the human framework and, based on the
`and then switching to buffer (PBS-0.05% polysorbate 20). Data points from
`models of the murine and humanized F(ab)s, the residue at position
`0-4500 s were used for off-rate kinetic analysis. The dissociation rate constant
`H49 (Ala in humans) was changed to the murine Gly. In addition,
`(koe) Was obtained from the slope of the plot of In(RO/R) versus time, where RO
`F(ab)s that consisted of the chimeric heavy chain/F(ab)-1 light chain
`is the signal at t = 0 andRisthe signal at each time point.
`[F(ab)-2] and F(ab)-1 heavy chain/chimeric light chain [F(ab)-3] were
`On-rate kinetics were measured using 2-fold serial dilutions of F(ab)
`(0.0625-2 mm). The slope, K,, was obtained from the plot of In(—dR/dt)
`generated and tested for binding. F(ab)-1 exhibited a bindingaffinity
`versus time for each F(ab) concentration using the BIAcore kinetics eval-
`greater than 1000-fold reduced from the chimeric F(ab) (Table 1).
`uation software as described in the Pharmacia Biosensor manual. R is the
`Comparing the binding affinities of F(ab)-2 and F(ab)-3 suggested
`signalat time r. Data between 80 and 168, 148, 128, 114, 102, and 92 s were
`that framework residues in the F(ab)-1 VH domain needed to be
`used for 0.0625, 0.125, 0.25, 0.5, 1, and 2 mm F(ab), respectively. The
`altered to increase binding.
`association rate constant (k,,,) was obtained from the slope ofthe plot of K,
`Previous humanizations (31, 32, 43, 44) as well as studies of
`versus F(ab) concentration. At the end of each cycle, bound F(ab) was
`F(ab)-antigen crystal structures (45, 47) have shownthat residues H71
`removed by injecting 5 ul of 50 mm HCIat a flow rate of 20 l/min to
`and H73 can have a profound effect on binding, possibly by influ-
`regenerate the chip.
`encing the conformations of CDR-H1 and CDR-H2. Changing the
`Endothelial Cell Growth Assay. Bovine adrenal cortex-derived capillary
`humanresidues to their murine counterparts in F(ab)-4 improved
`endothelial cells were cultured in the presence of low glucose DMEM(Life
`Technologies, Inc.) supplemented with 10% calf serum, 2 mM glutamine, and
`binding by 4-fold (Table 1). Inspection of the models of the murine
`4595
`
`divided by the concentration of the standard corresponding to the midpoint
`absorbanceofthe standard titration curve. Assays for full-length IgG were
`the same as for the F(ab)s except that the assay buffer contained 10%
`humanserum.
`
`

`

`HUMANIZATION OF AN ANTI-VEGF MONOCLONAL ANTIBODY
`
`Variable Heavy
`
`A.4.6.1
`
`F(ab)-12
`
`humIII
`
`EIQLVOSGPELKQPGETVRISCKASGYTFINYGMNWVKQAPGKGLKWMG
`*
`*
`kk *
`keke
`*&
`*
`*
`x *
`EVQLVESGGGLVOPGGSLRLSCAASGYTFINYGMNWVROAP GKGLEWVG
`* kek &
`*
`EVQLVESGGGLVQPGGSLRLSCAASGFTFSS YAMSWVROQAPGKGLEWVS
`1
`10
`20
`30
`40
`
`A.4.6.1
`
`F(ab) -12
`
`humIII
`
`WINTYTGEPTYAADFKRRFTFSLETSASTAYLQISNLKNDDTATYFCAK
`*
`*
`kkk kkKK
`xk *
`‘TFSLDTSKSTAYLQMNSLRAEDTAVYYCAK
`nz kek
`x *
`*
`kak *
`wk KKK KKK
`VISGDGGSTYYADSVKGRFTISRDNSKNTLYLOMNSLRAEDTAVYYCAR
`50
`a
`60
`70
`80
`abc
`90
`
`A.4.6.1
`
`XPHYYGSSHWYFDVWGAGTTVIVSS
`*
`*
`F(ab)-12 XYPHYYGSSHWYFDVWGOQGTLVTVSS
`*
`*
`G----------FDYWGQGTLVTVSS
`110
`
`humIII
`
`A.4.6.1
`
`Variable Light
`DIQMTOTTSSLSASLGDRVI ISCSASODISNYLNWYQQKPDGTVKVLIY
`ak
`*
`x
`*&
`keke
`F(ab)-12 DIQMTQSPSSLSASVGDRVTITCSASODISNYLNWYOQOKPGKAPKVLIY
`*
`*
`*
`x
`
`humKI
`
`DIQMTOSPSSLSASVGDRVT ITCRASQSISNYLAWYQQKPGKAPKLLIY
`1
`10
`20
`30
`40
`
`A.4.6.1
`
`EISSLHSGVP SRFSGSGSGTDYSLTISNLEPEDIATYYCOQQYSTVPWIF
`xk
`x *
`*
`
`xk
`*
`kkk
`F(ab)-12 EISSLHSGVPSRFSGSGSGTDFTLTISSLOPEDFATYYCOOYSTVPWTF _
`humKI
`AASSLESGVPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQYNSLPWIF
`50
`60
`70
`80
`90
`
`A.4.6.1
`
`GGGTKLEIKR
`*
`*
`F(ab)-12 GQGTKVEIKR
`
`humKI
`
`GQGTKVEIKR
`100
`
`Fig. 1. Amino acid sequence of variable heavy and light domains of muMAbVEGF
`A.4.6.1, humanized F(ab) with optimal VEGF binding [F(ab)-12] and human consensus
`frameworks (humill, heavy subgroup III; humx/, light x subgroup I). Asterisks, differ-
`ences between humanized F(ab)-12 and the murine MAbor between F(ab)-12 and the
`human framework. CDRsare underlined.
`
`H94, human and murine sequences most often have an Arg (30). In
`F(ab)-12, this Arg was replaced by the rare Lys found in the murine
`antibody (Fig. 1), andthis resulted in binding that was less than 2-fold
`from the chimeric F(ab) (Table 1). F(ab)-12 was also compared to the
`chimeric F(ab) using the BlAcore system (Pharmacia). Using this
`technique, the K, of the humanized F(ab)-12 was 2-fold weaker than
`that of the chimeric F(ab) due to both a slower k,,, and faster koe,
`(Table 2).
`Full-length MAbs were constructed by fusing the VL and VH
`domains of the chimeric F(ab) and variant F(ab)-12 to the constant
`domains of human « light chain and human IgG] heavy chain. The
`full-length 12-IgG1 [F(ab)-12 fused to human IgG1] exhibited bind-
`ing that was 1.7-fold weaker than the chimeric IgG1 (Table 3). Both
`12-IgG1 and the chimeric IgG1 boundslightly less well than the
`original muMAb VEGFA.4.6.1 (Table 3).
`Biological Studies. rhuMAb VEGF and muMAbVEGFA.4.6.1
`were compared for their ability to inhibit bovine capillary endo-
`thelial cell proliferation in response to a near maximally effective
`concentration of VEGF,,., (3 ng/ml). In several experiments, the
`two MAbswerefoundto be essentially equivalent, both in potency
`and efficacy. The ED.9s were, respectively, 50 + 5 and 48 + 8
`ng/ml (~0.3 nm). In both cases, 90% inhibition was achieved at the
`concentration of 500 ng/ml (~3 nm). Fig. 3 illustrates a represent-
`ative experiment. Neither muMAb VEGF A.4.6.1 nor rhuMAb
`VEGFhadanyeffect on basal or bFGF-stimulated proliferation of
`capillary endothelial cells (data not shown), confirming that the
`inhibition is specific for VEGF.
`To determine whethersimilar findings could be obtained also in an
`in vivo system, we compared the two antibodies for their ability to
`suppress the growth of human A673 rhabdomyosarcomacells in nude
`mice. Previous studies have shown that muMAb VEGFA.4.6.1 has a
`dramatic inhibitory effect in this tumor model (20, 22). As shown in
`Fig. 4, at both doses tested (0.5 and 5 mg/kg), the two antibodies
`markedly suppressed tumor growth as assessed by tumor weight
`measurements 4 weeks after cell inoculation. The decreases in tumor
`weight compared to the control group were, respectively, 85 and 93%
`at each dosein the animals treated with muMAb VEGFA.4.6.1 versus
`90 and 95% in those treated with rhuMAb VEGF.Similar results were
`obtained with the breast carcinomacell line MDA-MB 435 (data not
`shown).
`
`DISCUSSION
`
`and humanized F(ab)s suggested that residue L46, buried at the
`VL-VHinterface and interacting with CDR-H3(Fig. 2), might also
`play a role either in determining the conformation of CDR-H3
`The murine MAb A.4.6.1, directed against human VEGF(42),
`and/or affecting the relationship of the VL and VH domains. When
`was humanized using the same consensus frameworksfor the light
`the murine Val was exchangedfor the human Leuat L46 [F(ab)-5],
`and heavy chains used in previous humanizations (31, 32, 43, 44),
`the binding affinity increased by almost 4-fold (Table 1). Three
`other buried framework residues were evaluated based on the
`i.e., V«I and VHIII (30). Simply transferring the CDRs from the
`murine antibody to the human frameworkresulted in a F(ab) that
`molecular models: H49, H69, and H78. Position H69 mayaffect
`exhibited binding to VEGF reduced by over 1000-fold compared to
`the conformation of CDR-H2, whereas position H78 mayaffect the
`the parent murine antibody. Seven non-CDR, frameworkresidues
`conformation of CDR-H1 (Fig. 2). When each was individually
`in the VH domain and one in the VL domain were altered from
`changed from the human to murine counterpart, the binding im-
`human to murine to achieve binding equivalent
`to the parent
`proved by 2-fold in each case [F(ab)-6 and F(ab)-7; Table 1]. When
`both were simultaneously changed, the improvement in binding
`murine antibody.
`In the VH domain,residuesat positions H49, H69, H71, and H78
`was 8-fold [F(ab)-8; Table 1]. Residue H49 wasoriginally in-
`are buried or partially buried and probably effect binding by
`cluded as the murine Gly; when changed to the human consensus
`influencing the conformation of the CDR loops. Residues H73 and
`counterpart Ala,
`the binding was reduced by 15-fold [F(ab)-9;
`H76 should be solvent exposed (Fig. 2) and hence may interact
`Table 1).
`directly with the VEGF;these two residues are in a non-CDR loop
`Wehave found during previous humanizations that residues in a
`adjacent to CDRs H1 and H2 and have been showntoplaya role
`framework loop, FR-3 (30) adjacent to CDR-H1 and CDR-H2,can
`in binding in previous humanizations (31, 32, 44). The requirement
`affect binding (44). In F(ab)-10 and F(ab)-11, two residuesin this loop
`for lysine at position H94 was surprising given that this residue is
`were changed to their murine counterparts: AsnH76 to murine Ser
`arginine in the human framework(Fig. 1). In some crystal struc-
`[F(ab)-10] and LysH75 to murine Ala [F(ab)-11]. Both effected a
`tures of F(ab)s, ArgH94 forms a hydrogen-bondedsalt-bridge with
`relatively small improvementin binding (Table 1). Finally, at position
`4596
`
`

`

`HUMANIZATION OF AN ANTI-VEGF MONOCLONAL ANTIBODY
`
`Fig. 2. Ribbon diagram of the model of humanized F(ab)-12 VL and VH domains. VL domain is shown in brown with CDRs in tan. The side chain of residue L46 is shown in
`yellow. VH domain is shown in purple with CDRs in pink. Side chains of VH residues changed from human to murine are shown in yellow.
`
`Table 2 Binding of anti-VEGF F(ab) variants to VEGF using the BlAcore system*
`
`\
`Amountof (Fab)
`Kg (nm)
`kon (M7 'S7')
`kore (87!)
`bound (RU)
`Variant
`0.91
`6.5 x 10*
`5.9 x 1075
`4250
`chim-F(ab)”
`1.8
`3.5 x 10*
`63x 107°
`3740
`F(ab)-12
`* The amount of F(ab) bound,in resonance units (RU), was measured using a BIAcore
`system when 2 yg F(ab) was injected onto a chip containing 2480 RU of immobilized
`VEGF.Off-rate kinetics (kop) were measured bysaturating the chip with F(ab) and then
`monitoring dissociation after switching to buffer. On-rate kinetics (k,,) were measured
`using 2-fold serial dilutions of F(ab). Ky, the equilibrium dissociation constant, was
`calculated as kop@kon-
`A chim-F(ab) is a chimeric F(ab) with murine VL and VH domainsfused to human CL
`and CHI heavy domains.
`
`adjacent to CDR-H3, suggests that CDR-H3plays a major role in the
`binding of the antibody to VEGF.
`The humanized version with optimal binding, 12-IgG1, exhib-
`ited only a 2-fold reduction in binding compared to the parent
`murine antibody (Table 3). An analysis of the binding kinetics of
`the humanized and chimeric F(ab)s showed that both had similar
`off-rates but that the humanized F(ab) had a 2-fold slower on-rate
`(Table 2), which accounts for the 2-fold reduction in binding.
`However, this modest reduction in on-rate did not result in any
`decreased ability to antagonize VEGFbioactivity. The two anti-
`
`
`Table 3 Binding of anti-VEGF IgG variants to VEGF?
`
`AspH101 (33, 48). Substitution of lysine for arginine might con-
`ceivably alter this salt-bridge and perturb the conformation of
`CDR-H3.
`In the VL domain, only one framework residue had to be changed
`to murine to optimize the humanization. Position L46 is at the VL-VH
`interface, whereit is buried andinteracts directly with CDR-H3(Fig.
`2). The requirement for murine valine (as opposed to human leucine)
`implies that this residue plays an importantrole in the conformation of
`CDR-H3. The necess

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