throbber
[CANCER RESEARCH 57. 4593—4599. October l5. I997]
`
`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 Ferraral
`Depamnenrr of Immunology. Process Sciences, Molecular Biology. Bioanalytical Technology and Cardiovascular Research. Genenlech. Inc. South San Francisco. California
`94080
`
`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 murlne anti-human VEGF monoclonal antibody
`(muMAb VEGF) A.4.6.l has been shown to potently suppress angio-
`genesis and growth in a variety of human tumor cells 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 muMAb VEGF A.4.6.l. by site-directed mutagenesis of a human
`framework. Not only the residues involved in the six complementarity-
`determining regions but also several framework residues were changed
`from human to murine. Humanized anti-VEGF F(ab) and IgGI 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
`tumor growth in vivo with potency and efficacy very similar to those of
`muMAb VEGF A.4.6.l. Therefore, recombinant humanized MAb
`VEGF is 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 syndromes such as proliferative retinopathies
`or AMD,2 rheumatoid arthritis, and psoriasis (l, 2, 3). In the case of
`solid tumors. the neovascularization allows the tumor cells 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 cancer as 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 (i. 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 Mr 16,000 NHz-terminal fragment of prolac-
`tin (8), angiostatin (9), and endostatin (10).
`Work done over the last several years has established the key role
`of VEGF in the regulation of normal and abnormal angiogenesis (l l).
`The finding that the loss of even a single VEGF allele results in
`
`embryonic lethality points to an irreplaceable role played by this
`factor in the development and differentiation of the vascular system
`(1 1). Also, VEGF has been shown to be a key mediator of neovas-
`cularization associated with tumors and intraocular disorders (II).
`The VEGF mRNA is 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 VEGF in choroidal neovascular membranes in patients
`affected by AMD (18).
`The muMAb VEGF A.4.6.l (19) has been used extensively to
`test the hypothesis that VEGF is a mediator of pathological angio-
`genesis in vivo. This high affinity MAb is able to recognize all
`VEGF isoforms (l9) and has been shown to inhibit potently and
`reproducibly the growth of a variety of human tumor cell lines in
`nude mice (1 1, 20—23). Moreover, intraocular administration of
`muMAb VEGF A.4.6.l resulted in virtually complete inhibition of
`iris neovascularization secondary to retinal ischemia in a primate
`model (24).
`A major limitation 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, are still capable of eliciting a significant immune
`response (27). A powerful approach to overcome these limitations in
`the clinical use of monoclonal antibodies is “humanization" of the
`
`murine antibody. This approach was pioneered by Jones er al. (28)
`and Riechman et al. (29), who first transplanted the CDRs of a murine
`antibody into human V domains antibody.
`In the present article, we report on the humanization of muMAb
`VEGF A.4.6.l. Our strategy was to transfer the six CDRs, as defined
`by Kabat er a1. (30), from muMAb VEGF A.4.6.l
`to a consensus
`human framework used in previous humanizations (3l—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 VEGF A.4.6.l. This humanized MAb is suit-
`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/l6/97.
`The costs of publication of this article were defrayed in part by the payment of page
`charges. This article must therefore be hereby marked advertisement in accordance with
`IS 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.
`lnc.. 460 Point San Bruno Boulevard. South San
`Francisco. CA 94080. Phone: (4l5)225-2968: Fax: (4l5)225—6327; E-mail: Ferrara.
`Napoleone@gene.com.
`2 fire 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.l and Construction of Mouse-Human
`Chimeric Fab. Total RNA was isolated from hybridoma cells producing
`the anti-VEGF MAb A.4.6.l using RNAsol (Tel-Test) and reverse-tran-
`scribed to cDNA using Oligo-dT primer and the SuperScript ll system (Life
`Technologies. Inc., Gaithersburg. MD). Degenerate oligonucleotide primer
`pools. based of the NHz-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 KV and heavy chain subgroup ll (30).
`After PCR amplification. DNA fragments were ligated to a TA cloning
`vector (lnvitrogen. San Diego, CA). Eight clones each of the light and
`4593
`Pfizer v. Genentech
`
`Pfizer v. Genentech
`IPR2017-01488
`IPR2017—01488
`Genentech Exhibit 2021
`Genentech Exhibit 2021
`
`

`

`HUMANIZATION OF AN AN'I'LVEGF MONOCLONAL ANTIBODY
`
`heavy chains were sequenced. One clone with a consensus sequence for the
`light chain VL domain and one with a consensus sequence for the heavy
`chain VH domain were subcloned. respectively, into the pEMXl 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.l VH domain fused to a human CH1 domain at amino acid SerHl 13,
`and the entire murine A.4.6.l VL domain fused to a human CL domain at
`
`amino acid LysL107. Expression and purification of the chimeric F(ab)
`were identical to those of the humanized F(ab)s. The chimeric F(ab) was
`used as the standard in the binding assays.
`Computer Graphies Models of Murine and Humanized F(ab)s. Se-
`quences of the VL and VH domains (Fig. l) were used to construct a computer
`graphics model of the murine A.4.6.1 VL—VH domains. This model was used
`to detemrine which framework residues should be incorporated into the hu-
`manized antibody. A model of the humanized F(ab) was also constructed to
`verify conect selection of murine framework residues. Construction of models
`was performed as described previously (32, 33).
`Construction _of Humanized F(ab)s. The plasmid pEMXl used for mu-
`tagenesis and expression of F(ab)s in Escherichia cali has been described
`previously (31). Briefly, the plasmid contains a DNA fragment encoding a
`consensus human K subgroup I light chain (VLKI-CL) and a consensus human
`subgroup III heavy chain (VHIIl-CHl) and an alkaline phosphatase promoter.
`The use of the consensus sequences for VL and VH has been described
`previously (32).
`the first F(ab) variant of humanized A.4.6.1, F(ab)-l.
`To construct
`site—directed mutagenesis (34) was performed on a deoxyuridine-containing
`template of pEMXl. The six CDRs were changed to the murine A.4.6.1
`sequence; the residues included in each CDR were from the sequence-based
`CDR definitions (30). F(ab)-1, therefore, consisted of a complete human
`framework (VL K subgroup l and VH subgroup III) with the six complete
`murine CDR sequences. Plasmids for all other F(ab) variants were con-
`structed from the plasmid template of F(ab)-1. Plasmids were transformed
`into E. coli strain XL-l Blue (Stratagene, San Diego. CA) for preparation
`of double— and single-stranded DNA. For each variant, DNA coding for
`light and heavy chains was completely sequenced using the dideoxynucle-
`otide method (Sequenase; U.S. Biochemical Corp.. Cleveland, OH). Plas-
`mids were transformed into E. coli strain 16C9, a derivative of MM294,
`plated onto Luria broth plates containing 50 ug/ml carbenicillin, and a
`single colony selected for protein expression. The single colony was grown
`in 5 ml of Luria broth—100 [ig/ml carbenicillin for 5—8 h at 37°C. The 5-m1
`culture was added to 500 ml of AP5—50 [Lg/ml carbenicillin and allowed to
`grow for 20 h in 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),
`0.19 g of MgSO4 (anhydrous), 1.07 g of NH‘CI. 3.73 g of KC], 1.2 g of
`NaCl, 120 ml of l M triethanolamine. pH 7.4, to 1 liter of water and then
`sterile filtered through a 0.1-mm Sealkeen filter. Cells were harvested by
`
`centrifugation in a l-liter centrifuge bottle at 3000 X g. and the supernatant
`was removed. After freezing for l 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
`added to inhibit proteolysis. After gentle stirring on ice for 3 h. the sample
`was centrifuged at 40,000 X g for 15 min. The supernatant was then applied
`to a protein G-Sepharose CL-4B (Pharmacia Biotech, Inc., Uppsala, Swe-
`den) column (0.5-ml bed volume) equilibrated with 10 mM Tris-l mM
`EDTA, pH 7.5. The column was washed with 10 ml of 10 mM Tris-l mM
`EDTA, pH 7.5, and eluted with 3 ml of 0.3 M glycine, pH 3.0. into 1.25 ml
`of l M Tris, pH 8.0. The F(ab) was then buffer exchanged into PBS using
`a Centricon-30 (Amicon, Beverly. MA) and concentrated to a final volume
`of 0.5 ml. SDS-PAGE gels of all F(ab)s were run to ascertain purity, and
`the molecular weight of each variant was verified by electrospray mass
`spectrometry.
`Construction, Expression, and Purification of Chimeric and Human-
`ized lgG Variants. For the generation of human IgGl variants of chimeric
`(chIgGl) and humanized (rhuMAb VEGF) A.4.6.l, the appropriate murine or
`humanized VL and VH (F(ab)-12; Table 1) domains were subcloned into
`separate, previously described pRK vectors (35). The DNA coding for the
`entire light and the entire heavy chain of each variant was verified by
`dideoxynucleotide sequencing.
`For transient expression of variants. heavy and light chain plasmids were
`cotransfected into human 293 cells (36) using a high efficiency procedure (37).
`Media were changed to serum free and harvested daily for up to 5 days.
`Antibodies were purified from the pooled supematants using protein A-
`Sepharose CL-4B (Pharrnacia). The eluted antibody was buffer exchanged into
`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.
`For stable expression of the final humanized IgGl variant (rhuMAb
`VEGF), Chinese hamster ovary (CHO) cells were transfected with dicis-
`tronic vectors designed to coexpress both heavy and light chains (38).
`Plasmids were introduced into DP12 cells, a proprietary derivative of the
`CHO-Kl DUX 811 cell line developed by L. Chasin (Columbia University,
`New York. NY), via lipofection and selected for growth in glycine/
`hypoxanthine/thymidine (GHT)-free medium (39). Approximately 20 un-
`amplified clones were randomly chosen and reseeded into 96-well plates.
`Relative specific productivity of each colony was monitored using an
`ELISA to quantitate the full-length human lgG accumulated in each well
`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
`clones were chosen for further amplification in the presence of increasing
`concentrations of methotrexate. Individual clones surviving at 10. 50, and
`100 nM methotrexate were chosen and transferred to 96-well plates for
`productivity screening. One clone. which reproducibly exhibited high spe—
`cific productivity, was expanded in T-flasks and used to inoculate a spinner
`
`Table 1 Binding of humanized anti-VEGF F(ab) variants to VEGF“
`
`Variant
`chim-F(ab)
`F(ab)-l
`F(ab)-2
`
`F(abH
`
`F(ab)-4
`
`F(abys
`F(ab)«6
`F(ab)-7
`F(ab)-8
`
`Template
`Chimeric F(ab)
`Human FR
`
`Changesb
`
`F(ab)-l
`
`F(ab)-4
`F(ab)«5
`F(ab)-5
`F(ab)—5
`
`ArgH71_Lg
`AspH73fl
`hut/WE
`LeuH78Ala
`namfi
`[leH69E
`LeuH78Ala
`
`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
`Framework
`VL-VH interface
`CDR-H1 conformation
`CDR-HZ conformation
`CDR-H2 conformation
`CDR-H1 conformation
`
`Mean
`
`>1350
`>145
`
`2.6
`
`>295
`
`80.9
`36.4
`45.2
`9.6
`
`ECSO F(ab)-X
`
`ECSO chimeric F(ab)‘
`SD
`
`0.1
`
`6.5
`4.2
`2.3
`0.9
`
`>150
`CDR—H2 conformation
`flfl49fl
`F(ab)-8
`F(ab)-9
`6.4
`Framework
`Aan76Ser
`F(ab)-8
`F(ab)- 10
`3.3
`Framework
`LysH75ATa
`F(ab)-10
`F(ab)-l 1
`1.6
`CDR-H3 conformation
`ArgH94L—ys
`F(ab)-10
`F(ab)-12
`" Anti-VEGF F(ab) variants were incubated with biotinylated VEGF and then transferred to ELISA plates coated with KDR-lgG (40).
`b Murine residues are underlined; residue numbers are according to Kabat et a1. (30).
`‘ Mean and SD are the average of the ratios calculated for each of the independent assays; the EC50 for chimeric F(ab) was 0.049 1‘ 0.013 mg/ml (1.0 M).
`4594
`
`l .2
`0.4
`0.6
`
`N
`
`2
`3
`
`2
`
`3
`
`2
`2
`2
`4
`
`2
`4
`2
`4
`
`

`

`HUMANIZATION OF AN ANTI~VEGF MONOCLONAL ANTIBODY
`
`antibiotics (growth medium). essentially as described previously (42). For
`mitogenic assays, endothelial cells were seeded at a density of 6 X I03
`cells/well in 6—well plates in growth medium. Either muMAb VEGF A.4.6.l or
`rhuMAb VEGF was then added at concentrations ranging between I and 5000
`ng/ml. After 2—3 h. purified E. cali-expressed rhVEGF”,5 was added to a final
`concentration of 3 ng/ml. For specificity control, each antibody was added to
`endothelial cells at the concentration of 5000 ng/ml, either alone or in the
`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
`(Coulter Electronics. Hialeah. FL). The variation from the mean did not exceed
`10%. Data were analyzed by a four-parameter curve fitting program (Kalei-
`daGraph).
`In Vivo Tumor Studies. Human A673 rhabdomyosarcoma cells (Amer-
`ican Type Culture Collection; CRL l598) were cultured as described
`previously in DMEM/FIZ supplemented with 10% fetal bovine serum. 2
`mM glutamine. and antibiotics (20, 22). Female BALB/c nude mice. 6-40
`weeks old. were injected s.c. with 2 X 10" tumor cells in the dorsal area in
`a volume of 200 [1.1. Animals were then treated with muMAb VEGF
`A.4.6.l. rhuMAb VEGF. or a control murine MAb directed against the
`gpl20 protein. Both anti-VEGF MAbs were administered at the doses of
`0.5 and 5 mg/kg; the control MAb was given at the dose of 5 mg/kg. Each
`MAb was administered twice weekly i.p. in a volume of mo pl. starting
`24 h after tumor cell inoculation. Each group consisted of to mice. Tumor
`size was determined at weekly intervals. Four weeks after tumor cell
`inoculation. animals were euthanized. and the tumors were removed and
`weighed. Statistical analysis was performed by ANOVA.
`
`RESULTS
`
`culture. After several passages. the suspension-adapted cells were used to
`inoculate production cultures in GHT-containing. serum-free media sup-
`plemented with various hormones and protein hydrolysates. Harvested cell
`culture fluid containing rhuMAb VEGF was purified using protein A-
`Sepharose CL-4B. The purity after this step was ~99%. Subsequent
`purification to homogeneity was carried out using an ion exchange chro-
`matography step. The endotoxin content of the final purified antibody was
`<0.IO eu/mg.
`F(ab) and IgG Quantitation. For quantitating F(ab) molecules. ELISA
`plates were coated with 2 [Lg/ml of goat anti-human lgG Fab (Organon
`Teknika. Durham. NC) in 50 mM carbonate buffer. pH 9.6. at 4°C overnight
`and blocked with PBS-0.5% BSA (blocking buffer) at room temperature for
`I h. Standards [0.78—50 ng/ml human F(ab)] were purchased from Chemicon
`(Temecula. CA). Serial dilutions of samples in PBS-0.5% BSA-0.05% poly-
`sorbate 20 (assay buffer) were incubated on the plates for 2 h. Bound F(ab) was
`detected using horseradish peroxidase-labeled goat anti-human lgG F(ab)
`(Organon Teknika), followed by 3.3’,5.5'-tetramethylbenzidine (Kirkegaard &
`Perry Laboratories. Gaithersburg. MD) as the substrate. Plates were washed
`between steps. Absorbance was read at 450 nm on a VM plate reader
`(Molecular Devices. Menlo Park. CA). The standard curve was fit using a
`four—parameter nonlinear
`regression curve-fitting program developed at
`Genentech. Data points that fell in the range of the standard curve were used
`for calculating the F(ab) concentrations of samples.
`The concentration of full-length antibody was determined using goat anti-
`human IgG Fe (Cappel. Westchester. PA) for capture and horseradish perox-
`idase-labeled goat anti-human Fc (Cappel) for detection. Human IgGl (Chemi-
`con) was used as standard.
`VEGF Binding Assays. For measuring the VEGF binding activity of
`F(ab)s, ELISA plates were coated with 2 ug/ml rabbit F(ab’)2 to human
`IgG Fc (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 VEGF for l h in tubes.
`The solutions from the tubes were then transferred to the ELISA plates and
`incubated for l 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
`absorbance of the titration curve of the standard were calculated and then
`
`divided by the concentration of the standard corresponding to the midpoint
`absorbance of the standard titration curve. Assays for full-length IgG were
`the same as for the F(ab)s except that the assay buffer contained l0%
`human serum.
`
`Humanization. The consensus sequence for the human heavy
`chain subgroup III and the light chain subgroup K I were used as the
`framework for the humanization (Ref. 30; Fig. I). This framework has
`been successfully used in the humanization of other murine antibodies
`(3l. 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).
`Two definitions of CDR residues have been proposed. One is based
`on sequence hypervariability (30) and the other on crysz 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-HI; CDR—H1 includes residues H3I-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. VEGF binding of the humanized and chimeric
`with H). We. therefore, defined CDR-HI as a combination of the two,
`F(ab)s were compared using a BlAcore biosensor (4l). Concentrations of
`i.e.. including residues H26 —H35. The other CDRs were 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) was used as the standard in the binding assays.
`manufacturer‘s instructions (Pharmacia). Off-rate kinetics were measured by
`In the initial variant. F(ab)-l, the CDR residues were transferred from
`saturating the chip with F(ab) [35 p.l of 2 uM F(ab) at a flow rate of 20 ul/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 5 were used for off-rate kinetic analysis. The dissociation rate constant
`H49 (Ala in humans) was changed to the murine Gly. In addition,
`(km) was obtained from the slope of the plot of ln(R0/R) versus time, where R0
`F(ab)s that consisted of the chimeric heavy chain/F(ab)-l light chain
`is the signal at t = 0 and R is the signal at each time point.
`On-rate kinetics were measured using 2-fold serial dilutions of F(ab)
`[F(ab)-2] and F(ab)-l heavy chain/chimeric light chain [F(ab)-3] were
`(0.0625—2 mM). The slope. K,. was obtained from the plot of ln(-dR/dt)
`generated and tested for binding. F(ab)-I exhibited a binding affinity
`versus time for each F(ab) concentration using the BlAcore kinetics eval—
`greater than lOOO-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
`signal at time I. Data between 80 and I68. I48. l28. I I4, 102. and 92 s were
`that framework residues in the F(ab)-l VH domain needed to be
`used for 0.0625. 0.125. 0.25. 0.5. l, and 2 mM F(ab). respectively. The
`altered to increase binding.
`association rate constant (km) was obtained from the slope of the plot of K5
`Previous humanizations (3|. 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 shown that residues H7]
`removed by injecting 5 pl of 50 mM HCI at a flow rate of 20 ul/min to
`and H73 can have a profound effect on binding. possibly by influ-
`regenerate the chip.
`encing the conformations of CDR-HI and CDR-H2. Changing the
`Endothelial Cell Growth Assay. Bovine adrenal cortex-derived capillary
`endothelial cells were cultured in the presence of low glucose DMEM (Life
`human residues to their murine counterparts in F(ab)-4 improved
`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
`
`

`

`HUMANIZATION OF AN ANTI-VEGF MONOCLONAL ANTIBODY
`
`Variable Heavy
`
`A. 4 . 6 . 1
`
`P (ab) - 12
`
`humI I I
`
`E IQLVQSGPELKQPGETVRISCKASGXIETNXGMNWVKQAPGKGLKWMG
`t
`t
`* I t
`t i k i
`*
`fl *
`EVOLVESGGGLVQPGGSLRLSCAASWMNWVRQAP GKGLEWVG
`i
`t i
`t
`t
`EVOLVESGGGLVQPGGSLRLSCAASGE‘TFSS YAMSWVRQAPGKGLEWVS
`1
`1 0
`2 0
`3 0
`4 0
`
`A. 4 . 6 . 1 WTFSLETSASTAYLQISNLKNDDTATYFCAK
`* t i R t 'k
`i i
`P (ab) - 12 WTFSLDTSKSTAYLQMNSLRAEDTAVYYCAK
`* ttti it!
`it! t
`i i
`'k
`t *
`v1 SGDGGs'rvaDSVKGRF'rIsanNSKN'rLYLmiNSLRAEDTAWXCAR
`5 0
`a
`60
`7 0
`8 0
`abc
`90
`
`humI I I
`
`A - 4 - 6 - 1 WGAGTTVTVSS
`i
`*
`F (ab) -12 WGQGTLVTVSS
`*
`t
`G——————————FDYWGQGTLVTVSS
`1 10
`
`humI I I
`
`A. 4 . 6 . 1
`
`Variable Light
`DIQMTQTTSSLSASLGDRVIISCSASQQISWWYQQKPDGTVKVLIY
`* i
`t
`i *
`* i t i
`F (ab) — 12 DIQMTQSPSSLSASVGDRVT ITCWQQKPGKAPKVLIY
`t
`t
`t
`*
`
`humKI
`
`DIQMTQSPSSLSASVGDRVTITCRASQSISNYLAWYQQKPGKAPKLLIY
`1
`1 o
`20
`30
`4 o
`
`A. 4 . 6 . 1
`
`EISSLHSGVPSRFSGSGSGTDYSLTISNLEPEDIATvvcmxammF
`I t
`t t
`t
`
`* i
`t
`“1:
`F (ab) — 12 WGVPSRFSGSGSGTDFTLTISSLQPEDE‘ATYYCQQXSMEIFy
`humKI
`AASSLESGVPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQYNSLPWTF
`5 0
`60
`7 0
`8 0
`90
`
`A.4.6.1
`
`E‘ (ab) -12
`
`GGGTKLEIKR
`t
`t
`GQGTKVEIKR
`
`humKI
`
`GQGTKVE IKR
`1 00
`
`Fig. 1. Amino acid sequence of variable heavy and light domains of muMAbVEGF
`A.4.6.l, humanized F(ab) with optimal VEGF binding [F(ab)-12] and human consensus
`frameworks (humlll. heavy subgroup [I]; humxl, light K subgroup I). Asterisks. differ-
`ences between humanized F(ab)-12 and the murine MAb or between F(ab)-12 and the
`human framework. CDRs are underlined.
`
`and humanized F(ab)s suggested that residue L46, buried at the
`VL—VH interface and interacting with CDR-H3 (Fig. 2), might also
`play a role either in determining the conformation of CDR-H3
`and/or affecting the relationship of the VL and VH domains. When
`the murine Val was exchanged for the human Leu at L46 [F(ab)-5],
`the binding affinity increased by almost 4-fold (Table 1). Three
`other buried framework residues were evaluated based on the
`
`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), and this 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 BIAcore system (Phannacia). Using this
`technique, the Kd of the humanized F(ab)-12 was 2-fold weaker than
`that of the chimeric F(ab) due to both a slower kon and faster koff
`(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 K light chain and human IgG1 heavy chain. The
`full-length 12-IgGl [F(ab)—12 fused to human IgGl] exhibited bind-
`ing that was 1.7-fold weaker than the chimeric IgGl (Table 3). Both
`lZ-IgGl and the chimeric lgGl bound slightly less well than the
`original muMAb VEGF A.4.6.1 (Table 3).
`Biological Studios. rhuMAb VEGF and muMAb VEGF A.4.6.l
`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 MAbs were found to be essentially equivalent, both in potency
`and efficacy. The EDsos were, respectively, 50 t 5 and 48 t 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
`VEGF had any effect on basal or bFGF-stimulated proliferation of
`capillary endothelial cells (data not shown), confirming that the
`inhibition is specific for VEGF.
`To determine whether similar 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 rhabdomyosarcoma cells in nude
`mice. Previous studies have shown that muMAb VEGF A.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 dose in the animals treated with muMAb VEGF A.4.6.l versus
`90 and 95% in those treated with rhuMAb VEGF. Similar results were
`obtained with the breast carcinoma cell line MDA-MB 435 (data not
`shown).
`
`DISCUSSION
`
`The murine MAb A.4.6.1, directed against human VEGF (42),
`was humanized using the same consensus frameworks for the light
`and heavy chains used in previous humanizations (31, 32, 43, 44),
`i.e., VKI and VHIII (30). Simply transferring the CDRs from the
`murine antibody to the human framework resulted in a F(ab) that
`exhibited binding to VEGF reduced by over lOOO-fold compared to
`the parent murine antibody. Seven non-CDR, framework residues
`in the VH domain and one in the VL domain were altered from
`
`molecular models: H49, H69, and H78. Position H69 may affect
`the conformation of CDR-H2, whereas position H78 may affect the
`conformation of CDR-H1 (Fig. 2). When each was individually
`changed from the human to murine counterpart, the binding im-
`human to murine to achieve binding equivalent
`proved by 2-fold in each case [F(ab)-6 and F(ab)-7; Table 1]. When
`murine antibody.
`both were simultaneously changed, the improvement in binding
`In the VH domain, residues at positions H49, H69, H71, and H78
`was 8—fold [F(ab)-8; Table 1]. Residue H49 was originally 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;
`Table 1].
`H76 should be solvent exposed (Fig. 2) and hence may interact
`directly with the VEGF; these two residues are in a non-CDR loop
`We have found during previous humanizations that residues in a
`adjacent to CDRs H1 and H2 and have been shown to play a 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)-l 1, two residues in this loop
`were changed to their murine counterparts: Aan76 to murine Ser
`for lysine at position H94 was surprising given that this residue is
`[F(ab)-10] and LysH75 to murine Ala [F(ab)-ll]. Both effected a
`arginine in the human framework (Fig. 1). In some crystal struc-
`relatively small improvement in binding (Table 1). Finally, at position
`tures of F(ab)s, ArgH94 forms a hydrogen-bonded salt-bridge with
`4596
`
`to the parent
`
`

`

`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 [/46 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 ofanli-VEGF F(ab) variants to VEGF using the BlAcare system"
`Amount of (Fab)
`Kd (m)
`km (M_'s_')
`1:0,Y (s-l)
`bound (RU)
`Variant
`0.91
`6.5 x 10“
`5.9 x 10'5
`4250
`chain-F(ab)"
`l.8
`3.5 x 10“
`6.3 x to"
`3740
`F(ab)—12
`" The amount of F(ab) bound. in resonance units (RU). was measured using a BlAcore
`system when 2 pg F(ab) was injected onto a chip containing 2480 RU of immobilized
`VEGF. Off-rate kinetics (km) were measured by saturating the chip with F(ab) and then
`monitoring dissociation after switching to buffer. On-rate kinetics (Iron) were measured
`using 2-fold serial dilutions of F(ab). Kd. the equilibrium dissociation constant. was
`calculated as knnlkm.
`b chim-F(ab) is a chimeric F(ab) with murine VL and VH domains fused to human CL
`and CHI heavy domains.
`
`AsleOl (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-Vl-l
`interface. where it is buried and interacts directly with CDR-H3 (Fig.
`2). The requirement for murine valine (as opposed to human leucine)
`implies that this residue plays an important role in the conformation of
`CDR-H3. The necessity of retaining LysH94 in VH, which is also
`4597
`
`adjacent to CDR-H3, suggests that CDR-H3 plays a major role in the
`binding of the antibody to VEGF.
`The humanized version with optimal binding, lZ-lgGl, 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

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