Title of Invention

ANTIBODIES AGAINST IL-13 RECEPTOR ALPHA 1 AND USES THEREOF

Abstract An antibody binding to IL-13Ral, inhibiting IL-13 bioactivity and comprising a variable heavy and a variable light chain, characterized in that the variable heavy chain amino acid sequence CDR3 of said antibody is selected from the group consisting of the heavy chain CDR3 sequences of SEQ ID NO: 1, 3, 5, 7 or 9 is useful in the treatment of asthma and allergic diseases.
Full Text

Antibodies against IL-13 receptor alphal and uses thereof
The present invention relates to human antibodies against IL-13 receptor alphal (IL-13Ra (alpha)l), methods for their production, and uses.
Background of the Invention
IL-13 is a secreted monomelic peptide produced mainly by Th2 cells but also by mast cells and NK cells. Biological functions of IL-13 include regulation of IgE production and modulation of Th2 development. IL-13 binds to a receptor complex consisting of IL-13 receptor alphal (IL-13Ral) chain and IL-4 receptor alpha (IL-4Ra) chain. IL-13 binding triggers signal transduction events mainly through STAT6. IL-13 binds with low affinity to the IL-13 Ral alone and does not bind to IL-4Ral. Contrary to this, IL-4 binds to lL-4Ra alone and does not bind to IL-13Ral alone. Another receptor for IL-13 has been described, the IL-13Ra2. IL-13 binds with high affinity to this receptor. Likely this receptor acts as a decoy receptor.
Inducible overexpression of IL-13 in transgenic mice results in a phenotype that shares many characteristics with asthmatic patients. They show mucus metaplasia, macrophage, lymphocyte and eosinophil-rich inflammation, upregulation of proteases like MMP-9, -12, -13, -2 and -14, cathepsin B, H, K and S and they also present subepithelial fibrosis. Knockout mice for IL-13 show a significant reduction in Th2 cytokine production due to impairment in Th2 development. These mice do not develop airway hyperreactivity (AHR) in spite of the presence of eosinophil inflammation. The AHR was restored by administration of IL-13, indicating that IL-13 is necessary and sufficient for the induction of AHR in mouse. Other important biological functions of IL-13 in relationship with asthma include the induction of goblet cell metaplasia and mucus production. It acts directly on airway epithelial cells, fibroblasts and airway smooth muscle cells and induces different transcriptional programs in each of this cell types. Interestingly, IL-13 decreases the alpha-adrenergic response in smooth muscle cells, contributing to airway narrowing. IL-13 promoter polymorphism is associated with increased risk of allergic asthma. Polymorphisms in the IL-13 gene are associated with high serum IgE levels. Single nucleotide polymorphism in the intergene sequence between the IL-4 and IL-13 genes is associated with atopic asthma.

JL-13 antagonists have been utilized in animal models. For example a soluble mouse IL-13Ra2-IgGFc fusion protein has been used to show efficacy in completely reversing ovalbumin-induced AHR and the number of mucus containing cells. The reversal was obtained even if the treatment is given after full development of the phenotype. In addition, treatment of mice with an IL-13 fusion cytotoxin molecule resulted in reduction of all features of airway disease in a chronic fungal-induced allergic inflammation. In conclusion, IL-13 is a critical mediator of the effector arm of the allergic response.
IL-13Ral is a member of the hemapoietin receptor superfamily (type 1 cytokine receptor family) and identified and described by Obiri N. I., et ah, J. Biol. Chem., 270 (1995) 8797-8804) and WO 96/29417. It is a protein of 427 amino acids including the signal sequence. Its DNA and protein sequences are described in WO 97/15663 and SwissProt No. P78552. IL-13Ral is a glycosylated protein binding to IL-13 with low affinity, but, when linked with IL-4Ra to a heterodimer, it binds IL-13 with high affinity. This complex is also a receptor for IL-4.
Antibodies against IL-13Ral are known from WO 96/29417, WO 97/15663, WO 03/080675, Graber P., et al., Eur. J. Immunol., 28 (1998) 4286-4298; Poudrier J., et al., J. Immunol., 163 (1999) 1153-1161; Poudrier J., et al., Eur. J. Immunol., 30 (2000) 3157-3164; Aikawa M., et al., Cytokine, 13 (2001) 75-84. Antibodies against IL-13Ral are commercially available from R&D Systems Inc. USA.
Summary of the Invention
The invention comprises an antibody binding to IL-13Ral and inhibiting IL-13 bioactivity, characterized in that the variable heavy chain amino acid sequence CDR3 of said antibody is selected from the group consisting of the heavy chain CDR3 sequences of SEQ ID NO: 1, 3, 5, 7 or 9.
The antibody is preferably a human antibody.
The antibody is preferably characterized by an affinity of 10~9 M (KD) or less, preferably of 10"9 to 1013 M for binding to IL-13Ral.
Preferably the antibody is characterized in that its heavy chain CDR1, CDR2 and CDR3 sequences are selected from the group consisting of the heavy chain CDR1, CDR2 and CDR3 sequences of SEQ ID NO: 1, 3, 5, 7 or 9.

The antibody is preferably characterized in that the variable light chain amino acid sequences CDR1, CDR2 and CDR3 of said antibody are selected from the group consisting of the light chain CDR sequences of SEQ ID NO: 2, 4, 6, 8 or 10.
The antibody is preferably characterized in that the variable heavy chain amino acid sequences CDR1, CDR2 and CDR3 of said antibody are selected from the group consisting of the heavy chain CDR sequences of SEQ ID NO: 1, 3, 5, 7 or 9 and the variable light chain amino acid sequences CDR1, CDR2 and CDR3, of said antibody are selected from the group consisting of the light chain CDR sequences of SEQ ID NO: 2, 4, 6, 8 or 10.
The CDR sequences are preferably selected independently of each other and are separated by FR (framework) regions.
The antibody is preferably characterized in comprising as heavy chain CDRs the CDRs of SEQ ID NO: 1 and as light chain CDRs the CDRs of SEQ ID NO: 2, as heavy chain CDRs the CDRs of SEQ ID NO: 3 and as light chain CDRs the CDRs of SEQ ID NO: 4, as heavy chain CDRs the CDRs of SEQ ID NO: 5 and as light chain CDRs the CDRs of SEQ ID NO: 6, as heavy chain CDRs the CDRs of SEQ ID NO: 7 and as light chain CDRs the CDRs of SEQ ID NO: 8 or as heavy chain CDRs the CDRs of SEQ ID NO: 9 and as light chain CDRs the CDRs of SEQ ID NO: 10.
The CDR sequences can be determined according to the standard definition of Kabat et ah, Sequences of Proteins of Immunological Interest, 5th ed., Public Health Service, National Institutes of Health, Bethesda, MD (1991). On this basis, the complementarity determining regions (CDRs) of SEQ ID NO: 1-8 have the following sequences:
Heavy chain CDRs: CDR1 (aa 31-35) of SEQ ID NO: 1, 3, 5, 7, 9, CDR2 (aa 50-66) of SEQ ID NO: 1, 3, 5, 7, 9, CDR3 (aa 99-108) of SEQ ID NO: 1, 3, 9, CDR3 (aa 99-107) of SEQ ID NO: 5, CDR3 (aa 99-112) of SEQ ID NO: 7;
Light chain CDRs: CDR1 (aa 24-34 ) of SEQ ID NO: 2, 4, 6, 10, CDR1 (aa 24-35 ) of SEQ ID NO: 8, CDR2 (aa 50-56) of SEQ ID NO: 2, 4, 6, 10, CDR2 (aa51-57)ofSEQIDNO:8 and CDR3 (aa 89-97) of SEQ ID NO: 2, 4, 6, 10, CDR3 (aa 90-97) of SEQ ID NO: 8.

Preferably, the invention provides an antibody comprising as complementarity determining regions (CDRs) the following sequences:
a) an antibody heavy chain comprising heavy chain CDRs of SEQ ID NO:l, 3, 5, 7 or 9;
b) an antibody light chain comprising light chain CDRs of SEQ ID NO:2, 4, 6, 8 or 10,
wherein the CDRs are selected independently of each other.
The antibody is preferably characterized in comprising as heavy chain variable region SEQ ID NO: 1 and as light chain variable region SEQ ID NO: 2, as heavy chain variable region SEQ ID NO: 3 and as light chain variable region of SEQ ID NO: 4, as heavy chain variable region SEQ ID NO: 5 and as light chain variable region SEQ ID NO: 6, as heavy chain variable region SEQ ID NO: 7 and as light chain variable region SEQ ID NO: 8 or as heavy variable region SEQ ID NO: 9 and as light chain variable region SEQ ID NO: 10.
The antibody is preferably characterized in comprising
a) as heavy chain variable region SEQ ID NO: 1, as light chain variable region SEQ ID NO: 2, as K light chain constant region SEQ ID NO: 11 and as yl heavy chain constant region SEQ ID NO: 12 optionally with mutations L234A and L235A or D265A and N297A,
b) as heavy chain variable region SEQ ID NO: 3 and as light chain variable region of SEQ ID NO: 4, as K light chain constant region SEQ ID NO: 11 and as yl heavy chain constant region SEQ ID NO: 12 optionally with mutations L234A and L235A or D265A and N297A,
c) as heavy chain variable region SEQ ID NO: 5 and as light chain variable region SEQ ID NO: 6, as K light chain constant region SEQ ID NO: 11 and as yl heavy chain constant region SEQ ID NO: 12 optionally with mutations L234A and L235A or D265A and N297A,
d) as heavy chain variable region SEQ ID NO: 7 and as light chain variable region SEQ ID NO: 8, as K light chain constant region SEQ ID NO: 11 and as yl heavy chain constant region SEQ ID NO: 12 optionally with mutations L234A and L235A or D265A and N297A, or
e) as heavy variable region SEQ ID NO: 9 and as light chain variable region SEQ ID NO: 10, as K light chain constant region SEQ ID NO: 11 and as yl

heavy chain constant region SEQ ID NO: 12 optionally with mutations L234A and L235A or D265A and N297A.
Preferably the antibody is characterized in binding to IL-13Ral in competition to antibody LC5002-002, LC5002-003, LC5002-005, LC5002-007 and/or LC5002-018. Preferably the antibody is characterized in comprising as variable regions the variable regions of LC5002-002, LC5002-003, LC5002-005, LC5002-007 or LC5002-018. The variable regions of these antibodies are shown in SEQ ID NO: 1-10. Useful constant regions are well known in the state of the art. Examples are shown in SEQ ID NO: 11-12.
The antibody is preferably a monoclonal or a recombinantly produced antibody.
In one embodiment of the invention the antibody is a class-altered human antibody.
In a preferred embodiment of the invention the antibody contains a human yl heavy chain comprising
a) amino acid sequence Pro233Val234Ala235 with deletion of Gly236 and/or amino acid sequence Gly327Leu32sPro329Ser33oSer33i ,
b) amino acid sequence Ala234Ala235 or
c) amino acids Ala265 and Ala297-
Preferably the antibody according to the invention inhibits IL-13 induced Stat-6 phosphorylation with an IC50 value of 6 nM or lower, inhibits IL-13 induced eotaxin production with an IC50 value of 20 nM or lower and/or inhibits IL-13 or IL-4 induced cell proliferation, preferably of TF-1 cells (ATCC CRL 2003) with an IC50 value of 10 nM or lower (IL-13) and 60 nM or lower (IL-4). IL-13 induced Stat-6 phosphorylation, eotaxin production and induction of cell proliferation are determined according to examples 6 to 8.
The antibody according to the invention preferably does not bind to denatured IL-13R(X1(KD for binding affinity 10"6 M or higher). The antibody is preferably characterized by showing substantially no crossreactivity with IL-13Ra2 and IL-4Ra (KD for binding affinity 10"6 M or higher).

The invention further provides hybridoma cell lines which produce antagonistic monoclonal antibodies against IL-13Ral .
The preferred hybridoma cell lines according to the invention (hu-MABLC.5002-002 (DSM ACC2709), hu-MABLC.5002-003 (DSM ACC2710), hu-MABLC.5002-005 (DSM ACC2711), hu-MABLC.5002-007 (DSM ACC2712)) were deposited 13.01.2005 with Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH (DSMZ), Germany.
The antibodies obtainable from said cell lines are embodiments of the invention.
The invention further provides nucleic acids encoding polypeptides of which antibodies according to the invention are comprised, expression vectors comprising said nucleic acids, and host cells for the recombinant production of such antibodies. The invention further provides methods for the recombinant production of such antibodies.
The polypeptides encoded by the nucleic acids according to the invention are
a) an antibody heavy chain comprising heavy chain CDRs of SEQ ID NO: NO:l,3, 5, 7 or 9 and
b) an antibody light chain comprising light chain CDRs of SEQ ID NO: 2, 4, 6, 8 or 10.
These polypeptides are capable of assembling together with the respective other antibody chain to generate an antibody.
Antibodies according to the invention show benefits for patients in need of corticosteroid therapy. The antibodies according to the invention have new and inventive properties causing a benefit for a patient suffering from asthma or an allergic disease.
The invention further provides methods for treating asthma and allergic diseases.
The invention further comprises the use of an antibody according to the invention for asthma treatment and for the manufacture of a pharmaceutical composition according to the invention. In addition, the invention comprises a method for the manufacture of a pharmaceutical composition according to the invention.

The invention further comprises a pharmaceutical composition comprising an antibody according to the invention with a pharmaceutically effective amount, optionally together with a buffer and/or an adjuvant useful for the formulation of antibodies for pharmaceutical purposes.
The invention further provides pharmaceutical compositions comprising such antibodies in a pharmaceutically acceptable carrier. In one embodiment, the pharmaceutical composition may be included in an article of manufacture or kit.
The invention further comprises a vector comprising a nucleic acid according to the invention, capable of expressing said nucleic acid in a prokaryotic or eukaryotic host cell.
The invention further comprises a prokaryotic or eukaryotic host cell comprising a vector according to the invention.
The invention further comprises a method for the production of a recombinant human antibody according to the invention, characterized by expressing a nucleic acid according to the invention in a prokaryotic or eukaryotic host cell and recovering said antibody from said cell. The invention further comprises the antibody obtainable by such a recombinant method.
The invention further comprises a method for the preparation of a pharmaceutical composition characterized in selecting an antibody against IL-13Ral from a plurality of antibodies against IL-13Ral when compared to such an assay without said antibody, producing said antibody by means of recombinant expression, recovering said antibody and combining said antibody with a pharmaceutical acceptable buffer and/or adjuvant Preferably the antibody has one or more of the above mentioned additional properties.
Detailed Description of the Invention
The terms "IL-13Ral, murine IL-13Ral, IL-13, IL-13Ra2 and IL-4Ra" and their domains are well known in the state of the art and e.g. defined by SwissProt P78552, G09030, P35225, Q14627 and P24394. If not otherwise stated, the terms "IL-13Ral, IL-13, IL-13Ra2 and IL-4Ra" therefore denotes the human polypeptides IL-13Ral, IL-13, IL-13Ra2 and IL-4Ra.

The term "human antibody", as used herein, includes antibodies having variable and constant regions (domains) which can be assigned to defined human germ line immunoglobulin sequences because of their high sequence similarity or identity with these germ line sequences. Human antibodies are well-known in the state of the art (van Dijk, M.A., and van de Winkel, J.G., Curr. Opin. Chem. Biol. 5 (2001) 368-374). Human antibodies can also be produced in transgenic animals (e.g. mice) that are capable, upon immunization, of producing a full repertoire or a selection of human antibodies in the absence of endogenous immunoglobulin production. Transfer of the human germ-line immunoglobulin gene array in such germ-line mutant mice results in the production of human antibodies upon antigen challenge (see, e.g., Jakobovits, A., et al., Proc. Natl. Acad. Sci. USA 90 (1993) 2551-2555; Jakobovits, A., et al., Nature 362 (1993) 255-258; Bruggemann, M., et al., Year Immunol. 7 (1993) 33-40). Human antibodies can also be produced in phage display libraries (Hoogenboom, H.R., and Winter, G., J. Mol. Biol. 227 (1992) 381-388; Marks, J.D., et al., J. Mol. Biol. 222 (1991) 581-597). The techniques of Cole et al. and Boerner et al. are also available for the preparation of human monoclonal antibodies (Cole et al., Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, p. 77 (1985); and Boerner, P., et al., J. Immunol. 147 (1991) 86-95). A human antibody encompasses the various forms of antibodies, preferably monoclonal antibodies including but not being limited to whole antibodies, antibody fragments, class-altered antibodies and genetically engineered antibodies (variant or mutant antibodies) as long as the characteristic properties according to the invention are retained. Especially preferred are recombinant human antibodies. The term "monoclonal antibody" as used herein refers to a preparation of antibody molecules all having substantially the same amino acid sequence.
The term "recombinant human antibody", as used herein, is intended to include all human antibodies that are prepared, expressed, created or isolated by recombinant means, such as antibodies isolated from a host cell such as a NSO or CHO cell or from an animal (e.g. a mouse) that is transgenic for human immunoglobulin genes or antibodies expressed using a recombinant expression vector transfected into such a host cell. Such recombinant human antibodies have variable and constant regions in a rearranged form. The recombinant human antibodies according to the invention have been subjected to in vivo somatic hypermutation. Thus, the amino acid sequences of the VH and VL regions of the recombinant antibodies are sequences that can be assigned to defined human germ line VH and VL sequences, but may not naturally exist within the human antibody germ line repertoire in vivo.

The term "class-altered antibody" refers lo a monoclonal antibody, preferably a human antibody, comprising a variable region, i.e., binding region, from one source or germ line and at least a portion of a constant region that matches a constant region of an antibody from a different source or germ line, usually prepared by recombinant DNA techniques. Such class-altered antibodies are not naturally occurring and therefore not available directly from xenograft mice. Forms of "class-altered antibodies" encompassed by the present invention are those in which the constant region has differences from the wild-type constant region sequence that result in an antibody having different properties according to the invention, especially in regard to Clq binding and/or Fc receptor (FcR) binding, i.e. by change or mutation of Fc. Class-altered antibodies are the product of expressed immunoglobulin genes comprising DNA segments encoding immunoglobulin variable regions and DNA segments encoding immunoglobulin constant regions. Methods for producing class-altered antibodies involve conventional recombinant DNA and gene transfection techniques are well known in the art (see, e.g., Morrison, S.L., et ah, Proc. Natl. Acad. Sci. USA 81 (1984) 6851-6855; US Patent Nos. 5,202,238 and 5,204,244).
The "variable region" (variable region of a light chain (VL), variable region of a heavy chain (VH)) as used herein denotes the part of each of the pair of light and heavy chains which is involved directly in binding the antibody to the antigen. The domains of variable human light and heavy chains have the same general structure and each domain comprises four framework (FR) regions whose sequences are widely conserved, connected by three "hypervariable regions" (or complementarity determining regions, CDRs). The framework regions adopt a P(beta)-sheet conformation and the CDRs may form loops connecting the P-sheet structure. The CDRs in each chain are held in their three-dimensional structure by the framework regions and form together with the CDRs from the other chain the antigen binding site. The antibody heavy and light chain CDR3 regions, preferably the heavy chain CDR3, play a particularly important role in the binding specificity/affinity of the antibodies according to the invention and therefore provide a further object of the invention.
The terms "hypervariable region" or "antigen-binding portion of an antibody" when used herein refer to the amino acid residues of an antibody which are responsible for antigen-binding. The hypervariable region comprises amino acid residues from the "complementarity determining regions" or "CDRs". "Framework" or "FR" regions are those variable domain regions other than the hypervariable region

residues as herein defined. Therefore, the light and heavy chains of an antibody comprise from N- to C-terminus the domains FR1, CDR1, FR2, CDR2, FR3, CDR3, and FR4. CDR and FR regions are determined according to the standard definition of Kabat et al., Sequences of Proteins of Immunological Interest, 5th ed., Public Health Service, National Institutes of Health, Bethesda, MD (1991).
The "constant domains" are not involved directly in binding of an antibody to an antigen, but exhibit various effector functions. Depending on the amino acid sequence of the constant region of their heavy chains, antibodies or immunoglobulins are divided into the classes: IgA, IgD, IgE, IgG and IgM, and several of these may be further divided into subclasses (isotypes), e.g. IgGl, IgG2, IgG3, and IgG4, IgAl and IgA2. The heavy chain constant regions that correspond to the different classes of immunoglobulins are called m 5, y, a, and e, respectively. The antibodies according to the invention are preferably of IgGl type.
The Fc part of an antibody is directly involved in complement activation, Clq binding, C3 activation and Fc receptor binding. Binding to Clq is caused by defined binding sites in the Fc part. Such binding sites are known in the state of the art and described e.g. by Lukas, T.J., et al., J. Immunol. 127 (1981) 2555-2560; Brunhouse, R., and Cebra, J.J., Mol. Immunol. 16 (1979) 907-917; Burton, D.R., et al., Nature 288 (1980) 338-344; Thommesen, J.E., et al., Mol. Immunol. 37 (2000) 995-1004; Idusogie, E.E., et al., J. Immunol. 164 (2000) 4178-4184; Hezareh, M., et al., J. Virol. 75 (2001) 12161-12168; Morgan, A., et al., Immunology 86 (1995) 319-324; and EP 0 307 434. Such binding sites are e.g. L234, L235, D270, N297, E318, K320, K322, P331 and P329 (numbering according to EU index of Kabat, see below). Antibodies of subclass IgGl, IgG2 and IgG3 usually show complement activation, C1 q binding and C3 activation, whereas IgG4 antibodies do not activate the complement system, do not bind Clq and do not activate C3. As used herein the term "Fc part derived from human origin" denotes a Fc part which preferably has an amino acid sequence of a Fc part of a human antibody of the subclass IgGl modified in such a way that no Clq binding, C3 activation and/or FcR binding can be detected or binding is at least reduced for 50%, preferably 70%, compared to a human IgGl antibody. An "Fc part of an antibody" is a term well known to the skilled artisan and defined on the basis of papain cleavage of antibodies. The antibodies according to the invention contain as Fc part, preferably with an amino acid sequence of a Fc part derived from human origin and preferably all other parts of the human constant regions. Preferably the Fc part is a mutated human Fc part

from human IgGl subclass. Mostly preferred are Fc parts comprising a yl-heavy chain constant region (an example is shown in SEQ ID NO: 11) with mutations L234A and L235A or D265A and N297A (W099/51642).
Human constant chains, e.g. yl-heavy chains are described in detail by Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD. (1991), and by Briiggemann, M., et al., J. Exp. Med. 166 (1987) 1351-1361; Love, T.W., et al., Methods Enzymol. 178 (1989) 515-527. The constant domains preferred in the invention provide no complement binding. The "variable region" (variable region of a light chain (VL), variable region of a heavy chain (VH)) as used herein denotes each of the pair of light and heavy chains which is involved directly in binding the antibody to the antigen.
The term nucleic acid or nucleic acid molecule, as used herein, is intended to include DNA molecules and RNA molecules. A nucleic acid molecule may be single-stranded or double-stranded, but preferably is double-stranded DNA.
A nucleic acid is "operably linked" when it is placed into a functional relationship with another nucleic acid sequence. For example, DNA for a presequence or secretory leader is operably linked to DNA for a polypeptide if it is expressed as a preprotein that participates in the secretion of the polypeptide; a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the sequence; or a ribosome binding site is operably linked to a coding sequence if it is positioned so as to facilitate translation. Generally, "operably linked" means that the DNA sequences being linked are cis, and, in the case of a secretory leader, contiguous and in reading frame. However, enhancers do not have to be contiguous. Linking is accomplished by ligation at convenient restriction sites. If such sites do not exist, the synthetic oligonucleotide adaptors or linkers are used in accordance with conventional practice.
As used herein, the expressions "cell," "cell line," and "cell culture" are used interchangeably and all such designations include progeny. Thus, the words "transformants" and "transformed cells" include the primary subject cell and cultures derived therefrom without regard for the number of transfers. It is also understood that all progeny may not be precisely identical in DNA content, due to deliberate or inadvertent mutations. Variant progeny that have the same function or

biological activity as screened for in the originally transformed cell are included. Where distinct designations are intended, it will be clear from the context.
The term "binding to IL-13Raln as used herein means the binding of the antibody to IL-13Ral in an in vitro assay, preferably in a binding assay in which the antibody is bound to a surface and binding of IL-13Ral is measured by Surface Plasmon Resonance (SPR). Binding means a binding affinity (KD) of 10" M or less, preferably 10"13 to 10"9 M. "No binding" means a KDof 10"6 M or more. The antibodies according to the invention bind to the extracellular domain of human IL-13Ral and preferably also of mouse IL-13Ral.
Binding to IL-13Ral can be investigated by a BIAcore assay (Pharmacia Biosensor AB, Uppsala, Sweden). The affinity of the binding is defined by the terms ka (rate constant for the association of the antibody from the antibody/antigen complex), kd (dissociation rate), and KD (kd/ka).
The binding of 1L-13 to IL-13Ral is inhibited by the antibodies according to the invention. The inhibition is measured as IC50 in an ELISA for binding of IL-13 to IL-13Ral/IL-4Ra heterodimer. For performing such an assay lL-13Ral is immobilized and IL-13 and IL-4Ra are added. The IC50 values of the antibodies according to the invention for the binding of IL-13 to IL-13Ral are no more than 6 nM. IC50 values are measured as average or median values of at least three independent measurements. Single IC50 values may be out of the scope.
The antibodies according to the invention show preferably a binding to the same epitopes of IL-13Ral as an antibody selected from the group consisting of antibodies LC5002-002, LC5002-003, LC5002-005, LC5002-007 or LC5002-018 or are inhibited in binding to IL~13Rctl due to steric hindrance of binding by these antibodies. Binding inhibition can be detected by an SPR assay using an immobilized antibody selected from the group consisting of antibodies LC5002-002, LC5002-003, LC5002-005, LC5002-007 or LC5002-018 and IL-13Ral at a concentration of 20-50 nM and the antibody to be detected at a concentration of 100 nM. A signal reduction of 50% or more shows that the antibody competes with an antibody selected from the group consisting of antibodies LC5002-002, LC5002-003, LC5002-005, LC5002-007 or LC5002-018. The term "epitope" means a protein determinant capable of specific binding to an antibody. Epitopes usually consist of chemically active surface groupings of molecules such as amino acids or sugar side chains and usually have specific three dimensional structural

characteristics, as well as specific charge characteristics. Conformational and nonconformational epitopes are distinguished in that the binding to the former but not the latter is lost in the presence of denaturing solvents. The invention comprises also a human antibody binding to IL-13Ral and inhibiting IL-13 bioactivity, characterized by an affinity of 10"9 M (KD) or less, preferably of 10"9 to 10"13 M for binding to IL-13Ral and by an affinity of 10"7 M (KD) or less, preferably of 10~8 to 10"9M for binding to murine IL-13Ral.
In a preferred embodiment of the invention, the antibodies according to the invention are further characterized by one or more of the characteristics selected from the group selected from the binding parameters ka, kd and KD, binding to the same epitope to which an antibody selected from the group consisting of antibodies LC5002-002, LC5002-003, LC5002-005, LC5002-007 or LC5002-018 binds.
The antibodies according to the invention are preferably produced by recombinant means. Such methods are widely known in the state of the art and comprise protein expression in prokaryotic and eukaryotic cells with subsequent isolation of the antibody polypeptide and usually purification to a pharmaceutically acceptable purity. For the protein expression, nucleic acids encoding light and heavy chains or fragments thereof are inserted into expression vectors by standard methods. Expression is performed in appropriate prokaryotic or eukaryotic host cells like CHO cells, NSO cells, SP2/0 cells* HEK293 cells, COS cells, yeast, or E.coli cells, and the antibody is recovered from the cells (supernatant or cells after lysis).
Recombinant production of antibodies is well-known in the state of the art and described, for example, in the review articles of Makrides, S.C., Protein Expr. Purif. 17 (1999) 183-202; Geisse, S., et ah, Protein Expr. Purif. 8 (1996) 271-282; Kaufman, R.J., Mol. Biotechnol. 16 (2000) 151-161; Werner, R.G., Drug Res. 48 (1998)870-880.
The antibodies may be present in whole cells, in a cell lysate, or in a partially purified or substantially pure form. Purification is performed in order to eliminate other cellular components or other contaminants, e.g. other cellular nucleic acids or proteins, by standard techniques, including alkaline/SDS treatment, CsCl banding, column chromatography, agarose gel electrophoresis, and others well known in the art. See Ausubel, F., et al., ed. Current Protocols in Molecular Biology, Greene Publishing and Wiley Interscience, New York (1987).

Expression in NSO cells is described by, e.g., Barnes, L.M., el al., Cytotechnology 32 (2000) 109-123; and Barnes, L.M., et al., Biotech. Bioeng. 73 (2001) 261-270. Transient expression is described by, e.g., Durocher, Y., et al., Nucl. Acids. Res. 30 (2002) E9. Cloning of variable domains is described by Orlandi, R., et al., Proc. Natl. Acad. Sci. USA 86 (1989) 3833-3837; Carter, P., et al., Proc. Natl. Acad. Sci. USA 89 (1992) 4285-4289; and Norderhaug, L., et ah, J. Immunol. Methods 204 (1997) 77-87. A preferred transient expression system (HEK 293) is described by Schlaeger, E.-J., and Christensen, K., in Cytotechnology 30 (1999) 71-83 and by Schlaeger, E.-J., in J. Immunol. Methods 194 (1996) 191-199.
The control sequences that are suitable for prokaryotes, for example, include a promoter, optionally an operator sequence, and a ribosome binding site. Eukaryotic cells are known to utilize promoters, enhancers and polyadenylation signals.
The monoclonal antibodies are suitably separated from the culture medium by conventional immunoglobulin purification procedures such as, for example, protein A-Sepharose, hydroxylapatite chromatography, gel electrophoresis, dialysis, or affinity chromatography. DNA and RNA encoding the monoclonal antibodies is readily isolated and sequenced using conventional procedures. The hybridoma cells can serve as a source of such DNA and RNA. Once isolated, the DNA may be inserted into expression vectors, which are then transfected into host cells such as CHO cells, HEK 293 cells, or myeloma cells that do not otherwise produce immunoglobulin protein, to obtain the synthesis of recombinant monoclonal antibodies in the host cells.
The antibodies according to the invention include, in addition, such antibodies having "conservative sequence modifications", nucleotide and amino acid sequence modifications which do not affect or alter the above-mentioned characteristics of the antibody according to the invention. Modifications can be introduced by standard techniques known in the art, such as site-directed mutagenesis and PCR-mediated mutagenesis. Conservative amino acid substitutions include ones in which the amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined in the art. These families include amino acids with basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g. glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine, tryptophan), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine), beta-branched side chains

(e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine). Thus, a predicted nonessential amino acid residue in a human anti-IL-13Ral antibody can be preferably replaced with another amino acid residue from the same side chain family.
Amino acid substitutions can be performed by mutagenesis based upon molecular modeling as described by Riechmann, L., et al., Nature 332 (1988) 323-327 and Queen, C, et al., Proc. Natl. Acad. Sci. USA 86 (1989)10029-10033.
Amino acid sequence variants of human lL-13Ral antibody are prepared by introducing appropriate nucleotide changes into the antibody DNA, or by peptide synthesis. Such modifications can be performed, however, only in a very limited range, e.g. as described above. For example, the modifications do not alter the abovementioned antibody characteristics such as the IgG isotype and epitope binding, but may improve the yield of the recombinant production, protein stability or facilitate the purification.
Any cysteine residue not involved in maintaining the proper conformation of the anti- IL-13Ral antibody also may be substituted, generally with serine, to improve the oxidative stability of the molecule and prevent aberrant crosslinking. Conversely, cysteine bond(s) may be added to the antibody to improve its stability (particularly where the antibody is an antibody fragment such as an Fv fragment).
Another type of amino acid variant of the antibody alters the original glycosylation pattern of the antibody. By altering is meant deleting one or more carbohydrate moieties found in the antibody, and/or adding one or more glycosylation sites that are not present in the antibody. Glycosylation of antibodies is typically N-linked. N-linked refers to the attachment of the carbohydrate moiety to the side chain of an asparagine residue. The tripeptide sequences asparagine-X-serine and asparagine-X-threonine, where X is any amino acid except proline, are the recognition sequences for enzymatic attachment of the carbohydrate moiety to the asparagine side chain. Thus, the presence of either of these tripeptide sequences in a polypeptide creates a potential glycosylation site. Addition of glycosylation sites to the antibody is conveniently accomplished by altering the amino acid sequence such that it contains one or more of the above-described tripeptide sequences (for N-linked glycosylation sites).

Nucleic acid molecules encoding amino acid sequence variants of anti-IL-I3Rctl antibody are prepared by a variety of methods known in the art. These methods include, but are not limited to, isolation from a natural source (in the case of naturally occurring amino acid sequence variants) or preparation by oligonucleotide-mediated (or site-directed) mutagenesis, PCR mutagenesis, and cassette mutagenesis of an earlier prepared variant or a non-variant version of anti-IL-13Ral antibody.
Another type of covalent modification involves chemically or enzymatically coupling glycosides to the antibody. These procedures are advantageous in that they do not require production of the antibody in a host cell that has glycosylation capabilities for N- or O-linked glycosylation. Depending on the coupling mode used, the sugar(s) may be attached to (a) arginine and histidine, (b) free carboxyl groups, (c) free sulfhydryl groups such as those of cysteine, (d) free hydroxyl groups such as those of serine, threonine, or hydroxyproline, (e) aromatic residues such as those of phenylalanine, tyrosine, or tryptophan, or (f) the amide group of glutamine. These methods are described in WO 87/05330, and in Aplin, J.D., and Wriston, J.C. Jr., CRC Crit. Rev. Biochem. (1981) 259-306.
Removal of any carbohydrate moieties present on the antibody may be accomplished chemically or enzymatically. Chemical deglycosylation can be accomplished by exposing the antibody to trifluoromethanesulfonic acid, or an equivalent compound. This treatment results in the cleavage of most or all sugars except the linking sugar (N-acetylglucosamine or N-acetylgalactosamine), while leaving the antibody intact. Chemical deglycosylation is described by Sojahr, H.T., and Bahl, O.P., Arch. Biochem. Biophys. 259 (1987) 52-57 and by Edge, A.S., et al. Anal. Biochem. 118 (1981) 131-137. Enzymatic cleavage of carbohydrate moieties on antibodies can be achieved by the use of a variety of endo- and exo-glycosidases as described by Thotakura, N.R., and Bahl, O.P., Meth. Enzymol. 138(1987)350-359.
Another type of covalent modification of the antibody comprises linking the antibody to one of a variety of nonproteinaceous polymers, e.g., polyethylene glycol, polypropylene glycol, or polyoxyalkylenes, in the manner set forth in US Patent Nos. 4,640,835; 4,496,689; 4,301, 144; 4,670,417; 4,791,192 or 4,179,337.
In yet another aspect, the invention provides isolated B-cells from a transgenic non-human animal, e.g. a transgenic mouse, which express the human anti-IL-13Ral

antibodies according to the invention. Preferably, the isolated B cells are obtained from a transgenic non-human animal, e.g., a transgenic mouse, which has been immunized with a purified or enriched preparation of IL-13Ral antigen and/or cells expressing IL-13Ral. Preferably, the transgenic non-human animal, e.g. a transgenic mouse, has a genome comprising a human heavy chain transgene and a human light chain transgene encoding all or a portion of an antibody of the invention. The isolated B-cells are then immortalized to provide a source (e.g. a hybridoma) of human anti-IL-13Ral antibodies. Accordingly, the present invention also provides a hybridoma capable of producing human monoclonal antibodies according to the invention. In one embodiment, the hybridoma includes a B cell obtained from a transgenic non-human animal, e.g., a transgenic mouse having a genome comprising a human heavy chain transgene and a human light chain transgene encoding all or a portion of an antibody of the invention, fused to an immortalized cell.
In a particular embodiment, the transgenic non-human animal is a transgenic mouse having a genome comprising a human heavy chain transgene and a human light chain transgene encoding all or a portion of an antibody of the invention. The transgenic non-human animal can be immunized with a purified or enriched preparation of IL-13Ral antigen and/or cells expressing IL-13Ral. Preferably, the transgenic non-human animal, e.g. the transgenic mouse, is capable of producing IgGl isotypes of human monoclonal antibodies to IL-13Ral.
The human monoclonal antibodies according to the invention can be produced by immunizing a transgenic non-human animal, e.g. a transgenic mouse, having a genome comprising a human heavy chain transgene and a human light chain transgene encoding all or a portion of an antibody of the invention, with a purified or enriched preparation of IL-13Ral antigen and/or cells expressing IL-13Ral. B cells (e.g. splenic B cells) of the animal are then obtained and fused with myeloma cells to form immortal, hybridoma cells that secrete human monoclonal antibodies against IL-13Ral.
In a preferred embodiment, human monoclonal antibodies directed against IL-13Ral can be generated using transgenic mice carrying parts of the human immune system rather than the mouse system. These transgenic mice, referred to herein as "HuMab" mice, contain a human immunoglobulin gene miniloci that encodes unrearranged human immunoglobulin genes which include the heavy (\i and y) and K light chain (constant region genes), together with targeted mutations that

inactivate the endogenous u and K chain loci (Lonberg, N., et al., Nature 368 (1994) 856-859). Accordingly, the mice exhibit reduced expression of mouse IgM or K, and in response to immunization, the introduced human heavy and light chain transgenes undergo class switching and somatic mutation to generate high affinity human IgG monoclonal antibodies (Lonberg, N., et al, Nature 368 (1994) 856-859; reviewed in Lonberg, N., Handbook of Experimental Pharmacology 113 (1994) 49-101; Lonberg, N., and Huszar, D., Intern. Rev. Immunol. 25 (1995) 65-93; and Harding, F., and Lonberg, N., Ann. N. Acad. Sci 764 (1995) 536-546). The preparation of HuMab mice is described in Taylor, L.? et al., Nucleic Acids Research 20 (1992) 6287-6295; Chen, J., et al., International Immunology 5 (1993) 647-656; Tuaillon, N., et al., Proc. Natl. Acad. Sci USA 90 (1993) 3720-3724; Choi, T.K., et al., Nature Genetics 4 (1993) 117-123; Chen, J., et al., EMBO J. 12 (1993) 821-830; Tuaillon, N., et al., Immunol. 152 (1994) 2912-2920; Lonberg, N., et al., Nature 368 (1994) 856-859; Lonberg, N., Handbook of Experimental Pharmacology 113 (1994) 49-101; Taylor, L., et al., Int. Immunol. 6 (1994) 579-591; Lonberg, N., and Huszar, D., Intern. Rev. Immunol. 25 (1995) 65-93; Harding, F., and Lonberg, N., Ann. N. Acad. Sci 764 (1995) 536-546; Fishwild, D.M., et al., Nat. Biotechnol. 14 (1996) 845-851, the contents of all of which are hereby incorporated by reference in their entirety. See further, US Patent Nos. 5,545,806; 5,569,825; 5,625,126; 5,633,425; 5,789,650; 5,877,397; 5,661,016; 5,814,318; 5,874,299; 5,545,807; 5,770,429; WO 98/24884; WO 94/25585; WO 93/1227; WO 92/22645; and WO 92/03918.
To generate fully human monoclonal antibodies to IL-13Ral, HuMab mice can be immunized with a purified or enriched preparation of IL-13R
immune response can be monitored over the course of the immunization protocol with plasma samples being obtained by retroorbital bleeds. The plasma can be screened by EL1SA, and mice with sufficient titers of anti-IL-13Ral human immunoglobulin can be used for immortalization of corresponding B cells. Mice can be boosted intravenously with antigen 3 to 4 days before sacrifice and removal of the spleen and lymph nodes. It is expected that 2-3 fusions for each antigen may need to be performed. Several mice will be immunized for each antigen. For example, a total of five to twelve HuMab mice of the HCo7 and HCol2 strains can be immunized.
The HCo7 mice have a JKD disruption in their endogenous light chain (kappa) genes (as described in Chen, J., et aL, EMBO J. 12 (1993) 821-830), a CMD disruption in their endogenous heavy chain genes (as described in Example 1 of WO 01/14424), a KCo5 human kappa light chain transgene (as described in Fishwild, D.M., et aL, Nat. Biotechnol. 14 (1996) 845-851), and a HCo7 human heavy chain transgene (as described in US Patent No. 5,770,429).
The HCol2 mice have a JKD disruption in their endogenous light chain (kappa) genes (as described in Chen, J., et aL, EMBO J. 12 (1993) 821-830), a CMD disruption in their endogenous heavy chain genes (as described in Example 1 of WO 01/14424), a KCo5 human kappa light chain transgene (as described in Fishwild, D.M., et aL, Nat. Biotechnol. 14 (1996) 845-851), and a HCol2 human heavy chain transgene (as described in Example 2 of WO 01/14424).
The mouse lymphocytes can be isolated and fused with a mouse myeloma cell line using PEG based on standard protocols to generate hybridomas. The resulting hybridomas are then screened for the production of antigen-specific antibodies. For example, single cell suspensions of splenic and lymph node-derived lymphocytes from immunized mice are fused to one-sixth the number of SP 2/0 non secreting mouse myeloma cells (ATCC, CRL 1581) with 50% PEG. Cells are plated at approximately 2x10 in flat bottom microtiter plate, followed by about two weeks incubation in selective medium.
Individual wells are then screened by ELISA for human anti-IL-13Ral monoclonal ■ IgM and IgG antibodies. Once extensive hybridoma growth occurs, medium is analyzed, usually after 10-14 days. The antibody secreting hybridomas are replated, screened again, and if still positive for human IgG, anti-IL-13Ral monoclonal antibodies, can be subcloned at least twice by limiting dilution. The stable

subclones are then cultured in vitro to produce antibody in tissue culture medium for characterization.
Because CDR sequences are responsible for antibody-antigen interactions, it is possible to express recombinant antibodies according to the invention by constructing expression vectors that include the CDR sequences according to the invention onto framework sequences from a different human antibody (see, e.g., Riechmann, L., et al., Nature 332 (1998) 323-327; Jones, P., et al., Nature 321 (1986) 522-525; and Queen, C, et al., Proc. Natl. Acad. See. U.S.A. 86 (1989)10029-10033). Such framework sequences can be obtained from public DNA databases that include germline human antibody gene sequences. These germline sequences will differ from mature antibody gene sequences because they will not include completely assembled variable genes, which are formed by V(D)J joining during B cell maturation. Germline gene sequences will also differ from the sequences of a high affinity secondary repertoire antibody at individual evenly across the variable region.
The invention preferably comprises a nucleic acid fragment encoding a polypeptide binding to IL-13Ral, whereby said polypeptide inhibits the binding of IL-13to IL-13Ral, selected from the group consisting of
a) an antibody heavy chain comprising heavy chain CDRs of SEQ ID NO: 1, 3, 5, 7 or 9;
b) an antibody light chain comprising light chain CDRs of SEQ ID NO: 2, 4, 6, 8 or 10.
The reconstructed heavy and light chain variable regions are combined with sequences of promoter, translation initiation, constant region, 31 untranslated, polyadenylation, and transcription termination to form expression vector constructs. The heavy and light chain expression constructs can be combined into a single vector, co-transfected, serially transfected, or separately transfected into host cells which are then fused to form a single host cell expressing both chains.
Accordingly, the invention provides a method for the production of a recombinant human antibody according to the invention, comprising expressing a nucleic acid encoding

a) an antibody heavy chain comprising heavy chain CDRs of SEQ ID NO:l, 3, 5, 7 or 9;
b) an antibody light chain comprising light chain CDRs of SEQ ID NO: 2, 4, 6, 8 or 10.
The invention further comprises the use of an antibody according to the invention for the detection of IL-13Ral in vitro, preferably by an immunological assay determining the binding between IL-13Ral of a sample and the antibody according to the invention.
In another aspect, the present invention provides a composition, e.g. a pharmaceutical composition, comprising one or a combination of human monoclonal antibodies, or the antigen-binding portion thereof, of the present invention, formulated together with a pharmaceutically acceptable carrier.
As used herein, "pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible. Preferably, the carrier is suitable for intravenous, intramuscular, subcutaneous, parenteral, spinal or epidermal administration (e.g. by injection or infusion).
A "pharmaceutically acceptable salt" refers to a salt that retains the desired biological activity of the antibody and does not impart any undesired toxicological effects (see e.g. Berge, S.M., et al, J. Pharm. Sci. 66 (1977) 1-19). Such salts are included in the invention. Examples of such salts include acid addition salts and base addition salts. Acid addition salts include those derived from nontoxic inorganic acids, such as hydrochloric salts.
A composition of the present invention can be administered by a variety of methods known in the art. As will be appreciated by the skilled artisan, the route and/or mode of administration will vary depending upon the desired results.
To administer a compound of the invention by certain routes of administration, it may be necessary to coat the compound with, or coadminister the compound with, a material to prevent its inactivation. For example, the compound may be administered to a subject in an appropriate carrier, for example, liposomes, or a diluent. Pharmaceutically acceptable diluents include saline and aqueous buffer solutions.

Pharmaceutically acceptable carriers include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion. The use of such media and agents for pharmaceutically active substances is known in the art.
The phrases "parenteral administration" and "administered parenterally" as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal, epidural and intrasternal injection and infiision.
These compositions may also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of presence of microorganisms may be ensured both by sterilization procedures, supra, and by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol, sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption such as aluminum monostearate and gelatin.
Regardless of the route of administration selected, the compounds of the present invention, which may be used in a suitable hydrated form, and/or the pharmaceutical compositions of the present invention, are formulated into pharmaceutically acceptable dosage forms by conventional methods known to those of skill in the art.
Actual dosage levels of the active ingredients in the pharmaceutical compositions of the present invention may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient. The selected dosage level will depend upon a variety of pharmacokinetic factors including the activity of the particular compositions of the present invention employed, or the ester, salt or amide thereof, the route of administration, the time of administration, the rate of excretion of the particular compound being employed, the duration of the treatment, other drugs, compounds

and/or materials used in combination with the particular compositions employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts.
The composition must be sterile and fluid to the extent that the composition is deliverable by syringe. In addition to water, the carrier can be an isotonic buffered saline solution, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyetheylene glycol, and the like), and suitable mixtures thereof.
Proper fluidity can be maintained, for example, by use of coating such as lecithin, by maintenance of required particle size in the case of dispersion and by use of surfactants. In many cases, it is preferable to include isotonic agents, for example, sugars, polyalcohols such as mannitol or sorbitol, and sodium chloride in the composition. Long-term absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate or gelatin.
The following examples, references, sequence listing and figures are provided to aid the understanding of the present invention, the true scope of which is set forth in the appended claims. It is understood that modifications can be made in the procedures set forth without departing from the spirit of the invention.
Description of the Sequence Listing
SEQ ID NO:l heavy chain variable domain of HuMab LC5002-002
SEQ ID NO:2 light chain variable domain of HuMab LC5002-002
SEQ ID NO:3 heavy chain variable domain of HuMab LC5002-003
SEQ ID NO:4 light chain variable domain of HuMab LC5002-003
SEQ ID NO:5 heavy chain variable domain of HuMab LC5002-005
SEQ ID NO:6 light chain variable domain of HuMab LC5002-005
SEQ ID NO:7 heavy chain variable domain of HuMab LC5002-007

SEQ ID NO:8 light chain variable domain of HuMab LC5002-007
SEQ ID NO:9 heavy chain variable domain of HuMab LC5002-018
SEQ ED NO:10 light chain variable domain of HuMab LC5002-018
SEQ ID NO: 11 K light chain constant region
SEQ ID NO:12 y\ heavy chain constant region
Description of the Figures
Figure 1 shows binding of anti-IL-13Ral antibodies to immobilized
recombinant human IL-13Ral polypeptide. Included are polyclonal rabbit-anti-human IL-13Ral antibody AF152 (R&D systems) and anti-KLH as a negative control HuMab.
Figure 2 shows inhibition of IL-13 binding to immobilized IL-13Ral/IL-
4Ra receptor by anti-IL-13Ral antibodies.
Figure 3 shows the blockade of IL-13 binding to CHO cells (expressing
IL-13Ralphal and IL-4Ra2) by anti-IL-13Ral antibodies. As a positive control, a commercially available polyclonal anti-IL-13Ral antibody (AF152, R&D Systems, Minneapolis, MN) was included.
Figure 4 shows binding of anti-IL-13Rctl antibodies to hIL-13Ral and
binding properties to functionally related receptors hIL-13Ra2 and hIL-4Ra.
Figure 5 shows the capacity of anti-IL-13Ral antibodies to bind to
immobilized recombinant murine IL-13Ra 1 polypeptide. Included are polyclonal goat-anti- human IL-13Ral antibody AF152 (R&D Systems) and anti-KLH as a negative control HuMab.

Examples
Example 1
Generation of hybridomas
The human monoclonal antibodies according to the invention can be produced by immunizing a transgenic non-human animal, e. g. a transgenic mouse having a genome comprising a human heavy chain transgene and a human light chain transgene encoding all or a portion of an antibody of the invention, with cells expressing human IL-13Ral. B cells (e.g. splenic B cells) of the animal are then obtained and fused with myeloma cells to form immortal, hybridoma cells that secrete human monoclonal antibodies against IL-13Ral. Human monoclonal antibodies directed against human IL-13Ral can be generated using transgenic mice carrying parts of the human immune system rather than the mouse system. These transgenic mice, referred to herein as "HuMab" mice, contain a human immunoglobulin gene minilocus that encodes unrearranged human immunoglobulin genes which include the heavy (\i and y) and K (kappa) light chain (constant region genes), together with targeted mutations that inactivate the endogenous nand kappa chain loci (Lonberg N., et al., Nature 368 (1994) 856-859) Accordingly, the mice exhibit reduced expression of mouse IgM or K, and in response to immunization, the introduced human heavy and light chain transgenes undergo class switching and somatic mutation to generate high affinity human IgG monoclonal antibodies. To generate fully human monoclonal antibodies to human IL-13Ral, HuMab mice can be immunized with cells expressing human IL-13Ral in accordance with the general method, as described by Lonberg, N., et al., Nature 368 (1994) 856-859; Fishwild, D.M., et al., Nat. Biotechnol. 14 (1996) 845-851 and WO 98/24884. Preferably, the mice will be 6-16 weeks of age upon the first immunization. For example, IL-13Ral transfected cells can be used to immunize the HuMab mice intraperitoneally. The immune response can be monitored over the course of the immunization protocol with plasma samples being obtained by retroorbital bleeds. The plasma can be screened by EL1SA and/or FACS. Mice with sufficient titers of anti-human IL-13Ral human immunoglobulin can be used for immortalization of corresponding B cells. Mice can be boosted intravenously with antigen 3 to 4 days before sacrifice and removal of the spleen and lymph nodes. For example, HuMab mice of the HCo7 or HCol2 strain can be immunized. The HCo7 mice have a JKD disruption in their endogenous light chain (kappa) genes (as described in Chen et al. (1993) EMBO J. 12: 821-830), a CMD disruption in their endogenous heavy chain genes (as described in Example 1 of WO 01/14424),

a KCo5 human kappa light chain transgene (as described in Fishwild, -D.M., et a!. (1996) Nature Biotechnology 14:845-851), and a HCo7 human heavy chain transgene (as described in US 5,770,429). The HCol2 mice have a JKD disruption in their endogenous light chain (kappa) genes (as described in Chen, J., et al., EMBO J. 12 (1993) 821-830), a CMD disruption in their endogenous heavy chain genes (as described in Example 1 of WO 01/14424)) a KCo5 human kappa light chain transgene (as described in Fishwild, D.M., et al., Nat. Biotechnol. 14 (1996) 845-851, and a HCol2 human heavy chain transgene (as described in Example 2 of WO 01/14424). The mouse lymphocytes can be isolated and ftised with a mouse myeloma cell line using PEG based on standard protocols to generate hybridomas. The resulting hybridomas are then screened for the production of antigen-specific antibodies. For example, single cell suspensions of splenic and lymph node derived lymphocytes from immunized mice are ftised to SP 2/0 nonsecreting mouse myeloma cells (ATCC, CRL 1581) with 50% PEG. Cells are plated at approximately 0.75 x 10 in flat bottom micro titer plate, followed by about two weeks incubation in selective medium. Individual wells are then screened by ELISA and/or FACS for human anti-IL-13Ral monoclonal IgM and IgG antibodies. Once extensive hybridoma growth occurs, the antibody secreting hybridomas are replated, screened again, and if still positive for human IgG, anti-IL-13Ral monoclonal antibodies, can be subcloned at least twice by limiting dilution. The stable subclones are then cultured in vitro to produce antibody in tissue culture medium for characterization.
Immunization procedure of transgenic mice
Three HCo7 mice (3 males), strain GG2201 (Medarex, San Jose, CA, USA) and 2 HCol2 mice (1 male and 1 female), strain GG2198 (Medarex, San Jose, CA, USA) were immunized with 1 x 106 HEK293 cells, transfected with an expression vector for IL-13Ral. In total eight immunizations were given alternating intraperitoneally (i.p.) and subcutaneous (s.c.) at the tail base. For the first immunization, 100 |al of 1 x 106 HEK293: IL-13Ral cells, was mixed with 100 fil complete Freund's adjuvant (CFA; Difco Laboratories, Detroit, USA). For all other immunizations, 100 u1 of cells in PBS was mixed with 100 uj.1 incomplete Freund's adjuvant (ICFA; Difco).

Boosting of mice
When serum titers of anti-IL-13Ral were found to be sufficient, mice were additionally boosted twice with 1 xlO6 HEK293: IL-13Ral cells in 200 \i\ PBS intravenously (i.v.) 4 and 3 days before fusion.
Esampte 2
Testing the binding of HuMab to immobilized IL-13Ral by ELISA
To determine the ability of the antibodies of the invention to bind to recombinant IL-13Ral, the extracellular domain of IL-13Ral (R&D Systems, UK) was dissolved in PBS (1 ng/ml) and allowed to adsorb to microtiter plates (NUNC Maxisorb) by incubation over night at 4°C. After washing the plates with washing buffer (WB = 0.9 % NaCl; 0.1% Tween® 20) unspecific binding sites were blocked by addition of 100 u.1 incubation buffer (IB = PBS with 1% crotein C and 0.1% Tween® 20) and incubation for 30 min at room-temperature (RT). Then, serially diluted HuMab and control antibodies (100 \i\ /well; dilutions in IB) were added and incubated for 1 hour at RT. The plates were again washed, and bound human antibodies were detected by incubation with peroxidase-conjugated rabbit anti-human kappa (DAKO, Denmark) in a final dilution of 1:500 in IB. Polyclonal goat anti-hIL-13Ra 1 antibodies were detected with peroxidase-conjugated polyclonal donkey anti-goat IgG (Santa Cruz; dilution 1:1000 in IB). After incubation for Ih at RT and a subsequent washing step, the plates were developed with ready-to-use ABTS® solution (Roche Diagnostics GmbH) at RT in the dark. Absorbance was measured at 405 nm after absorbance of the highest concentration reached a sufficient OD.
All antibodies against IL-13Ralphal tested were able to bind to immobilized extracelluar domains of human IL-13Ral. The EC50 values determined were in the range of 0.5 - 2 nM for the various LC antibodies tested. The negative control HuMab anti-KLH did not bind to the immobilized extracellular domains of IL-13Ral. Polyclonal goat-anti human IL-13Ral antibody, included as positive control, also bound efficiently to the immobilized extracellular domains of IL-13RQ1 (Fig.l).

Example 3
Inhibition of IL-13 binding to IL-13Ral/IL-4Ra heterodimer (ELISA)
Microliter plates were coated with 100 |il hIL-13Ral:hFc chimeric protein (R&D Systems, UK) in PBS at 3 |ig/ml at 4°C overnight on a shaker. After washing the plates with WB, serially diluted HuMab and control antibodies (100 \i\ /well; dilutions in IB) were added and incubated for 30 min at RT. The plates were again washed, and then a mixture of IL-13 (R&D Systems, UK; 0.5 |ig/ml; dilution with IB) and IL-4Ra (R&D Systems, UK; 0.75 |ig/ml; dilution with IB) were added and incubated for 1 h at RT. After washing the plates 100 \i\ biotinylated anti IL-13 antibody (BAF213; R&D Systems, UK) in a concentration of 0.4 jig/ml was added and incubated for Ih at RT. After washing the plates, bound IL-13 was detected by peroxidase-coupled streptavidin (Roche Diagnostics GmbH, DE) in a dilution of 1:5000 in IB (incubation period lh at RT). Finally, plates were washed and developed with ready-to-use ABTS® solution (Roche Diagnostics GmbH, DE) at room temperature (RT) in the dark. Absorbance was measured at 405 nm after 45 to 60 minutes.
Antibodies LC5002-002, LC5002-003, LC5002-005, LC5002-007 and LC5002-018 were able to inhibit binding of IL-13 to the heterodimeric receptor with maximal inhibition values ranging from approximately 50% to 80-85%. Positive control was AF152 (polyclonal rabbit antibody). As expected, the negative control anti-KLH did not inhibit binding of IL-13 to the heterodimeric receptor. The IC50 values obtained were between 1.5 nM and 10.1 nM for LC5002-002, LC5002-003, LC5002-005, LC5002-007 and LC5002-018 (Fig.2).
Example 4
Radioligand Binding Assay
I-EL-13 binding assay was performed using CHO cells expressing human IL-13Rctl and human IL-4Ra in binding buffer (25 mM HEPES, 150 mM NaCl, 1 mM CaCb, 5 mM MgCfe, and 0.5% bovine serum albumin, adjusted to pH 7.2). lxlO5 cells per well were mixed with the antibodies and preincubated for 15 minutes to 1 hour. 0.1 nM 125I-IL-13 was added, and the mix was incubated at 4°C for 4 hours. The concentration of I-IL-13 used in the assay was determined from saturation binding analysis, competition analysis and determination of input I-IL-13 to reach equilibrium binding with the cell line. Samples were harvested onto a GF/C filter plate pretreated with 1% PEI/0.5% BSA and counted on Packard

TopCount Scintillation counter. Data analysis was performed in PRISM using nonlinear regression curve fit (GraphPad Software, San Diego, CA).
All antibodies aganist IL-13Ralphal tested block binding of labeled IL-13 to the IL-13Ral/IL-4Rct complex. The calculated IC50 values for antibodies LC5002-002, LC5002-003, LC5002-005, LC5002-007 and LC5002-018 were between 0.09 nM and 0.32 nM and 84.8nM for AF152 (Fig.3).
Example 5
Inhibition of IL-13 induced upregulation of CD23 on human B cells and monocytes by HuMab
Peripheral blood mononuclear cells (PBMC) were isolated by a Ficoll Hypaque density gradient. After washing the cells with RPMI they were resuspended in RPMI/10% FCS and distributed at 3xl05 PBMC/well (volume 50 nl) in 96 well flat bottom microtiter plates (Corning Incorporated Costar). Then, 25 \i\ of an anti-human CD40 antibody (Immunotech) in a final concentration 0.5 ^g/ml in RPMI/10% FCS and 25 nl of an anti-human IgA+IgG+IgM antibody (Immunoresearch) in a final concentration of 10 \ig/m\ in RPMI/10%FCS were added. Then serially diluted HuMab and control antibodies (50 jil/well; dilutions in RPMI/10% FCS) were added and the cells incubated for 30 min in the incubator (37°C; 5% CO2). Then recombinant human IL-13 (R&D Systems) in a final concentration of 0.67 ng/ml was added (50 fil/well) and the cells incubated for 72h at 37°C/5% CO2. After this incubation the plates were centrifuged and the medium aspirated. For detachment of adherent cells 200 jil of Accutase (PAA) was added and the cells incubated for approximately 5 min at 37°C; 5% CO2. The cells were detached by repeated flushing and transferred to a round bottom plate. After centrifiigation and aspiration of the supernatants the cells were incubated with 200 pi of a mixture of anti-CD23-PE, anti-CD20-FITC and anti-CD14-APC (all from BD Biosciences Pharmingen, San Diego, CA). The cells were incubated for 30 min at 4°C, then centrifuged and the supernatants aspirated. This washing step was repeated once, and finally the cells were resupended in 200 \i\ of PBS/0.1% human . serum albumin and analysed in a FACS Calibur flow cytometer (BD Biosciences Pharmingen, San Diego, CA) using the CellQuest software. In most cases, 10 000 events were acquired and gated on a light scatter gate to include only viable lymphocytes and monocytes. The cells were pregated on a CD 19 positive cluster

for B lymphocytes or a CD 14 positive cluster for monocytes and analyzed further forCD23 expression.
The observed IC50 values for inhibiting of CD 23 upregulation on B-Iymphocytes were between 0.5 nM and >70 nM for antibodies LC5002-002, LC5002-003, LC5002-005, LC5002-007 and LC5002-018, and 13.6 nM for AF152. A similar profile was found for inhibition of IL-13 induced CD23 upregulation on human monocytes. On monocytes the IC50 values were between 0.1 nM and 62.8 nM for antibodies LC5002-002, LC5002-003, LC5002-005, LC5002-007 and LC5002-018, and 62.9 nM for AF152.
Example 6
TF-1 proliferation assay in response to IL-13 or IL-4 as stimulus
TF-1 cells (ATCC # CRL 2003) were grown in media containing ATCC modified RPMI, 10% FBS, IX Pencillin/Streptomycin, 2ng/ml GM-CSF. A day prior to the assay the cells were maintained in GM-CSF free media. 5x10 cells per well were incubated with appropriate concentrations of anti~IL-13Rotl antibodies at 37°C for 1 hour. Then the cells were stimulated with 2ng/ml of human IL-13 (R&D Systems, Minneapolis, MN) or 0.1 ng/ml of human IL-4 (R&D Systems, Minneapolis, MN) and incubated at 37°C for 48 hours. The cells were pulsed with 0.5 jxCi 3H-Thymidine and incubated at 37°C for 16-18 hours. Samples were harvested onto GFC plates pretreated with 1% PEI/0.25% BSA using Perkin Elmer Filtermate 96 harvester. The GFC plates were counted on a Perkin Elmer Top count Scintillation counter. Data analysis was performed in PRISM using nonlinear regression curve fit (GraphPad Software, San Diego, CA).
Anti-KLH antibody did not show any inhibition in this assay. The same was true for LC5002-007. All other antibodies inhibited the response, even though LC5002-007 inhibited the response with higher IC value than the other antibodies. The observed IC50 values for the different antibodies were: 13.50 nM for AF152, 9.21 nM for LC5002-002, 3.07 nM for LC5002-003 and 0.39 nM for LC5002-005. A similar profile was found for IL-4-induced TF-1 cell proliferation, however the potency of the antibodies was decreased compared to IL-13-induced responses. IC50 values for IL-4-induced proliferation were 0.02 nM for anti-IL4R antibody, 74.37 nM for AF152 and for antibodies LC5002-002, LC5002-003, LC5002-005, LC5002-007 and LC5002-018 between 4.68 nM and 60 nM.

Example 7
Inhibition of eotaxin production in response to IL-13 by human lung fibroblast
The assay was performed using HFL-1 cells (Human Lung Fibroblast, ATCC # CCL-153). Cells were plated at a density of 100,000 cells per well in a 12-well plate and incubated at 37°C for 72 hours to reach confluency. Cells were then starved in serum free medium for 24 h and treated with anti-IL-13Ral antibody at 37°C for 1 h. Following this treatment cells were stimulated with 10 ng/ml IL-13 (R&D Systems, Minneapolis, MN) at 37°C for 48 h. Supernatants were collected and eotaxin determinations were done using commercially available ELISA from R&D Systems (Cat. No. DTX00). Absorbance was read using Spectromax microplate reader and the data was analyzed using PRISM (GraphPad Software, San Diego, CA).
The antibodies tested showed different capability to inhibit eotaxin release. With the exeption of LC5002-007 all other antibodies tested showed some inhibition. The calculated average IC50 values from 3 to 4 different experiments were 11.5 nM for AF152 and between 2.45 nM and 19.8nM for antibodies LC5002-002, LC5002-003, LC5002-005, LC5002-007 and LC5002-018.
ifaaropte 8
Inhibition of IL-13-induced Stat-6 phosphorylation in human bronchial smooth muscle cells
Human Bronchial Smooth Muscle Cells (BSMC; Clonetics, Cat No CC-2576) were grown following the manufacturer's instructions. Cells were grown in 12-well tissue culture plates until they reached confluency. Cells were starved for 24h in serum-free medium and variable amounts of antibody were added. Plates were incubated for lh and then stimulated with 2.5 ng/ml IL-13 (R&D System). After 15 min incubation, supernatant was removed, cells were washed with phosphate buffer and 100 nl of lysis buffer was added. The mix was briefly sonicated on ice and centrifiiged. Lysate was used for Western Blot detection of phophorylated Stat-6. Equal amounts of protein were loaded onto an SDS-gel, run and transferred to a membrane. Anti-Stat-6 antibody was from Santa Cruz Biotechnology (Cat. No. SC-11762R) and a secondary antibody coupled to peroxidase was used. Detection was

done using the ECL Plus System from Amersham (Cat No RPN 2132). Quantitation was done in a Typhoon 9400 Imager.
Both HuMabs tested in this assay (LC50002-003 and LC5002-005) inhibited IL-13-induced Stat-6 phosphorylation. The potency found in this assay was similar to that of other functional assays. The calculated IC50 values were 18.64 nM for AF152, 5.98 nM for LC5002-003 and 1.18 nM for LC5002-005.
Example 9
Cloning and sequence analysis of anti-hIL-13Ral HuMab variable domains (K-light and yl-heavy chains)
The nucleotide sequences coding for the light chain variable region VL and the heavy chain variable region VH of the anti hIL-13Ral HuMabs were isolated by a standard cDNA synthesis / PCR procedure. Total RNA was prepared from lxl06 -1x10 hybridoma cells using the GeneRacer™ Kit (Invitrogen). Hybridoma derived RNA was used as a template for the 1st strand cDNA synthesis and ligation of the GeneRacer™ Oligo-dT Primer. 2nd-strand cDNA synthesis and further PCR amplification of VL and VH encoding cDNA fragments were performed with reverse light and heavy chain primers complementary to nucleotide sequences of the K-light and yl-heavy chain constant region and 5'-specific GeneRacer™ primers, respectively. The PCR products were cloned using the TOPO™ TA cloning kit from Invitrogen Life Technologies and pCR4-TOPO™ as a cloning vector. Cloned PCR products were identified by restriction mapping of the appropriate plasmids using EcoRI for digestion and expected / calculated DNA fragment sizes of about 740 and 790 bp for VL and VH, respectively. The DNA sequence of cloned PCR fragments was determined by double strand sequencing. The GCG (Genetics Computer Group, Madison, Wisconsin) software package version 10.2 and Vector-NTI 8 (InforMax, Inc) was used for general data processing. DNA and protein sequences were aligned using the GCG modul CLUSTALW. Sequence alignments were made using the program GENEDOC (version 2.1).
Example 10
Construction of expression plasmids for an anti-hIL-13Ral IgGl HuMab
The anti-hIL-13Ral HuMab light and heavy chain encoding genes were separately assembled in mammalian cell expression vectors. Thereby the gene segments

encoding the anti-hIL-13Ral HuMab light chain variable region (Vi.) and the human K-light chain constant region (CL, SEQ ID NO: 11) were joined as were gene segments for the anti-hIL-13Ral HuMab heavy chain variable region (VH) and the human yl-heavy chain constant region (Cm-Hinge-Cm-Cm, SEQ ID NO: 12). General information regarding the nucleotide sequences of human light and heavy chains from which the codon usage is given in: Kabat, E. A., Wu, T. T., Perry, H. M., Gottesman, K. S., and Foeller, C. ,(1991) Sequences of Proteins of Immunological Interest, Fifth Ed., NIH Publication No. 91-3242. The transcription unit of the anti-hIL-13Ral HuMab K-light chain is composed of the following elements:
• The immediate early enhancer and promoter from the human cytomegalovirus (HCMV),
• A synthetic 5'-UT including a Kozak sequence,
• A murine immunoglobulin heavy chain signal sequence including the signal sequence intron,
• The cloned anti-hIL-13Ral HuMab variable light chain cDNA arranged with a unique BsmI restriction site at the 5' end and a splice donor site and a unique NotI restriction site at the 3' end,
• The genomic human K-gene constant region, including the intron 2 mouse Ig-K enhancer [Picard, D., and Schaffiier, W., Nature 307 (1984) 80-82] and
• The human immunoglobulin K-polyadenylation ("poly A") signal sequence.
The transcription unit of the anti-hlL-13Ral HuMab yl-heavy chain is composed of the following elements:
• The immediate early enhancer and promoter from the human cytomegalovirus (HCMV),
• A synthetic 5'-UT including a Kozak sequence,
• A modified murine immunoglobulin heavy chain signal sequence including the signal sequence intron,
• The cloned anti-hIL-13Ral HuMab variable heavy chain cDNA arranged with a unique BsmI restriction site at the 5' and a splice donor site and a unique NotI restriction site at the 3' end,
• The genomic human yl-heavy gene constant region, including the mouse Ig ^-enhancer (Neuberger, M.S., EMBO J. 2 (1983) 1373-1378),
• The human yl-immunoglobulin polyadenylation ("poly A") signal sequence.

Functional elements of the anti-hIL-13Ral HuMab K-light chain and yl-heavy chain expression plasmids:
Besides the anti-hIL-13Ral HuMab K-light chain or yl-heavy chain expression cassette these plasmids contain
• A hygromycin resistance gene
• An origin of replication, oriP, of Epstein-Barr virus (EBV)
• An origin of replication from the vector pUC18 which allows replication of this plasmid in E. coli, and
• A B-lactamase gene which confers ampicillin resistance in E. coli.
Example 11
Construction of expression plasmids for mutant (variant) anti-hIL-13Ral IgGl
Expression plasmids encoding mutant anti-hIL-13Ral yl-heavy chains can be created by site-directed mutagenesis of the wild type expression plasmids using the QuickChange Site-Directed mutagenesis Kit (Stratagene) and are desribed in table 1. Amino acids are numbered according to EU numbering [Edelman, G.M., et al., Proc. Natl. Acad. Sci. USA 63 (1969) 78-85; Kabat, E. A., Wu, T. T., Perry, H. M., Gottesman, K. S., and Foeller, C, (1991) Sequences of Proteins of Immunological Interest, Fifth Ed., NIH Publication No. 91-3242].

Example iz
Production of recombinant anti-hIL-l3Ral HuMabs
Recombinant HuMabs were generated by transient transfection of adherent HEK293-EBNA cells (ATTC CRL-10852) cultivated in DMEM (Gibco) supplemented with 10 % ultra-low IgG FCS (Gibco), 2 mM Glutamine (Gibco), l% v/v nonessential aminoacids (Gibco) and 250 |ig/ml G418 (Roche Diagnostics GmbH, DE). For transfection Fugene™ 6 (Roche Diagnostics GmbH, DE) transfection reagent was used in a ratio of reagent (jil) to DNA (\ig) ranging from 3:1 to 6:1. Immunoglobulin light and heavy chains were expressed from two different plasmids using a molar ratio of light chain to heavy chain encoding plasmid from 1:2 to 2:1. HuMab containing cell culture supernatants were harvested at day 4 to 11 after transfection.
General information regarding the recombinant expression of human antibody in e.g. HEK293 is given in: Meissner, P., et al., Biotechnol Bioeng 75 (2001) 197-203.

Exaropte 13
a) Affinity analysis of HuMabs LC5002-003, -005, and -007 using chimeric hIL-13Ral:hFc
For interaction analysis a Biacore 3000 instrument was used. As running and reaction buffer, HBS-P (10 mM HEPES, 150 mM NaCl, 0.005 % polysurfactant P, pH 7.4) at 25°C was used. Capturing molecules (goat anti-human-IgG, Fey specifc) were amine-coupled at a concentration of 20 \ig/m\ at a flow rate of 5 jil/min for 20 minutes. HuMabs were injected at a concentration of 1 |ig/ml at a flow rate of 10 jil/min for 1 minute. Blocking of the free goat anti human IgG, FcY was achieved by injecting human gamma globulin at 500 nM and 30 nl/min for 3 minutes. Analyte (hIL-13Ral:hFc chimeric protein) was injected for two minutes at five concentrations between 5.63 nM and 90 nM and washed with HBS-P for five minutes. Regeneration of the surface was accomplished by two injections of 100 mM HC1 for 1 min each. The chip, assay format and sequence of injections and kinetic data correspond to the description in table 2. Negative control data (e.g. buffer curves) were subtracted from sample curves for correction of system intrinsic baseline drift and for noise signal reduction. BiaEvaluation version 4.01 was used for analysis of sensorgrams and for calculation of affinity data. Kinetic data were calculated by fitting kinetic data to a 1:1 Langmuir binding model (Table 2).
Table 2: Affinity analysis of HuMabs using hIL-13Ral:hFc. Data analysis based on 1:1


b) Affinity analysis of HuMabs LC5002-003, -005 and -007 using the extracellular domain of hIL-13Ral cleaved from of hIL-13Ral:hFc chimeric protein
For interaction analysis, a Biacore 3000 instrument was used. Running and reaction buffer was HBS-P (10 mM HEPES, 150 mM NaCl, 0.005 % polysurfactant P, pH 7.4) at 25°C. Capturing antibody molecules (anti-hFcy) were amine-coupled at a concentration of 100 ^g/ml at a flow rate of 5 ^l/min for 20 minutes. HuMabs were injected at concentrations of 10 \ig/ml at a flow rate of 10 (il/min for 30 seconds. Cleaved hIL-13Ral molecules (Mw40kDa) were injected for 200 seconds at seven concentrations between 1.56 nM and 100 nM and washed with HBS-P for five minutes. Regeneration of the surface was accomplished by two injections of 100 mM HC1 for 1 min each at a flow rate of 10 fig/ml. The chip, assay format and sequence of injections and kinetic data correspond to the description in the following Table 3. Kinetic data were calculated by fitting kinetic data to a 1:1 Langmuir binding model.
Tohla 1*

c) Comparative affinity analysis of recombinant variants of LC5002-005
using the extracellular domain of hIL-13Ral cleaved from hIL-13Ral:hFc chimeric protein
In these experiments, the affinity of the original IgGl derived from hybridoma was compared with the affinities of the recombinant variant IgGl-Ala-Ala. For interaction analysis, a Biacore 3000 instrument was used. Running and reaction buffer was HBS-P (10 mM HEPES, 150 mM NaCl, 0.005 % polysurfactant P, pH 7.4) at 25°C. Capturing antibody molecules (anti-hFcy) were amine-coupled at a concentration of 20 ng/ml at a flow rate of 5 nl/min for 20 minutes. HuMabs were injected at concentrations of 10 jig/ml at a flow rate of 10 ^1/min for 1 minute. Cleaved hIL-13 Ral molelcules (analyte) were injected for five minutes at eight concentrations between 1,56 nM and 200 nM and washed with HBS-P for five minutes. Regeneration of the surface was accomplished by two injections of 100 mM HC1 for 1 min each. The chip, assay format and sequence of injections and kinetic data correspond to the description in Table 4. Kinetic data were calculated by fitting kinetic data to a bivalent analyte binding model.
Table 4:

Example 14
Testing the crossreactivity of HuMab with hIL-13Ra2 and hIL-4Ra by ELISA
Chimeric proteins WL-13Ra2:hFc and hIL-4Ra:hFc (R&D Systems, UK) were dissolved in PBS (1 fig/ml) and allowed to adsorb to microtiter plates (NUNC Maxisorb) by incubation over night at 4°C. After washing the plates with washing buffer (WB = 0.9 % NaCl; 0.1% Tween® 20) unspecific binding sites were

blocked by addition of 100 u1 incubation buffer (IB = PBS with 1% crotein C and 0.1% Tween® 20) and incubation for 30 min at room-temperature (RT). Then serially diluted HuMab and control antibodies (100 jil/well; dilutions in IB) were added and incubated for 1 hour at RT. The plates were again washed and bound antibody was detected by incubation with peroxidase conjugated rabbit anti-human kappa (DAKO, Denmark) in a final dilution of 1:500 in IB. After incubation for lh at RT and a subsequent washing step, the plates were developed with ready-to-use ABTS® solution (Roche Diagnostics GmbH, DE) at RT in the dark. Absorbance was measured at 405 nm after absorbance of the highest concentration reached a sufficient OD,
All antibodies against IL-13Ralphal tested were able to bind to immobilized extracelluar domains of human IL-13Ral, but not neither to hIL-13Ra2 nortohlL-4Ra (Fig. 4).
Example 15
Crossreactivity of HuMab with murine LL-13Ral
Chimeric protein murine IL-13Ral:hFc (R&D Systems, UK) was dissolved in PBS (1 ng/ml) and allowed to adsorb to microtiter plates (NUNC Maxisorb) by incubation over night at 4°C. After washing the plates with washing buffer (WB = 0.9 % NaCl; 0.1% Tween® 20), unspecific binding sites were blocked by addition of 100 fil incubation buffer (IB = PBS with 1% Crotein C and 0.1% Tween® 20) and incubation for 30 min at room-temperature (RT). Then, serially diluted HuMab and control antibodies (HuMab anti-KLH and polyclonal goat anti-hIL-13Ral (R&D Systems)) were added to the wells (100 nl /well; dilutions in IB) and incubated for 1 hour at RT. The plates were again washed and bound human antibodies were detected by incubation with peroxidase conjugated rabbit anti-human kappa (DAKO, Denmark) in a final dilution of 1:500 in IB. Goat anti-hlL-13Ral antibodies bound to the plates were detected by peroxidase-conjugated donkey anti-goat IgG (Santa Cruz; 1:1000 in IB). After incubation for lh at RT and a subsequent washing step, the plates were developed with ready-to-use ABTS® solution (Roche Diagnostics GmbH, DE) at RT in the dark. Absorbance was measured at 405 nm after absorbance of the highest concentration reached a sufficient OD (Fig. 5).

Example 16
Crossreactivity of HuMab with Cynomolgus IL-13Ral
The gene coding for IL-13Ral was isolated by RT-PCR from Cynomolgus tissue and transfected into in the murine cell line Ba/F3. In order to test whether the HuMabs crossreact with Cynomolgus IL-13Ral, the stably transfected Ba/F3 cells as well as the parental Ba/F3 cells were incubated with 10 jig/ml of HuMab and control antibodies. As positive control, a polyclonal goat-anti hIL-13Rctl(R&D Systems) was used. Negative controls included were: a human IgGl myeloma protein (Nordic) and normal goat serum. Bound antibodies were detected by FACS analysis using an antibody directed against human IgG conjugated with FITC to detect HuMabs and an antibody directed against goat IgG conjugated with FITC to detect goat antibodies. Mean fluorescence intensities (MFI) were compared for the individual antibodies tested on the transfected Ba/F3 line versus the parental line.
All HuMabs of the invention were able to bind to Cynomolgus IL-13Ral expressed in transfected Ba/F3 cells. As expected from the close homology between human and Cynomolgus IL-13Ral, the polyclonal AF152 antibody also bound to the Cynomolgus IL-13Ral. The negative control antibodies showed only a marginal increase in MFI when tested with the transfected Ba/F3 cell line (Table 5).


Example 17
Binding of IL-13Ral HuMabs to Fey receptors (binding to FcyRIIIa on NK cells)
To determine the ability of the antibodies of the invention to bind to FcyRIIIa (CD 16) on Natural Killer (NK) cells, Peripheral Blood Mononuclear Cells (PBMCs) were isolated and incubated with 20 jig/ml of HuMab antibody and control antibodies in the presence or absence of 20 ng/ml of a blocking mouse antibody to FcyRIIIa (anti-CD 16, clone 3G8, RDI, Flanders, NJ), to verify binding via FcyRIIIa. As negative controls, human IgG2 and IgG4 (The Binding Site), that do not bind FcyRIIIa, were used. Human IgGl and IgG3 (The Binding Site) were included as positive controls for FcyRIIIa binding. Bound antibodies on NK cells were detected by FACS analysis using a PE-labeled mouse anti-human CD56 (NK-cell surface marker) antibody (BD Biosciences Pharmingen, San Diego, CA) in combination with a FITC-labeled goat F(ab)2 anti-human IgG (Fc) antibody (Protos immunoresearch, Burlingame, CA). Maximum binding at 20 fig/ml (Bmax: MFI±st.dev) of the HuMab tested was determined.

LC5002-005 was able to bind to FeyRIIla efficiently (comparable to the control lgGl antibody) as indicated by a Bmax (MFI) value of 580.6±245.8. Addition of a blocking antibody against FeyRIIla dramatically reduced binding of LC5002-005 to NK cells (Bmax (MFI) value of 260.4±95.90) indicating specific binding to FeyRIIla.

List of References
Aikawa, M., et al., Cytokine 13 (2001) 75-84
Aplin, J.D., and Wriston, J.C. Jr., CRC Crit. Rev. Biochem. (1981) 259-306
Ausubel, F., et al., ed. Current Protocols in Molecular Biology, Greene Publishing
and Wiley Interscience, New York (1987) Barnes, L.M., et al., Cytotechnology 32 (2000) 109-123 Barnes, L.M., et al., Biotech. Bioeng. 73 (2001) 261-270 Berge, S.M., et al., J. Pharm. Sci. 66 (1977) 1-19 Boerner, P., et al., J. Immunol. 147 (1991) 86-95 Brueggemann M., et al., J. Exp. Med. 166 (1987) 1351-1361 Bruggemann, M., et al., Year Immunol. 7 (1993) 33-40 Brunhouse, R., and Cebra, J.J., Mol. Immunol. 16 (1979) 907-917 Burton, D.R., et al., Nature 288 (1980) 338-344 Carter, P., et al., Proc. Natl. Acad. Sci. USA 89 (1992) 4285-4289 Chen, J., et al., International Immunology 5 (1993) 647-656 Chen, J., et al., EMBO J. 12 (1993) 821-830 Choi, T.K., et al., Nature Genetics 4 (1993) 117-123
Cole et al., Monoclonal Antibodies and Cancer Therapy, Alan R. Liss (1985) p. 77 Durocher, Y., et al., Nicl. Acids. Res. 30 (2002) E9 Edelman, G.M., et al., Proc. Natl. Acad. Sci. USA 63 (1969) 78-85 Edge, A.S., et al., Anal. Biochem. 118 (1981) 131-137 EP 0 307 434
Fishwild, D.M., et al., Nat. Biotechnol. 14 (1996) 845-851 Geisse, S., et al., Protein Expr. Purif. 8 (1996) 271-282 Graber, P., et al., Eur. J. Immunol. 28 (1998) 4286-4298 Harding, F., and Lonberg, N., Ann. N. Acad. Sci 764 (1995) 536-546 Hezareh, M., etal., J. Virol. 75 (2001) 12161-12168 Hoogenboom, H.R., and Winter, G., J. Mol. Biol. 227 (1992) 381-388 Idusogie, E.E., et al, J. Immunol. 164 (2000) 4178-4184 Jakobovits, A., et al., Proc. Natl. Acad. Sci. USA 90 (1993) 2551-2555 Jakobovits, A., et al., Nature 362 (1993) 255-258 Jones, P., et al., Nature 321 (1986) 522-525 Kabat, E.A., et al., (1991) Sequences of Proteins of Immunological Interest, Fifth
Ed., Public Health Service, National Institutes of Health, Bethesda, MD.
(1991), NIH Publication No. 91-3242 Kaufman, R.J., Mol. Biotechnol. 16 (2000) 151-161 Lonberg, N., et al., Nature 368 (1994)856-859

Lonberg, N., Handbook of Experimental Pharmacology 113 (1994) 49-101
Lonberg, N., and Huszar, D., Intern. Rev. Immunol. 25 (1995) 65-93
Love, T.W., et al., Methods Enzymol. 178 (1989) 515-527
Lukas, T.J., et al., J. Immunol. 127 (1981) 2555-2560
Makrides, S.C., Protein Expr. Purif. 17 (1999) 183-202
Marks, J.D., et al., J. Mol. Biol. 222 (1991) 581-597
Meissner, P., et al., Biotechnol Bioeng 75 (2001) 197-203
Morgan, A., et al., Immunology 86 (1995) 319-324
Morrison, S.L., et al., Proc. Natl. Acad. Sci. USA 81 (1984) 6851-6855
Neuberger, M.S., EMBO J. 2 (1983) 1373-1378
Norderhaug, L., et al., J. Immunol. Methods 204 (1997) 77-87
Obiri, N.I., et al, J. Biol. Chem. 270 (1995) 8797-8804
Orlandi, R., et al., Proc. Natl. Acad. Sci. USA 86 (1989) 3833-3837
Picard, D., and Schaffiier, W., Nature 307 (1984) 80-82
Poudrier, J., et al., J. Immunol. 30 (2000) 3157-3164
Poudrier J., et al., J. Immunol., 163 (1999) 1153-1161
Queen, C, et al., Proc. Natl. Acad. Sci. USA 86 (1989) 10029-10033
Riechmann, L., et al., Nature 332 (1988) 323-327
Schlaeger, E.-J., and Christensen, K., Cytotechnology 30 (1999) 71-83
Schlaeger, E.-J., J. Immunol. Methods 194 (1996) 191-199
Sojahr, H.T., and Bahl, O.P., Arch. Biochem. Biophys. 259 (1987) 52-57
SwissProt No. 009030
SwissProt No. P24394
SwissProt No. P35225
SwissProt No. P78552
SwissProt No. Q14627
Taylor, L., et al., Nucleic Acids Research 20 (1992) 6287-6295
Taylor, L., et al., Int. Immunol. 6 (1994) 579-591
Thommesen, J.E., et al., Mol. Immunol. 37 (2000) 995-1004
Thotakura, N.R., and Bahl, O.P., Meth. Enzymol. 138 (1987) 350-359
Tuaillon, N., et al., Proc. Natl. Acad. Sci USA 90 (1993) 3720-3724
Tuaillon, N., et al., Immunol. 152 (1994) 2912-2920
US Patent No. 4,640,835
US Patent No. 4,496,689
US Patent No. 4,301,144
US Patent No. 4,670,417
US Patent No. 4,791,192
US Patent No. 4,179,337

US Patent No. 5,202,238
US Patent No. 5,204,244
US Patent No. 5,545,806
US Patent No. 5,545,807
US Patent No. 5,569,825
US Patent No. 5,625,126
US Patent No. 5,633,425
US Patent No. 5,661,016
US Patent No. 5,770,429
US Patent No. 5,789,650
US Patent No. 5,814,318
US Patent No. 5,874,299
US Patent No. 5,877,397
van Dijk, M.A., and van de Winkel, J.G., Curr. Opin. Chem. Biol 5 (2001) 368-374
Werner, R.G., Drug Res. 48 (1998) 870-880
WO 87/05330
WO 92/22645
WO 92/03918
WO 93/1227
WO 94/25585
WO 96/29417
WO 97/15663
WO 98/24884
WO 01/14424
WO 03/080675










Patent Claims
1. An antibody, characterized in that the variable heavy chain amino acid sequence CDR3 of said antibody is selected from the group consisting of the heavy chain CDR3 sequences of SEQ ID NO: 1, 3, 5, 7 or 9.
2. An antibody according to claim 1, characterized in that the variable heavy chain amino acid sequences CDR1 and CDR2 of said antibody are selected from the group consisting of the heavy chain CDR1 and CDR2 sequences of SEQ ID NO: 1,3, 5, 7 or 9.
3. An antibody according to claim 1 or 2, characterized in that the variable light chain amino acid sequences CDR1, CDR2 and CDR3 of said antibody are selected from the group consisting of the variable light chain amino acid sequences CDR1, CDR2 and CDR3 of SEQ ID NO: 2, 4, 6, 8 or 10.
4. An antibody according to claim 1 to 3, characterized in comprising a human yl -heavy chain comprising

a) amino acid sequence Pro234Val234Ala235 with deletion of Gly236 and/or amino acid sequence Gly327Leu32sPro329Ser33oSer33i,
b) amino acid sequence Ala234Ala235 or
c) amino acids Ala265 and Ala297-

5. An antibody according to claim 1 to 4, characterized in being a human antibody.
6. An antibody according to claim 1 to 5, characterized by an affinity of 10"9 M (KD) or less, preferably of 10"9 to 10"13 M for binding to IL-13Ral.
7. An antibody according to claims 1 to 6 obtained from hybridoma cell line DSM ACC2709, DSM ACC2710, DSM ACC2711 or DSM ACC2712.
8. An antibody according to claim 1 to 7, characterized in comprising as heavy chain variable region SEQ ID NO: 1 and as light chain variable region SEQ

ID NO: 2, as heavy chain variable region SEQ ID NO: 3 and as light chain variable region of SEQ ID NO: 4, as heavy chain variable region SEQ ID NO: 5 and as light chain variable region SEQ ID NO: 6, as heavy chain variable region SEQ ID NO: 7 and as light chain variable region SEQ ID NO: 8 or as heavy variable region SEQ ID NO: 9 and as light chain variable region SEQ ID NO: 10.
9. An antibody according to claim 1 to 6, characterized in comprising
a) as heavy chain variable region SEQ ID NO: I, as light chain variable
region SEQ ID NO: 2, as K light chain constant region SEQ ID NO: 11 and as y1 heavy chain constant region SEQ ID NO: 12 optionally with mutations L234A and L235A or D265A and N297A,
b) as heavy chain variable region SEQ ID NO: 3 and as light chain variable
region of SEQ ID NO: 4, as K light chain constant region SEQ ID NO: 11 and as yl heavy chain constant region SEQ ID NO: 12 optionally with mutations L234A and L235A or D265A and N297A,
c) as heavy chain variable region SEQ ID NO: 5 and as light chain variable
region SEQ ID NO: 6, as K light chain constant region SEQ ID NO: 11 and as yl heavy chain constant region SEQ ID NO: 12 optionally with mutations L234A and L235A or D265A and N297A,
d) as heavy chain variable region SEQ ID NO: 7 and as light chain variable
region SEQ ID NO: 8, as K light chain constant region SEQ ID NO: 11 and as yl heavy chain constant region SEQ ID NO: 12 optionally with mutations L234A and L235A or D265A and N297A, or
e) as heavy variable region SEQ ID NO: 9 and as light chain variable region
SEQ ED NO: 10, as K light chain constant region SEQ ID NO: 11 and as yl heavy chain constant region SEQ ID NO: 12 optionally with mutations L234A and L235A or D265A and N297A.
10. The use of an antibody according to claims 1 to 9 for the manufacture of a
pharmaceutical composition.

11. A pharmaceutical composition comprising an antibody according to claims 1 to 9 in a pharmaceutical]}/ effective amount.
12. A recombinant host cell capable of producing a recombinant antibody according to claim 1 to 9.
13. Method for the manufacture of a pharmaceutical composition comprising a pharmaceutically effective amount of an antibody according to claims 1 to 9.
14. A nucleic acid encoding a polypeptide capable of assembling together with the respectively other antibody chain defined below, whereas said polypeptide is either

a) an antibody heavy chain comprising heavy chain CDRs of SEQIDNO:l,3,5,7or9;
b) an antibody light chain comprising light chain CDRs of SEQ ID NO:2, 4, 6, 8 or 10
wherein the CDRs are selected independently of each other.
15. An expression vector comprising a nucleic acid according to claim 14, capable of expressing said nucleic acid in a prokaryotic or eukaryotic host cell.
16. A prokaryotic or eukaryotic host cell comprising the vector according to claim 15.
17. A method for the production of a polypeptide binding to IL-13Ral and inhibiting the binding of IL-13to IL-13Ral, characterized by expressing a nucleic acid encoding a heavy chain and a nucleic acid encoding a light chain according to claim 14 in a prokaryotic or eukaryotic host cell and recovering said polypeptide from said cell.

18. Method for the treatment of a patient in need of an asthma or antiallergic therapy, characterized by administering to the patient a therapeutically effective amount of an antibody according to claims 1 to 9.
19. Method for the preparation of a pharmaceutical composition characterized in producing an antibody according to claims 1 to 9 by means of recombinant expression, recovering said antibody and combining said antibody with a pharmaceutical acceptable buffer and/or adjuvant.
20. Hybridoma cell line DSM ACC2709, DSM ACC2710, DSM ACC2711 or
DSM ACC2712 Arr97i?

Documents:

2964-CHENP-2007 CORRESPONDENCE OTHERS 17-08-2011.pdf

2964-CHENP-2007 CORRESPONDENCE OTHERS 25-01-2012.pdf

2964-CHENP-2007 FORM-3 25-01-2012.pdf

2964-CHENP-2007 OTHER PATENT DOCUMENT 25-01-2012.pdf

2964-CHENP-2007 AMENDED PAGES OF SPECIFICATION 02-01-2012.pdf

2964-CHENP-2007 AMENDED CLAIMS 02-01-2012.pdf

2964-CHENP-2007 EXAMINATION REPORT REPLY RECEIVED 02-01-2012.pdf

2964-CHENP-2007 POWER OF ATTORNEY 02-01-2012.pdf

2964-chenp-2007-abstract.pdf

2964-chenp-2007-claims.pdf

2964-chenp-2007-correspondnece-others.pdf

2964-chenp-2007-description(complete).pdf

2964-chenp-2007-drawings.pdf

2964-chenp-2007-form 1.pdf

2964-chenp-2007-form 26.pdf

2964-chenp-2007-form 3.pdf

2964-chenp-2007-form 5.pdf

2964-chenp-2007-pct.pdf


Patent Number 250648
Indian Patent Application Number 2964/CHENP/2007
PG Journal Number 03/2012
Publication Date 20-Jan-2012
Grant Date 16-Jan-2012
Date of Filing 03-Jul-2007
Name of Patentee F. HOFFMANN-LA ROCHE AG
Applicant Address 124 GRENZACHERSTRASSE, CH-4070 BASEL,
Inventors:
# Inventor's Name Inventor's Address
1 ENDL, JOSEF ULMENSTRASSE 12, 82362 WEILHEIM,
2 FUENTES, MARIA, ELENA 1490 BITTERN DRIVE, SUNNYVALE, CA 94087, USA
3 GRAUS, YVO WERDORPERWAARD 3, NL-3984 PR ODIJK, THE NETHERLANDS
4 GROSSMANN, ADELBERT HEIMGARTEN-STRASSE 1, 82436 EGLFING, GERMANY
5 NEUMANN, SEBASTIAN EISVOGELSTRASSE 25, D-82362 WEILHEIM, GERMANY
6 PARREN, PAUL WEDORPERWAARD 17, NL-3984 PR ODIJK, THE NETHERLANDS
7 REBERS, FRANK ADRIAAN MENNICKKWARTIER 34, NL-3554 CT UTRECHT, THE NETHERLANDS
8 SCHUMACHER, RALF HOCHFELDSTRASSE 78, D-82377 PENZBERG, GERMANY
9 SEEBER, STEFAN BREUNETSRIEDER WEG 22, D-82377 PENZBERG, GERMANY
10 STRACKE, JAN BRUNECKERSTRASSE 7, D-81373 MUENCHEN, GERMANY
11 STUBENRAUCH, KAY-GUNNAR AN DER FREIHEIT 105, D-82377 PENZBERG, GERMANY
12 VAN DE WINKEL, JAN VERLENGDE SLOTLAAN 80, NL-3707 CK ZEIST, THE NETHERLANDS
13 VAN VUGT, MARTINE FLUWEELMOS 6, NL-3994 KR HOUTEN, THE NETHERLANDS
14 VEREECKEN-VERPLOEGEN, SANDRA HESSELAAN 19, NL-3431 CD NIEUWEGEIN, THE NETHERLANDS
15 REGULA, JORG LIERSTRASSE 15, D-80639 MUENCHEN, GERMANY
PCT International Classification Number C07K 16/28
PCT International Application Number PCT/EP06/00005
PCT International Filing date 2006-01-02
PCT Conventions:
# PCT Application Number Date of Convention Priority Country
1 05000003.3 2005-01-03 EUROPEAN UNION
2 05002229.2 2005-02-03 EUROPEAN UNION