AU2011200308A1 - Human antibody molecules for IL-13 - Google Patents

Human antibody molecules for IL-13 Download PDF

Info

Publication number
AU2011200308A1
AU2011200308A1 AU2011200308A AU2011200308A AU2011200308A1 AU 2011200308 A1 AU2011200308 A1 AU 2011200308A1 AU 2011200308 A AU2011200308 A AU 2011200308A AU 2011200308 A AU2011200308 A AU 2011200308A AU 2011200308 A1 AU2011200308 A1 AU 2011200308A1
Authority
AU
Australia
Prior art keywords
domain
seq
binding member
group
antibody
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
AU2011200308A
Other versions
AU2011200308B2 (en
Inventor
Lutz Jermutuz
Ralph Raymond Minter
Phillip David Monk
Celia Patricia Shorrock
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
MedImmune Ltd
Original Assignee
MedImmune Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from GBGB0407315.1A external-priority patent/GB0407315D0/en
Application filed by MedImmune Ltd filed Critical MedImmune Ltd
Priority to AU2011200308A priority Critical patent/AU2011200308B2/en
Publication of AU2011200308A1 publication Critical patent/AU2011200308A1/en
Application granted granted Critical
Publication of AU2011200308B2 publication Critical patent/AU2011200308B2/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/244Interleukins [IL]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Description

AUSTRALIA Patents Act 1990 FB RICE & CO Patent and Trade Mark Attorneys MedImmune Limited COMPLETE SPECIFICATION STANDARD PATENT Invention Title: Human antibody molecules for IL-13 The following statement is a full description of this invention including the best method of performing it known to us: 1 HUMAN ANTIBODY MOLECULES FOR IL-13 CROSS-REFERENCE TO RELATED APPLICATIONS This application is a divisional applicant pursuant to Section 5 79B of the Patents Act 1990 of Australian Patent Application No. 2004256976 filed July 15, 2004 which corresponds to International Application No. PCT/GB2004/003059 filed July 15, 2004 in the Australian national phase, which claims priority from GB0407315.1 filed March 31, 2004, USSN 60/573,791 filed L0 May 24, 2004, USSN 60/487,512 filed July 15, 2003 and USSN 60/558,216 filed March 31, 2004, the contents of which are incorporated herein by reference in their entirety. The present invention relates to specific binding members, in L5 particular human anti-IL-13 antibody molecules and especially those which neutralise IL-13 activity. It further relates to methods for using anti-IL-13 antibody molecules in diagnosis or treatment of IL-13 related disorders, including asthma, atopic dermatitis, allergic rhinitis, fibrosis, inflammatory ?0 bowel disease and Hodgkin's lymphoma. Preferred embodiments of the present invention employ the antibody VH and/or VL domain of the antibody molecule herein termed BAK502G9 and other antibody molecules of the BAK502G9 25 lineage and of the BAK278D6 lineage, as herein defined. Further preferred embodiments employ complementarity determining regions (CDRs) of the BAK278D6 lineage, and preferably BAK502G9, especially VH CDR3 in other antibody framework regions. Further aspects of the present invention 30 provide for compositions containing specific binding members of the invention, and their use in methods of inhibiting or neutralising IL-13, including methods of treatment of the human or animal body by therapy. 2 The present invention provides antibody molecules of particular value in binding and neutralising IL-13, and thus of use in any of a variety of therapeutic treatments, as indicated by the experimentation contained herein and further 5 by the supporting technical literature. Interleukin (IL)-13 is a 114 amino acid cytokine with an unmodified molecular mass of approximately 12 kDa [1,2]. IL-13 is most closely related to IL-4 with which it shares 30% LO sequence similarity at the amino acid level. The human IL-13 gene is located on chromosome 5q31 adjacent to the IL-4 gene [1][2]. This region of chromosome Sq contains gene sequences for other Th2 lymphocyte derived cytokines including GM-CSF and IL-5, whose levels together with IL-4 have been shown to [5 correlate with disease severity in asthmatics and rodent models of allergic inflammation [3][4][5][6][7][8]. Although initially identified as a Th2 CD4+ lymphocyte derived cytokine, IL-13 is also produced by Th1 CD4+ T-cells, CD8+ T 20 lymphocytes NK cells, and non-T-cell populations such as mast cells, basophils, eosinophils, macrophages, monocytes and airway smooth muscle cells. IL-13 is reported to mediate its effects through a receptor 25 system that includes the IL-4 receptor ax chain (IL-4Rx), which itself can bind IL-4 but not IL-13, and at least two other cell surface proteins, IL-13Ral and IL-13R2 [9][10]. IL-13Ral can bind IL-13 with low affinity, subsequently recruiting IL 4Ra to form a high affinity functional receptor that signals 30 [11][12]. The Genbank database lists the amino acid sequence and the nucleic acid sequence of IL-13Ral as NP_001551 and Y10659 respectively. Studies in STAT6 (signal transducer and activator of transcription 6) -deficient mice have revealed that IL-13, in a manner similar to IL-4, signals by utilising 3 the JAK-STAT6 pathway [13][14]. IL-13Ra2 shares 37% sequence identity with IL-13Ral at the amino acid level and binds IL-13 with high affinity [15][16]. However, IL-13Ra2 has a shorter cytoplasmic tail that lacks known signalling motifs. Cells 5 expressing IL-13Ra2 are not responsive to IL-13 even in the presence of IL-4Ra [17]. It is postulated, therefore, that IL 13Ra2 acts as a decoy receptor regulating IL-13 but not IL-4 function. This is supported by studies in IL-13Ra2 deficient mice whose phenotype was consistent with increased .0 responsiveness to IL-13 [18][19]. The Genbank database lists the amino acid sequence and the nucleic acid sequence of IL 13Ra2 as NP_000631 and Y08768 respectively. The signalling IL-13Ral/IL-4Ra receptor complex is expressed .5 on human B-cells, mast cells, monocyte/macrophages, dendritic cells, eosinophils, basophils, fibroblasts, endothelial cells, airway epithelial cells and airway smooth muscle cells. Bronchial asthma is a common persistent inflammatory disease !0 of the lung characterised by airways hyper-responsiveness, mucus overproduction, fibrosis and raised serum IgE levels. Airways hyper-responsiveness (AHR) is the exaggerated constriction of the airways to non-specific stimuli such as cold air. Both AHR and mucus overproduction are thought to be 25 responsible for the variable airway obstruction that leads to the shortness of breath characteristic of asthma attacks (exacerbations) and which is responsible for the mortality associated with this disease (around 2000 deaths/year in the United Kingdom). 30 The incidence of asthma, along with other allergic diseases, has increased significantly in recent years [20][21]. For 4 example, currently, around 10% of the population of the United Kingdom (UK) has been diagnosed as asthmatic. Current British Thoracic Society (BTS)and Global Initiative 5 for Asthma (GINA) guidelines suggest a stepwise approach to the treatment of asthma [22, 23]. Mild to moderate asthma can usually be controlled by the use of inhaled corticosteroids, in combination with beta-agonists or leukotriene inhibitors. However, due to the documented side effects of LO corticosteroids, patients tend not to comply with the treatment regime which reduces the effectiveness of treatment [24-26]. There is a clear need for new treatments for subjects with L5 more severe disease, who often gain very limited benefit from either higher doses of inhaled or oral corticosteroids recommended by asthma guidelines. Long term treatment with oral corticosteroids is associated with side effects such as osteoporosis, slowed growth rates in children, diabetes and ?0 oral candidiasis [88]. As both beneficial and adverse effects of corticosteroids are mediated via the same receptor, treatment is a balance between safety and efficacy. Hospitalisation of these patients, who represent around 6% of the UK asthma population, as a result of severe exacerbations 25 accounts for the majority of the significant economic burden of asthma on healthcare authorities [89]. It is believed that the pathology of asthma is caused by ongoing Th2 lymphocyte mediated inflammation that results from 30 inappropriate responses of the immune system to harmless antigens. Evidence has been accrued which implicates IL-13, rather than the classical Th2 derived cytokine IL-4, as the key mediator in the pathogenesis of established airway disease. 5 Administration of recombinant IL-13 to the airways of naive non-sensitised rodents caused many aspects of the asthma phenotype including airway inflammation, mucus production and 5 AHR [27][28][29][30]. A similar phenotype was observed in a transgenic mouse in which IL-13 was specifically overexpressed in the lung. In this model more chronic exposure to IL-13 also resulted in fibrosis [31]. 10 Further, in rodent models of allergic disease many aspects of the asthma phenotype have been associated with IL-13. Soluble murine IL-13Ra2, a potent IL-13 neutraliser, has been shown to inhibit AHR, mucus hypersecretion and the influx of inflammatory cells which are characteristics of this rodent L5 model [27][28][30]. In complementary studies, mice in which the IL-13 gene had been deleted, failed to develop allergen induced AHR. AHR could be restored in these IL-13 deficient mice by the administration of recombinant IL-13. In contrast, IL-4 deficient mice developed airway disease in this model 20 [32][33]. Using a longer-term allergen-induced pulmonary inflammation model, Taube at al. demonstrated the efficacy of soluble murine IL-13Ra2 against established airway disease [34]. 25 Soluble murine IL-13Ra2 inhibited AHR, mucus overproduction and to a lesser extent airway inflammation. In contrast, soluble IL-4Ra, which binds and antagonises IL-4, had little effect on AHR or airway inflammation in this system [35]. These findings were supported by Blease et al. who developed a 30 chronic fungal model of asthma in which polyclonal antibodies against IL-13 but not IL-4 were able to reduce mucus overproduction, AHR and subepithelial fibrosis [36]. 6 A number of genetic polymorphisms in the IL-13 gene have also been linked to allergic disease. In particular, a variant of the IL-13 gene in which the arginine residue at amino acid 130 is substituted with glutamine (R130Q) has been associated with 5 bronchial asthma, atopic dermatitis and raised serum IgE levels [37][38][39][40]. This particular IL-13 variant is also referred to as the Q11OR variant (arginine residue at amino acid 110 is substituted with glutamine) by some groups who exclude the 20 amino acid signal sequence from the amino acid 10 count. Arima et al, [41] report that this variant is associated with raised levels of IL-13 in serum. The IL-13 variant (R130Q) and antibodies to this variant are discussed in WO 01/62933. An IL-13 promoter polymorphism, which alters IL-13 production, has also been associated with allergic 15 asthma [42]. Raised levels of IL-13 have also been measured in human subjects with asthma, atopic rhinitis (hay fever), allergic dermatitis (eczema) and chronic sinusitis. For example levels 20 of IL-13 were found to be higher in bronchial biopsies, sputum and broncho-alveolar lavage (BAL) cells from asthmatics compared to control subjects [43][44][45][46]. Further, levels of IL-13 in BAL samples increased in asthmatic individuals upon challenge with allergen [47][48]. The IL-13 production 25 capacity of CD4(+) T cells has further been shown to be useful marker of risk for subsequent development of allergic disease in newborns [49] Li et al [114] have recently reported affects of a 30 neutralising anti-mouse IL-13 antibody in a chronic mouse model of asthma. Chronic asthma-like response (such as AHR, severe airway inflammation, hyper mucus productions) was induced in OVA sensitised mice. Li et al report that administration of an IL-13 antibody at the time of each OVA 7 challenge suppresses AHR, eosinophil infiltration, serum IgE levels, proinflammatory cytokine/chemokine levels and airway remodelling [14]. 5 In summary, these data provide indication that IL-13 rather than IL-4 is a more attractive target for the treatment of human allergic disease. IL-13 may play a role in the pathogenesis of inflammatory 10 bowel disease. Heller et al.[116] report that neutralisation of IL-13 by administration of soluble IL-13Ra2 ameliorated colonic inflammation in a murine model of human ulcerative colitis [116]. Correspondingly, IL-13 expression was higher in rectal biopsy specimens from ulcerative colitis patients when 15 compared to controls [117]. Aside from asthma, IL-13 has been associated with other fibrotic conditions. Increased levels of IL-13, up to a 1000 fold higher than IL-4, have been measured in the serum of 20 patients with systemic sclerosis [50] and in BAL samples from patients affected with other forms of pulmonary fibrosis [51]. Correspondingly, overexpression of IL-13 but not IL-4 in the mouse lung resulted in pronounced fibrosis [52][53]. The contribution of IL-13 to fibrosis in tissues other than the 25 lung has been extensively studied in a mouse model of parasite-induced liver fibrosis. Specific inhibition of IL-13 by administration of soluble IL-13Ra2 or IL-13 gene disruption, but not ablation of IL-4 production prevented fibrogenesis in the liver [54][55][56]. 30 Chronic Obstructive Pulmonary Disease (COPD) includes patient populations with varying degrees of chronic bronchitis, small airway disease and emphysema and is characterised by 8 progressive irreversible lung function decline that responds poorly to current asthma based therapy [90]. The incidence of COPD has risen dramatically in recent years to become the fourth leading cause of death worldwide (World 5 Health Organisation). COPD therefore represents a large unmet medical need. The underlying causes of COPD remain poorly understood. The "Dutch hypothesis" proposes that there is a common l0 susceptibility to COPD and asthma and therefore, that similar mechanisms may contribute to the pathogenesis of both disorders [57). Zheng et al [58] have demonstrated that overexpression of IL L5 13 in the mouse lung caused emphysema, elevated mucus production and inflammation, reflecting aspects of human COPD. Furthermore, AHR, an IL-13 dependent response in murine models of allergic inflammation, has been shown to be predictive of lung function decline in smokers [59]. A link has also been ?0 established between an IL-13 promoter polymorphism and susceptibility to develop COPD [60]. The signs are therefore that IL-13 plays an important role in the pathogenesis of COPD, particularly in patients with 25 asthma-like features including AHR and eosinophilia. mRNA levels of IL-13 have been shown to be higher in autopsy tissue samples from subjects with a history of COPD when compared to lung samples from subjects with no reported lung disease (J. Elias, Oral communication at American Thoracic Society Annual 30 Meeting 2002). In another study, raised levels of IL-13 were demonstrated by immunohistochemistry in peripheral lung sections from COPD patients [91]. 9 Hodgkin's disease is a common type of lymphoma, which accounts for approximately 7,500 cases per year in the United States. Hodgkin's disease is unusual among malignancies in that the neoplastic Reed-Sternberg cell, often derived from B-cells, 5 make up only a small proportion of the clinically detectable mass. Hodgkin's disease-derived cell lines and primary Reed Sternberg cells frequently express IL-13 and its receptor [61]. As IL-13 promotes cell survival and proliferation in normal B-cells, it was proposed that IL-13 could act as a 0 growth factor for Reed-Sternberg cells. Skinnider et al. have demonstrated that neutralising antibodies against IL-13 can inhibit the growth of Hodgkin's disease-derived cell lines in vitro [62]. This finding suggested that Reed-Sternberg cells might enhance their own survival by an IL-13 autocrine and .5 paracrine cytokine loop. Consistent with this hypothesis, raised levels of IL-13 have been detected in the serum of some Hodgkin's disease patients when compared to normal controls [63]. IL-13 inhibitors may therefore prevent disease progression by inhibiting proliferation of malignant Reed !0 Sternberg cells. Many human cancer cells express immunogenic tumour specific antigens. However, although many tumours spontaneously regress, a number evade the immune system (immunosurveillance) 25 by suppressing T-cell mediated immunity. Terabe et al.[64] have demonstrated a role of IL-13 in immunosuppression in a mouse model in which tumours spontaneously regress after initial growth and then recur. Specific inhibition of IL-13, with soluble IL-13Rx2, protected these mice from tumour 30 recurrence. Terabe et al [64) went on to show that IL-13 suppresses the differentiation of tumour specific CD8+ cytotoxic lymphocytes that mediate anti-tumour immune responses. 10 IL-13 inhibitors may, therefore, be used therapeutically to prevent tumour recurrence or metastasis. Inhibition of IL-13 has been shown to enhance anti-viral vaccines in animal models and may be beneficial in the treatment of HIV and other 5 infectious diseases [65]. It should be noted that generally herein reference to interleukin-13 or IL-13 is, except where context dictates otherwise, reference to human IL-13. This is also referred to LO in places as "the antigen". The present invention provides antibodies to human IL-13, especially human antibodies, that are cross-reactive with non-human primate IL-13, including cynomolgus and rhesus monkey IL-13. Antibodies in accordance with some embodiments of the present invention recognise a L5 variant of IL-13 in which the arginine residue at amino acid position 130 is replaced by glutamine. In other aspects and embodiments the present invention provides specific binding members against murine IL-13, specifically mouse IL-13. 0 Brief Description of Figures Figure 1 shows neutralisation potency (% inhibition) of BAK167All (closed squares) and its derivative BAK615E3 (open squares) as scFv against 25 ng/ml human IL-13 in the TF-1 cell 25 proliferation assay. The triangles represent an irrelevant scFv. Data represent the mean with standard error bars of triplicate determinations within the same experiment. Figure 2 shows the neutralisation potency (% inhibition) of 30 BAK278D6 (closed squares) and its derivative BAK502G9 (open squares) as scFv against 25 ng/ml human IL-13 in the TF-1 cell proliferation assay. The triangles represent an irrelevant scFv. Data represent the mean with standard error bars of triplicate determinations within the same experiment. 11 Figure 3 shows the neutralisation potency (% inhibition) of BAK209Bll (closed squares) as a scFv against 25 ng/ml murine IL-13 in the TF-1 cell proliferation assay. The triangles 5 represent an irrelevant scFv. Data represent the mean with standard error bars of triplicate determinations within the same experiment. Figure 4 shows the neutralisation potency (% inhibition) of LO BAK278D6 (closed squares) as a scFv against IL-13 in the TF-1 cell proliferation assay. The triangles represent an irrelevant scFv. Data represent the mean with standard error bars of triplicate determinations within the same experiment. Figure 4A show potency against 25 ng/ml human IL-13. L5 Figure 4B shows potency against 25 ng/ml human variant IL-13. Figure 4C shows potency against 50 ng/ml non-human primate IL-13. !0 Figure 5 shows a comparison of the potency of anti-human IL-13 antibodies in the TF-1 proliferation assay. Data represent the mean neutralisation potency with standard error bars over 5-7 experiments against 25 ng/ml human IL-13. The performance relative to the commercially available antibody, B-B13, was 25 evaluated statistically by performing a one-way ANOVA with Dunnett's test. * 2<0.05, ** 2<0.01 compared to B-B13. Figure 6 shows the neutralisation potency (% inhibition) of BAK502G9 (closed squares), BAKll67F2 (closed triangles) and 30 BAKl183H4 (closed inverted triangles) as human IgG4 against tagged IL-13 in the TF-1 cell proliferation assay. Open triangles represent an irrelevant IgG4. Data represent the mean with standard error bars of three separate experiments. Figure 6A shows potency against 25 ng/ml human IL-13. 12 Figure 6B shows potency against 25 ng/ml human variant IL-13. Figure 6C shows potency against 50 ng/ml non-human primate IL-13. 5 Figure 7 shows the neutralisation potency (% inhibition) of BAK502G9 (closed squares), BAK1l67F2 (closed triangles), BAKll83H4 (closed inverted triangles) as human IgG4 and commercial anti-human IL-13 antibodies (B-B13 - open squares; LO JES10-5A2 - open inverted triangles) in the native IL-13 dependent HDLM-2 cell proliferation assay. Open triangles represent an irrelevant IgG4. Data represent the mean with standard error bars of triplicate determinations within the same experiment. L5 Figure 8 shows a comparison of the potency of anti-human IL-13 antibodies in the NHLF assay. Data represent the mean neutralization potency (IC50 pM) with standard error bars over 4-5 experiments against 10 ng/ml human IL-13 in the NHLF 0 eotaxin release assay. The performance relative to the commercially available antibody, B-B13, was evaluated statistically by performing a one-way ANOVA with Dunnett's test. * P<0.05, ** P<0.01 compared to B-B13. 25 Figure 9 shows the neutralisation potency (% inhibition) of BAK502G9 (closed squares), BAK1l67F2 (closed triangles), BAKll83H4 (closed inverted triangles) as human IgG4 against VCAM-1 upregulation on the surface of HUVEC in response to lOng/ml human IL-13. Open triangles represent irrelevant IgG4. 30 Data represent the mean with standard error bars of triplicate determinations within the same experiment. Figure 10 shows the neutralisation potency (% inhibition) of BAK502G9 (closed squares), BAKll67F2 (closed triangles), 13 BAKll83H4 (closed inverted triangles) as human IgG4 against eotaxin release from VCAM-l upregulation on the surface of HUVEC in response to either lng/ml human IL-4 (Figure 10A) or 0.5ng/ml human IL-1 (Figure 10B). Open triangles represent an 5 irrelevant IgG4. Data represent the mean with standard error bars of triplicate determinations within the same experiment. Figure 11 shows the neutralisation potency (% inhibition) of BAK209Bll (squares) as a human IgG4 against 1 ng/ml murine IL LO 13 in the factor dependent B9 cell proliferation assay. Open triangles represent an irrelevant IgG4. Data represent the mean with standard error bars of triplicate determinations within the same experiment. L5 Figure 12 shows the relative level of IL-13 in lung homogenates from sensitised (s) (right-hand bar) and non sensitised (ns) (left-hand bar) mice post challenge in a murine model of acute pulmonary allergic inflammation. The effect of sensitisation was statistically evaluated by performing O Student's t-test using quantity of IL-13 data. *<0.05. **<0.01 compared to non-sensitised control animals (n=5-6 mice). Data represent the mean with standard error bars. Figure 13 illustrates the effects of i.v. administration of 25 BAK209B11 as human IgG4 in different amounts compared to an isotype matched IgG4 irrelevant control antibody on ovalbumin induced leukocyte recruitment to the lung in ovalbumin sensitised mice. The number of leukocytes is shown (x 104). The effect of antibody treatment was statistically evaluated 30 by performing one way ANOVA with Dunnett's test using differential cell count data. *<0.05. **<0.01 compared to ovalbumin challenged PBS control animals (=0% inhibition; n=5 8 mice). Data represent the mean with standard error bars. 14 Figure 14 illustrates the effects of i.v. administration of BAK209Bll as human IgG4 in different amounts compared to an isotype matched IgG4 irrelevant control antibody on ovalbumin induced eosinophil recruitment to the lung in ovalbumin 5 sensitised mice. The number of eosinophils is shown (x 104). The effect of antibody treatment was statistically evaluated by performing one way ANOVA with Dunnett's test using differential cell count data. *<0.05. **<0.01 compared to ovalbumin challenged PBS control animals (=0% inhibition; n=5 LO 8 mice). Data represent the mean with standard error bars. Figure 15 illustrates the effects of i.v. administration of BAK209Bll as human IgG4 in different amounts compared to an isotype matched IgG4 irrelevant control antibody on ovalbumin L5 induced neutrophil recruitment to the lung in ovalbumin sensitised mice. The number of neutrophils is shown (x 104). The effect of antibody treatment was statistically evaluated by performing one way ANOVA with Dunnett's test using differential cell count data. *<0.05. **<0.01 compared to 20 ovalbumin challenged PBS control animals (=0% inhibition; n=5 8 mice). Data represent the mean with standard error bars. Figure 16 illustrates the effects of i.v. administration of BAK209B1l as human IgG4 in different amounts compared to an 25 isotype matched IgG4 irrelevant control antibody on ovalbumin induced lymphocyte recruitment to the lung in ovalbumin sensitised mice. The induction of lymphocytes was dose dependently inhibited by BAK209B1l with maximal inhibition at 3pg/ml of BAK209Bll. The effect of antibody treatment was 30 statistically evaluated by performing one way ANOVA with Dunnett's test using differential cell count data. *<0.05. **<0.01 compared to ovalbumin challenged PBS control animals (=0% inhibition; n=5-8 mice). Data represent the mean with standard error bars. 15 Figure 17 illustrates the effects of i.v. administration of BAK209Bl1 as human IgG4 in different amounts compared to an isotype matched IgG4 irrelevant control antibody on ovalbumin 5 induced monocyte/macrophage recruitment to the lung in ovalbumin sensitised mice. There was no significant increase in the levels of monocytes/macrophages of sensitised animals when compared with control animals. However, such background levels of these cells were depressed by 236pg/ml BAK209Bll in LO sensitised animals. The effect of antibody treatment was statistically evaluated by performing one way ANOVA with Dunnett's test using differential cell count data. *<0.05. **<0.01 compared to ovalbumin challenged PBS control animals (=0% inhibition; n=5-8 mice). Data represent the mean with L5 standard error bars. Figure 18 shows the effects of a commercial anti-IL-13 neutralising antibody JES10-5A2 on the influx of cells (number 20 of leukocytes is shown (x 104)) into the murine airpouch elicited by administration of bacterially derived recombinant human IL-13. The effect of antibody treatment was statistically evaluated by performing one way ANOVA with Dunnett's test using differential cell count data. *<0.05. 25 **<0.01 compared to CMC control animals (=0% inhibition; n=ll 13 mice). Data represent the mean with standard error bars. Figure 19 shows an sequence alignment of cynomolgus IL-13 against human IL-13 amino acid sequences. The seven amino acid 30 residues that differ between human and cynomolgus IL-13 are shaded. Rhesus and cynomolgus IL-13 have an identical amino acid sequence. 16 Figure 20 illustrates the effects of single 10mg/kg i.v bolus dose of BAK502G9 as human IgG4 on serum IgE levels in 4 allergic but non-challenged cynomolgus primates (2 male/2 female) over 29 days. Serum IgE concentration is significantly 5 reduced from 100 % (predose) to 66 ± 10% of control values (p<0.05), at 4 and 5 days after dosing. This lowering of serum IgE concentration recovers to 88 ± 8 % of control levels by day 22. * = p<0.05 as compared to predose IgE levels, repeated measures ANOVA followed by Dunnett's multiple LO comparisons test (n=4 animals). Figure 20B shows relative serum IgE levels of male and female cynomolgus primates versus time following a single 10mg/kg intravenous dose of BAK502G9. Relative serum IgE data are expressed as arithmetic mean ± SEM percentage of baseline L5 value. Figure 21 illustrates the effects of intraperitoneal administration of BAK209Bll in different amounts (H=237pg/day, M=23.7tg/day and L=2.37tg/day) compared with an isotype ?0 matched IgG1 irrelevant control antibody on the lung function of ovalbumin sensitised and challenged mice. In Figure 21A lung function is represented by log PC 50 s (log methacholine concentration required to increase baseline PenH by 50%) before any treatment (day 0) and post sensitisation, challenge 25 and drug treatment (day 25). Figure 21A shows the raw data used to calculate the study endpoint, shown in figure 21B (Delta log PC 50 ) . Data represent the mean with standard error bars of n=8. 30 In Figure 21B changing lung function is shown by a change in an individual mouse's log PC 5 o (delta log PCso) . Delta log PCso is defined as an individuals change in log PC 5 o at day 25 verus day 0. Data represent group mean delta log PC 5 o (individual changes averaged within treatment groups) with standard error 17 bars. The effect of antibody treatment was statistically evaluated by performing one way ANOVA with Dunnett's test using delta log PC 5 o data. **p<0.01 compared to ovalbumin sensitised and challenged control animals (n=8 mice). 5 Figure 22 illustrates the effects of local (i.po.) and systemic (i.v.) administration of BAK502G9 as human IgG4 in different amounts compared to an isotype matched IgG4 irrelevant control antibody on the total leukocyte recruitment LO (Figure 22A) and eosinophil recruitment (Figure 22B) into the air pouch of BALB/C mice. Data represent the mean with standard error bars of n=10. The effect of antibody treatment was statistically evaluated by performing one way ANOVA with Dunnett's test using log-transformed data.. *p<0.05, **p<0.01 [5 compared to huIL-13 challenged mice (n=10). Figure 23: illustrates the effects of i.p. administration of BAK502G9 as human IgG4 compared to an isotype matched IgG4 irrelevant control antibody on the development of AHR !0 following intratracheal administration of human IL-13 to the airways of mice. The effect of antibody treatment was statistically evaluated by performing one way ANOVA with Dunnett's test using PC 200 Methacholine data. *<0.05. **<0.01 compared to the human IL-13 positive control group (n=6-8 25 mice). Data represent the mean with standard error bars. Figure 24 shows the neutralisation potency (% maximal response) of BAK502G9 (closed squares) as IgG4 against 30ng/ml IL-13 in a human B cell IgE production assay. Open squares 30 represent an irrelevant IgG4. Data represent the mean with standard error bars of six donors from separate experiments. Figure 25 shows the effects of BAK502G9 on IL-13 induced potentiation of agonist induced Ca 2 + signalling in bronchial 18 smooth muscle cells. The area under the curve (AUC) of the Ca 2 + signalling response to histamine was determined for each antibody +/- IL-13 pre-treatment condition. Combined data from three independent experiments are shown for irrelevant 5 antibody CAT-001 (a) and BAK502G9 (b) as the percentage difference versus untreated cells of AUC±SD (ns=not significant (p>0.05), *p<0.05, **p<0.01). The results were statistically evaluated utilising a one-way analysis of variance (ANOVA) with Bonferroni's multiple comparisons post LO test. Figure 26 shows effects of phase II administered BAK502G9. Figure 26A shows effect on AHR as measured by change in area under the histamine dose response curve (n=14). L5 Figure 26B shows effect on AHR as measured by change in
PC
3 o (n=18). Figure 26C shows effect on antigen priming (n=20). Figure 26D shows effect on BAL inflammation (n=21). ?0 Figure 27 shows effect of BAK502G9 on IL-13-induced CD23 expression. Data are presented as a percentage of the response to IL-13 alone (100%) and expressed as mean ± SEM % control of 6 separate experiments from 6 individual donors (performed in triplicate). 25 Figure 28 shows effect of BAK502G9 and irrelevant IgG4 on IL 13 and/or IL-4 induced PBMC CD23 expression. Data are presented as a percentage of the response to IL-4 alone (100%) and expressed as mean ± SEM % control of 4 separate 30 experiments from 4 individual donors (performed in triplicate). Figure 29A shows effect of BAK502G9 on NHLF eotaxin-1 production induced by 48h culture with IL-13/ TNF-a/ TGF-P1 19 containing media. Data are shown as an arithmetic mean ± SEM from triplicate determinations of the media used in this study to induce leukocyte shape change. 5 Figure 29B shows effect of BAK502G9 on shape change of human eosinophils induced by 1:16 diultion of conditioned media. Data points represented are mean ± SEM %blank media shape change from separate experiments from four individual donors. LO Figure 30 shows alignment of human IL-13 against murine IL13 highlighting the mutations that were introduced into human IL 13 to produce the first panel of IL-13 chimaeras. The four alpha helices are highlighted in boxes and loop 1 and loop 3 are labelled. Five chimeric proteins were produced where L5 helices B, C and D, and loops 1 and loop 3 were replaced with the murine sequence. Four further chimeric proteins were produced and numbered according to the amino acid in the human pre-protein (not to the numbering of the multiple aligment above) where arginine at residue 30 (position 34 above) was 0 mutated, residues 33 and 34 (position 37 and 38 above) were mutated, residues 37 and 38 (VH) were mutated (position 41 and 42 above), and residues 40 and 41 (TQ) were mutated (position 44 and 45 above). 25 Figure 31 shows alignment of human IL-13 against murine IL-13 highlighting the mutations that were introduced into human IL 13 to produce the second panel of IL-13 chimaeras. Six chimaeras were produced where the human residue(s) were substituted for the murine residue(s) (highlighted with 30 boxes). Four further chimeric proteins were produced (numbering is according to the amino acid position in the human pre-protein) where leucine at residue 58 (62 in above figure) was mutated, leucine at residue 119 (residue 123 above) was mutated, lysine at position 123 (residue 127 above) 20 was mutated, and arginine at residue 127 (residue 132 above was mutated. Figure 32 shows mutations made to human IL-13. Mutations in 5 dark grey reduced binding to BAK5O2G9, mutations in light grey did not alter binding. Linear sequence of pre-human IL-13 with the mutated residues indicated. In various aspects and embodiments of the invention there is LO provided the subject-matter of the claims included below. The present invention provides specific binding members for IL-13, in particular human and/or primate IL-13 and/or variant IL-13 (R130Q), and murine IL-13. Preferred embodiments within L5 the present invention are antibody molecules, whether whole antibody (e.g. IgG, such as IgG4) or antibody fragments (e.g. scFv, Fab, dAb). Antibody antigen binding regions are provided, as are antibody VH and VL domains. Within VH and VL domains are provided complementarity determining regions, ?O CDR's, which may be provided within different framework regions, FR's, to form VH or VL domains as the case may be. An antigen binding site may consist of an antibody VH domain and/or a VL domain. 25 An antigen binding site may be provided by means of arrangement of CDR's on non-antibody protein scaffolds such as fibronectin or cytochrome B etc. [115, 116]. Scaffolds for engineering novel binding sites in proteins have been reviewed in detail by Nygren et al [116]. Protein scaffolds for 30 antibody mimics are disclosed in WO/0034784 in which the inventors describe proteins (antibody mimics) which include a fibronectin type III domain having at least one randomised loop. A suitable scaffold into which to graft one or more 21 CDR's, e.g. a set of HCDR's, may be provided by any domain member of the immunoglobulin gene superfamily. Preferred embodiments of the present invention are in what is 5 termed herein the "BAK278D6 lineage". This is defined with reference to a set of six CDR sequences of BAK278D6 as follows: HCDR1 (SEQ ID NO: 1), HCDR2 (SEQ ID NO: 2), HCDR3 (SEQ ID NO: 3), LCDR1 (SEQ ID NO: 4), LCDR2 (SEQ ID NO: 5) and LCDR3 (SEQ ID NO: 6). In one aspect, the present invention LO provides a specific binding member for human IL-13, comprising an antibody antigen-binding site which is composed of a human antibody VH domain and a human antibody VL domain and which comprises a set of CDR's, wherein the VH domain comprises HCDR 1, HCDR2 and HCDR3 and the VL domain comprises LCDR1, LCDR2 [5 and LCDR3, wherein the HCDR1 has the amino acid sequence of SEQ ID NO: 1, the HCDR2 has the amino acid sequence of SEQ ID NO: 2, the HCDR3 has the amino acid sequence of SEQ ID NO: 3, the LCDR1 has the amino acid sequence of SEQ ID NO: 4, the LCDR2 has the amino acid sequence of SEQ ID NO: 5, and the !0 LCDR3 has the amino acid sequence of SEQ ID NO: 6; or wherein the set of CDR's contains one or two amino acid substitutions compared with the set of CDR's, wherein the HCDR1 has the amino acid sequence of SEQ ID NO: 1, the HCDR2 has the amino acid sequence of SEQ ID NO: 2, the HCDR3 has the amino acid 25 sequence of SEQ ID NO: 3, the LCDR1 has the amino acid sequence of SEQ ID NO: 4, the LCDR2 has the amino acid sequence of SEQ ID NO: 5, and the LCDR3 has the amino acid sequence of SEQ ID NO: 6. 30 The set of CDR's wherein the HCDR1 has the amino acid sequence of SEQ ID NO: 1, the HCDR2 has the amino acid sequence of SEQ ID NO: 2, the HCDR3 has the amino acid sequence of SEQ ID NO: 3, the LCDR1 has the amino acid sequence of SEQ ID NO: 4, the LCDR2 has the amino acid sequence of SEQ ID NO: 5, and the 22 LCDR3 has the amino acid sequence of SEQ ID NO: 6, are herein referred to as the "BAK278D6 set of CDR's". The HCDR1, HCDR2 and HCDR3 within the BAK278D6 set of CDR's are referred to as the "BAK278D6 set of HCDR's" and the LCDR1, LCDR2 and LCDR3 5 within the BAK278D6 set of CDR's are referred to as the "BAK278D6 set of LCDR's". A set of CDR's with the BAK278D6 set of CDR's, BAK278D6 set of HCDR's or BAK278D6 LCDR's, or one or two substitutions therein, is said to be of the BAK278D6 lineage. LO As noted, in one aspect the invention provides a specific binding member for human IL-13, comprising an antibody antigen-binding site which is composed of a human antibody VH domain and a human antibody VL domain and which comprises a L5 set of CDR's, wherein the set of CDR's is the BAK278D6 set of CDR's or a set of CDR's containing one or two substitutions compared with the BAK278D6 set of CDR's. In preferred embodiments, the one or two substitutions are at ?0 one or two of the following residues within the CDRs of the VH and/or VL domains, using the standard numbering of Kabat [107]. 31, 32, 34 in HCDR1 25 52, 52A, 53, 54, 56, 58, 60, 61, 62, 64, 65 in HCDR2 96, 97, 98, 99, 101 in HCDR3 30 26, 27, 28, 30, 31 in LCDR1 56 in LCDR2 95A, 97 in LCDR3 23 Preferred embodiments have two substitutions compared with the BAK278D6 set of CDR's, at HCDR3 residue 99 and LCDR1 residue 27. Of these embodiments, preferred embodiments have S 5 substituted for N at HCDR3 residue 99 and/or I substituted for N at LCDR 1 residue. 27. Still further embodiments have a substitution at HCDR3 residue 99 selected from the group consisting of S, A, I, R, P and K, and/or a substitution at LCDR1 residue 27 selected from the group consisting of I, L, 10 M, C, V, K, Y, F, R, T, S, A, H and G. In preferred embodiments one or two substitutions are made at one or two of the following residues within the BAK278D6 set of CDR's in accordance with the identified groups of possible L5 substitute residues: Position of Substitute Residue substitution selected from the group consisting of ?0 31 in HCDR1: Q, D, L, G and E 32 in HCDR1: T 34 in HCDR1: V, I and F 25 52 in HCDR2: D, N, A, R, G and E 52A in HCDR2: D, G, T, P, N and Y 53 in HCDR2: D, L, A, P, T, S, I and R 54 in HCDR2: S, T, D, G, K and I 30 56 in HCDR2: T, E, Q, L, Y, N, V, A, M and G 58 in HCDR2: I, L, Q, S, M, H, D and K 60 in HCDR2: R 61 in HCDR2: R 24 62 in HCDR2: K and G 64 in HCDR2: R 65 in HCDR2: K 5 96 in HCDR3: R and D 97 in HCDR3: N, D, T and P 98 in HCDR3: R 99 in HCDR3: S,A, I, R, P and K 101 in HCDR3: Y LO 26 in LCDR1: D and S 27 in LCDRl: I, L, M, C, V, K, Y, F, R, T, S, A, H and G 28 in LCDR1: V 30 in LCDR1: G L5 31 in LCDR1: R 56 in LCDR2: T 95A in LCDR3: N ?0 97 in LCDR3: I Preferred embodiments have the BAK278D6 set of CDR's with a substitution of S for N at residue 99 within HCDR3 and I for N at residue 27 within LCDR 1. The set of CDR's thus defined is 25 as follows: HCDR1 - SEQ ID NO: 7; HCDR2 - SEQ ID NO: 8, HCDR3 - SEQ ID NO: 9; LCDR1 - SEQ ID NO: 10, LCDR2 - SEQ ID NO: 11; LCDR3 - SEQ ID NO: 12. This set of CDR's is herein referred to as the "BAK502G9 set of CDR's". 30 Further preferred embodiments have the BAK278D6 set of CDR's with one or two substitutions within the CDR's, with the proviso that the pair of substitutions of S for N at residue 99 within HCDR3 and I for N at residue 27 within LCDR 1 is excluded. 25 Other preferred embodiments are as follows: BAK 1166G2: HCDR1- SEQ ID NO: 67, HCDR2- SEQ ID NO: 68, HCDR3- SEQ ID NO: 69, LCDR1 - SEQ ID NO: 70, LCDR2 - SEQ ID NO: 71; LCDR3 - SEQ 5 ID NO: 72. BAKll67F2 HCDR1- SEQ ID NO: 61, HCDR2- SEQ ID NO:62, HCDR3 SEQ ID NO:63, LCDR1 - SEQ ID NO: 64, LCDR2 - SEQ ID NO: 65; LCDR3 - SEQ ID NO: 66. .0 BAKll84C8: HCDR1- SEQ ID NO:73, HCDR2: SEQ ID NO:74, HCDR3 SEQ ID NO:75. LCDR1 - SEQ ID NO: 76, LCDR2 - SEQ ID NO: 77; LCDR3 - SEQ ID NO: 78. .5 BAK1185El: HCDR1- SEQ ID NO:79, HCDR2- SEQ ID NO:80, HCDR3 SEQ ID NO: 81. LCDR1 - SEQ ID NO: 82, LCDR2 - SEQ ID NO: 83; LCDR3 - SEQ ID NO: 84. BAK1167F4: HCDR1- SEQ ID NO: 85, HCDR2- SEQ ID NO:86, HCDR3 !0 SEQ ID NO:87. LCDR1 - SEQ ID NO: 88, LCDR2 - SEQ ID NO: 89; LCDR3 - SEQ ID NO: 90. BAKllllD10: HCDR1- SEQ ID NO: 91, HCDR2- SEQ ID NO: 92, HCDR3 SEQ ID NO: 93. LCDR1 - SEQ ID NO: 94, LCDR2 - SEQ ID NO: 95; 25 LCDR3 - SEQ ID NO: 96. BAK1183H4: HCDR1- SEQ ID NO: 97, HCDR2- SEQ ID NO: 98, HCDR3 SEQ ID NO: 99. LCDR1 - SEQ ID NO: 100, LCDR2 - SEQ ID NO: 101; LCDR3 - SEQ ID NO: 102. 30 BAK1l85F8: HCDR1- SEQ ID NO: 103, HCDR2- SEQ ID NO: 104, HCDR3- SEQ ID NO: 105. LCDR1 - SEQ ID NO: 106, LCDR2 - SEQ ID NO: 107; LCDR3 - SEQ ID NO: 108. All of these were derived 26 from BAK502G9 by heavy chain CDR1 and CDR2 randomisation and are thus of the BAK502G9 lineage. A VH domain comprising a set of CDR's HCDR1, HCDR2 and HCDR3 5 of any clone as shown in Table 1. Table 1 is also provided by the present invention, as is separately a VL domain comprising a set of CDR's LCDR1, LCDR2 and LCDR3 of the clones shown in Table 1. Preferably such a VH domain is paired with such a VL domain, and most preferably the VH and VL domain pairings are LO the same as in the clones as set out in Table 1. Further provided by the present invention is a VH domain comprising a set of CDR's HCDR1, HCDR2 and HCDR3 wherein the set of CDR's corresponds to that for any clone shown in Table .5 1 with one or two amino acid substitutions. Further provided by the present invention is a VL domain comprising a set of CDR's LCDR1, LCDR2 and LCDR3 wherein the set of CDR's corresponds to that for any clone shown in Table !0 1 with one or two amino acid substitutions. A specific binding member comprising an antibody antigen binding domain comprising such a VH and/or VL domain is also provided by the present invention. 25 The present inventors have identified the BAK278D6 lineage as providing human antibody antigen-binding domains against IL-13 which are of particular value. Within the lineage, BAK502G9 has been identified to be of special value. The BAK278D6 and 30 BAK502G9 sets of CDR's have been identified already above. Following the lead of computational chemistry in applying multivariate data analysis techniques to the structure/property-activity relationships [94], quantitative 27 activity-property relationships of antibodies can be derived using well-known mathematical techniques such as statistical regression, pattern recognition and classification [95-100]. The properties of antibodies can be derived from empirical and 5 theoretical models (for example, analysis of likely contact residues or calculated physicochemical property ) of antibody sequence, functional and three-dimensional structures and these properties can be considered singly and in combination. .0 An antibody antigen-binding site composed of a VH domain and a VL domain is formed by six loops of polypeptide: three from the light chain variable domain (VL) and three from the heavy chain variable domain (VH). Analysis of antibodies of known atomic structure has elucidated relationships between the .5 sequence and three-dimensional structure of antibody combining sites[101,102]. These relationships imply that, except for the third region (loop) in VH domains, binding site loops have one of a small number of main-chain conformations: canonical structures. The canonical structure formed in a particular .0 loop has been shown to be determined by its size and the presence of certain residues at key sites in both the loop and in framework regions [101,102]. This study of sequence-structure relationship can be used for 25 prediction of those residues in an antibody of known sequence, but of an unknown three-dimensional structure, which are important in maintaining the three-dimensional structure of its CDR loops and hence maintain binding specificity. These predictions can be backed up by comparison of the predictions 30 to the output from lead optimization experiments. In a structural approach, a model can be created of the antibody molecule [103] using an freely available or commercial package such as WAM [104]. A protein visualisation 28 and analysis software package such as Insight IT [105] or Deep View [106] may then be used to evaluate possible substitutions at each position in the CDR. This information may then be used to make substitutions likely to have a minimal or beneficial 5 effect on activity. The present inventors analysed sequence data of the panel of clones for which the sets of CDR's are shown in Table 1. LO The analysis tested the hypothesis that any binary combinations of listed amino acid variations in the CDR's from the presented set of scFv variants leads to a scFv variant with at least the starting potency of the parent scFv BAK278D6. L5 All scFv variants in the panel shown in Table 1 have been selected for improved affinity and have been confirmed to display higher potency. !0 The observed amino acid variations can either be favourable, non-favourable or neutral in their effect on the starting potency of scFv BAK278D6 in the TF-1 assay of 44nM. No linkage was observed between any two amino acid variations 25 confirming that there was no synergy, either "positive" or "negative", between any two selected amino acid variations. There are four scenarios where such binary combination will fulfil the hypothesis and three scenarios where the hypothesis 30 will not be valid. Synergistic amino acid variants are not considered as no linkage was observed. The hypothesis is valid where: 29 Al: mutation 1 is favourable and mutation 2 is favourable A2 : mutation 1 is favourable and mutation 2 is neutral A3: mutation 1 is neutral and mutation 2 is neutral A4: mutation 1 is favourable and mutation 2 is non-favourable 5 (with the effect of 1 outweighing the effect of 2) The hypothesis is not valid where: Bl: mutation 1 is non-favourable and mutation 2 is neutral 10 B2: mutation 1 is non-favourable and mutation 2 is non favourable B3: mutation 1 is favourable and mutation 2 is non-favourable (with the effect of 2 outweighing the effect of 1). L5 For A4 to be possible, mutation 1 needs to be highly favourable to counterbalance the negative effect of mutation 2 on potency. Since such highly favourable mutation would be present in the library of variants used for selection, it would be selected for and would therefore appear frequently in ?0 the panel of variants. Since synergy can be excluded, such mutation would be beneficial in any kind of sequence context and should therefore reappear in different scFv variants. An example for such frequent amino acid change is the change in the light chain CDR1 Asn27Ile. However, this mutation on its 25 own (in clone BAK531E2) has only a modest 2-fold effect on potency (final IC50 of 23.2nM). On its own this mutation would not allow the scenario depicted in A4, as it is not a highly favourable mutation. This suggests that every clone in the presented set of IL-13 binding clones (Table 1) which has a 30 light chain CDR1 Asn27Ile change along with one or more further mutations is at least as potent as the variant having the single light chain CDR1 Asn27Ile mutation. The other mutations are either neutral or positive but do not have a negative or detrimental affect. 30 A further example is in the heavy chain CDR3 Asn99Ser (see Table 1). As a clone carrying this particular single amino acid variation is not observed, the potency of such a clone 5 has been estimated to be approximately 12.OnM by the following rationale: BAK278D6 potency is 44nM. Alterations of VL CDR1 N271 + VH CDR3 N99S lead to BAK502G9 with potency 8nM, i.e. 5.5 fold .0 improvement. BAK278D6 potency is 44nM. Alteration of VL CDR1 N271 leads to BAK531E2 with potency 23 nM, i.e. 1.9 fold improvement .5 BAK278D6 potency is 44nM. Alteration VH CDR3 N99S to provide a possible clone with potency 12.2nM, i.e. 2.9 fold improvement (5.5/1.9 = 2.9). The binary combination of heavy chain CDR3 Asn99Ser with light .0 chain CDR1 Asn27Ile gives a scFv BAK0502G9 with a potency of 8nM. As synergy is excluded, the contribution of heavy chain CDR3 Asn99Ser change in BAK502G9 is therefore additive. Therefore every clone in the presented set of IL-13 binding 25 clones (Table 1) which has a heavy chain CDR3 AsnH99Ser change along with one or more further mutations would have a potency of at least 12nM or greater, within a permissive assay window of 2.5-fold for n=1-2. 30 Thus, the inventors note that a highly favourable amino acid variation which would be selected preferentially is not observed. As discussed above, two variations which were prominently represented in Table 1 of scFv variants were analysed closer. Any scFv variant in Table 1 with either of 31 these mutations along with one or more further mutations displayed a potency which was at least as improved as a clone containing any one of these two single amino acid variations in the parent BAK278D6. There is therefore no evidence that a 5 highly favourable amino acid variation, that would allow scenario A4, is present in the panel. This observation led the inventors to conclude that there were no non-favourable mutations present in this set of scFv .0 variants. This means scenarios A4 and B1 to B3 are not relevant and the hypothesis is valid. Accordingly, as noted already, the present invention provides specific binding members comprising the defined sets of CDR's, .5 in particular the set of CDR's of BAK278D6, and sets of CDR's of the BAK278D6 lineage, with one or two substitutions within the set of CDR's, e.g. the BAK502G9 set of CDR's. The relevant set of CDR's is provided within antibody :0 framework regions or other protein scaffold, e.g. fibronectin or cytochrome B [115, 1161. Preferably antibody framework regions are employed, and where they are employed they are preferably germline, more preferably the antibody framework region for the heavy chain may be DPl4 from the VHl family. 25 The preferred framework region for the light chain may be X3 3H. For the BAK502G9 set of CDR's it is preferred that the antibody framework regions are for VH FR1, SEQ ID NO: 27, for VH FR2, SEQ ID NO: 28, for VH FR3, SEQ ID NO 29, for light chain FR1, SEQ ID NO: 30, for light chain FR2, SEQ ID NO: 31, 30 for light chain FR3, SEQ ID NO: 32. In a highly preferred embodiment, a VH domain is provided with the amino acid sequence of SEQ ID NO: 15, this being termed "BAK502G9 VH domain". In a further highly preferred embodiment, a VL domain is provided with the amino acid sequence of SEQ ID NO: 32 16, this being termed "BAK502G9 VL domain". A highly preferred antibody antigen-binding site provided in accordance with the present invention is composed of the BAK502G9 VH domain, SEQ ID NO: 15, and the BAK502G9 VL domain, SEQ ID NO: 5 16. This antibody antigen-binding site may be provided within any desired antibody molecule format, e.g. scFv, Fab, IgG, IgG4, dAb etc., as is discussed further elsewhere herein. In a further highly preferred embodiment, the present 10 invention provides an IgG4 antibody molecule comprising the BAK502G9 VH domain, SEQ ID NO: 15, and the BAK502G9 VL domain, SEQ ID NO: 16. This is termed herein "BAK502G9 IgG4". Other IgG4 or other antibody molecules comprising the BAK502G9 15 VH domain, SEQ ID NO: 15, and/or the BAK502G9 VL domain, SEQ ID NO: 16, are provided by the present invention, as are other antibody molecules comprising the BAK502G9 set of HCDR's (SEQ ID NO: 7, 8 and 9) within an antibody VH domain, and/or the BAK502G9 set of LCDR's (SEQ ID NO: 10, 11 and 12) within an 20 antibody VL domain. It is convenient to point out here that "and/or" where used herein is to be taken as specific disclosure of each of the two specified features or components with or without the 25 other. For example "A and/or B" is to be taken as specific disclosure of each of (i) A, (ii) B and (iii) A and B, just as if each is set out individually herein. As noted, the present invention provides a specific binding 30 member which binds human IL-13 and which comprises the BAK502G9 VH domain (SEQ ID NO: 15) and/or the BAK502G9 VL domain (SEQ ID NO: 16). 33 Generally, a VH domain is paired with a VL domain to provide an antibody antigen binding site, although as discussed further below a VH domain alone may be used to bind antigen. In one preferred embodiment, the BAK502G9 VH domain (SEQ ID 5 NO: 15) is paired with the BAK502G9 VL domain (SEQ ID NO: 16), so that an antibody antigen binding site is formed comprising both the BAK502G9 VH and VL domains. In other embodiments, the BAK502G9 VH is paired with a VL domain other than the BAK502G9 VL. Light-chain promiscuity is well established in .0 the art. Similarly, any set of HCDR's of the BAK278D6 lineage can be provided in a VH domain that is used as a specific binding member alone or in combination with a VL domain. A VH domain .5 may be provided with a set of HCDR's of a BAK278D6 lineage antibody, e.g. as shown in Table 1, and if such a VH domain is paired with a VL domain, then the VL domain may be provided with a set of LCDR's of a BAK278D6 lineage antibody, e.g. as shown in Table 1. A pairing of a set of HCDR's and a set of !0 LCDR's may be as shown in Table 1, providing an antibody antigen-binding site comprising a set of CDR's as shown in Table 1. The framework regions of the VH and/or VL domains may be germline frameworks. Frameworks regions of the heavy chain domain may be selected from the VH-l family, and a 25 preferred VH-l framework is DP-14 framework. Framework regions of the light chain may be selected from the k3 family, and a preferred such framework is k3 3H. One or more CDRs may be taken from the BAK502G9 VH or VL 30 domain and incorporated into a suitable framework. This is discussed further herein. BAK502G9 HCDR's 1, 2 and 3 are shown in SEQ ID NO: 7, 8 and 9, respectively. BAK502G9 LCDR's 1, 2 and 3 are shown in SEQ ID NO: 10, 11 and 12, respectively. 34 The same applies for other BAK278D6 lineage CDR's and sets of CDR's as shown in Table 1. 5 Further embodiments of the invention relate to a specific binding member comprising the VH and/or VL domain, or an antigen binding site comprising CDRs of the VH and/or VL domain of the antibody molecule disclosed herein as 167A11 (VH: SEQ ID NO: 23 and VL: SEQ ID NO: 24) and its 10 derivatives 615E3 (VH:SEQ ID NO: 33 and VL: SEQ ID NO: 34) BAK582F7 (VH CDR's SEQ ID's 141-143) and BAK612B5 (VH CDR's SEQ ID's 147-149). These recognise human IL-13. The derivatives of 167A11 from VH CDR3 randomisation are potent scFv molecules (5-6nM). The 167All lineage may be employed in 15 any aspect and embodiment of the present invention as disclosed herein for other molecules, for instance methods of mutation and selection of antigen binding sites with improved potency. 20 Variants of the VH and VL domains and CDRs of the present invention, including those for which amino acid sequences are set out herein, and which can be employed in specific binding members for IL-13 can be obtained by means of methods of sequence alteration or mutation and screening. Such methods 25 are also provided by the present invention. Variable domain amino acid sequence variants of any of the VH and VL domains whose sequences are specifically disclosed herein may be employed in accordance with the present 30 invention, as discussed. Particular variants may include one or more amino acid sequence alterations (addition, deletion, substitution and/or insertion of an amino acid residue), may be less than about 20 alterations, less than about 15 alterations, less than about 10 alterations or less than about 35 5 alterations, 4, 3, 2 or 1. Alterations may be made in one or more framework regions and/or one or more CDR's. In accordance with further aspects of the present invention 5 there is provided a specific binding member which competes for binding to antigen with any specific binding member which both binds the antigen and comprises a specific binding member, VH and/or VL domain disclosed herein, or HCDR3 disclosed herein, or variant of any of these. Competition between binding LO members may be assayed easily in vitro, for example using ELISA and/or by tagging a specific reporter molecule to one binding member which can be detected in the presence of other untagged binding member(s), to enable identification of specific binding members which bind the same epitope or an L5 overlapping epitope. Thus, a further aspect of the present invention provides a specific binding member comprising a human antibody antigen binding site which competes with a BAK502G9 antibody molecule, !0 in particular BAK502G9 scFv and/or IgG4, for binding to IL-13. In further aspects the present invention provides a specific binding member comprising a human antibody antigen-binding site which competes with an antibody antigen-binding site for binding to IL-13, wherein the antibody antigen-binding site is 25 composed of a VH domain and a VL domain, and wherein the VH and VL domains comprise a set of CDR's of the BAK278D6 lineage. Various methods are available in the art for obtaining 30 antibodies against IL-13 and which may compete with a BAK502G9 antibody molecule, an antibody molecule with a BAK502G9 set of CDR's, or an antibody molecule with a set of CDR's of BAK278D6 lineage, for binding to IL-13. 36 In a further aspect, the present invention provides a method of obtaining one or more specific binding members able to bind the antigen, the method including bringing into contact a library of specific binding members according to the invention 5 and said antigen, and selecting one or more specific binding members of the library able to bind said antigen. The library may be displayed on the surface of bacteriophage particles, each particle containing nucleic acid encoding the 10 antibody VH variable domain displayed on its surface, and optionally also a displayed VL domain if present. Following selection of specific binding members able to bind the antigen and displayed on bacteriophage particles, nucleic L5 acid may be taken from a bacteriophage particle displaying a said selected specific binding member. Such nucleic acid may be used in subsequent production of a specific binding member or an antibody VH variable domain (optionally an antibody VL variable domain) by expression from nucleic acid with the ?0 sequence of nucleic acid taken from a bacteriophage particle displaying a said selected specific binding member. An antibody VH variable domain with the amino acid sequence of an antibody VH variable domain of a said selected specific 25 binding member may be provided in isolated form, as may a specific binding member comprising such a VH domain. Ability to bind IL-13 may be further tested, also ability to compete with BAK502G9 (e.g. in scFv format and/or IgG format, e.g. IgG4) for binding to IL-13. Ability to neutralise IL-13 30 may be tested, as discussed further below. A specific binding member according to the present invention may bind IL-13 with the affinity of a BAK502G9 antibody 37 molecule, e.g. scFv, or preferably BAK502G9 IgG4, or with an affinity that is better. A specific binding member according to the present invention 5 may neutralise IL-13 with the potency of a BAK502G9 antibody molecule, e.g. scFv, or preferably BAK502G9 IgG4, or with a potency that is better. A specific binding member according to the present invention LO may neutralise naturally occurring IL-13 with the potency of a BAK502G9 antibody molecule, e.g. scFv, or preferably BAK502G9 IgG4, or with a potency that is better. Binding affinity and neutralisation potency of different .5 specific binding members can be compared under appropriate conditions. The antibodies of the present invention have a number of advantages over existing commercial anti-IL-13 antibodies, in !0 particular three commercial rodent anti-human IL-13 antibodies namely, JES10-5A2 (BioSource), B-B13 (Euroclone) and clone 321166 (R&D Systems). The potency of the antibodies of the present invention was compared with commercial antibodies JES10-A2 and B-B13. Clone 321166 was not evaluated as previous 25 experiments revealed that this clone was considerably less potent than other known commercial antibodies. The efficacy and use of the rodent commercial IL-13 antibodies in man is likely to be limited, because of their increased 30 potential to induce immunogenic responses and therefore more rapid clearance from the body. Kinetic analysis of the antibodies of the present invention in non-human primates suggests that these antibodies have a clearance rate which is similar to that of other known human or humanised antibodies. 38 Antibodies provided by various embodiments of the present invention recognize non-human primate IL-13, including rhesus and cynomolgus IL-13. Determining efficacy and safety profiles 5 of an antibody in non-human primates is extremely valuable as it provides a means for predicting the antibody's safety, pharmacokinetic and pharmacodynamic profile in humans. Moreover, antibodies of various embodiments of the present LO invention further recognize the human IL-13 variant, R130Q, which is associated with asthma. Cross reactivity with variant IL-13 allows antibodies of the present invention and compositions comprising antibodies of the present invention to be used for the treatment of patients with wild-type and L5 variant IL-13. A preferred embodiment of the present invention comprises antibodies that neutralise naturally occurring IL-13 with a potency that is equal to or better than the potency of a IL-13 20 antigen binding site formed by BAK502G9 VH domain (SEQ ID NO:15) and the BAK502G9 VL domain (SEQ ID NO: 16). For example, the inventors have demonstrated that representative clones such as BAK502G9, 1167F2 and 1183H4 are significantly more potent against naturally occurring IL-13 than known 25 commercial antibodies (Figure 7). In addition to antibody sequences, a specific binding member according to the present invention may comprise other amino acids, e.g. forming a peptide or polypeptide, such as a folded 30 domain, or to impart to the molecule another functional characteristic in addition to ability to bind antigen. Specific binding members of the invention may carry a detectable label, or may be conjugated to a toxin or a 39 targeting moiety or enzyme (e.g. via a peptidyl bond or linker). In further aspects, the invention provides an isolated nucleic 5 acid which comprises a sequence encoding a specific binding member, VH domain and/or VL domains according to the present invention, and methods of preparing a specific binding member, a VH domain and/or a VL domain of the invention, which comprise expressing said nucleic acid under conditions to .0 bring about production of said specific binding member, VH domain and/or VL domain, and recovering it. Specific binding members according to the invention may be used in a method of treatment or diagnosis of the human or .5 animal body, such as a method of treatment (which may include prophylactic treatment) of a disease or disorder in a human patient which comprises administering to said patient an effective amount of a specific binding member of the invention. Conditions treatable in accordance with the !0 present invention include any in which IL-13 plays a role, especially asthma, atopic dermatitis, allergic rhinitis, fibrosis, chronic obstructive pulmonary disease, scleroderma, inflammatory bowel disease and Hodgkin's lymphoma. Further, the antibodies of the present invention may also be used in 25 treating tumours and viral infections as these antibodies will inhibit IL-13 mediated immunosupression [64, 65]. A further aspect of the present invention provides nucleic acid, generally isolated, encoding an antibody VH variable 30 domain and/or VL variable domain disclosed herein. Another aspect of the present invention provides nucleic acid, generally isolated, encoding a VH CDR or VL CDR sequence disclosed herein, especially a VH CDR selected from SEQ ID 40 NO's: 7, 8 and 9 or a VL CDR selected from SEQ ID NO's: 10, 11 and 12, most preferably BAK502G9 VH CDR3 (SEQ ID NO: 9). Nucleic acid encoding the BAK502G9 set of CDR's, nucleic acid encoding the BAK502G9 set of HCDR's and nucleic acid encoding 5 the BAK502G9 set of LCDR's are also provided by the present invention, as are nucleic acids encoding individual CDR's, HCDR's, LCDR's and sets of CDR's, HCDR's, LCDR's of the BAK278D6 lineage. 10 A further aspect provides a host cell transformed with nucleic acid of the invention. A yet further aspect provides a method of production of an antibody VH variable domain, the method including causing L5 expression from encoding nucleic acid. Such a method may comprise culturing host cells under conditions for production of said antibody VH variable domain. Analogous methods for production of VL variable domains and ?0 specific binding members comprising a VH and/or VL domain are provided as further aspects of the present invention. A method of production may comprise a step of isolation and/or purification of the product. 25 A method of production may comprise formulating the product into a composition including at least one additional component, such as a pharmaceutically acceptable excipient. 30 These and other aspects of the invention are described in further detail below. 41 TERMINOLOGY Specific binding member This describes a member of a pair of molecules which have 5 binding specificity for one another. The members of a specific binding pair may be naturally derived or wholly or partially synthetically produced. One member of the pair of molecules has an area on its surface, or a cavity, which specifically binds to and is therefore complementary to a 10 particular spatial and polar organisation of the other member of the pair of molecules. Thus the members of the pair have the property of binding specifically to each other. Examples of types of specific binding pairs are antigen-antibody, biotin-avidin, hormone-hormone receptor, receptor-ligand, L5 enzyme-substrate. The present invention is concerned with antigen-antibody type reactions. Antibody molecule This describes an immunoglobulin whether natural or partly or )0 wholly synthetically produced. The term also covers any polypeptide or protein comprising an antibody binding domain. Antibody fragments which comprise an antigen binding domain are molecules such as Fab, scFv, Fv, dAb, Fd; and diabodies. 25 It is possible to take monoclonal and other antibodies and use techniques of recombinant DNA technology to produce other antibodies or chimeric molecules which retain the specificity of the original antibody. Such techniques may involve introducing DNA encoding the immunoglobulin variable region, 30 or the complementarity determining regions (CDRs), of an antibody to the constant regions, or constant regions plus framework regions, of a different immunoglobulin. See, for instance, EP-A-184187, GB 2188638A or EP-A-239400, and a large body of subsequent literature. A hybridoma or other cell 42 producing an antibody may be subject to genetic mutation or other changes, which may or may not alter the binding specificity of antibodies produced. 5 As antibodies can be modified in a number of ways, the term "antibody molecule" should be construed as covering any specific binding member or substance having an antibody antigen-binding domain with the required specificity. Thus, this term covers antibody fragments and derivatives, including LO any polypeptide comprising an immunoglobulin binding domain, whether natural or wholly or partially synthetic. Chimeric molecules comprising an immunoglobulin binding domain, or equivalent, fused to another polypeptide are therefore included. Cloning and expression of chimeric antibodies are .5 described in EP-A-0120694 and EP-A-0125023, and a large body of subsequent literature. Further techniques available in the art of antibody engineering have made it possible to isolate human and !0 humanised antibodies. For example, human hybridomas can be made as described by Kontermann et al [107]. Phage display, another established technique for generating specific binding members has been described in detail in many publications such as Kontermann et al [107] and W092/01047 (discussed further 25 below). Transgenic mice in which the mouse antibody genes are inactivated and functionally replaced with human antibody genes while leaving intact other components of the mouse immune system, can be used for isolating human antibodies to human antigens [108]. 30 - Synthetic antibody molecules may be created by expression from genes generated by means of oligonucleotides synthesized and assembled within suitable expression vectors, for example as 43 described by Knappik et al. J. Mol. Biol. (2000) 296, 57-86 or Krebs et al. Journal of Immunological Methods 254 2001 67-84. It has been shown that fragments of a whole antibody can 5 perform the function of binding antigens. Examples of binding fragments are (i) the Fab fragment consisting of VL, VH, CL and CHl domains; (ii) the Ed fragment consisting of the VH and CH1 domains; (iii) the Fv fragment consisting of the VL and VH domains of a single antibody; (iv) the dAb fragment (Ward, 0 E.S. et al., Nature 341, 544-546 (1989), McCafferty et al (1990) Nature, 348, 552-554) which consists of a VH domain; (v) isolated CDR regions; (vi) F(ab')2 fragments, a bivalent fragment comprising two linked Fab fragments (vii) single chain Fv molecules (scFv), wherein a VH domain and a VL domain 5 are linked by a peptide linker which allows the two domains to associate to form an antigen binding site (Bird et al, Science, 242, 423-426, 1988; Huston et al, PNAS USA, 85, 5879 5883, 1988); (viii) bispecific single chain Fv dimers (PCT/US92/09965) and (ix) "diabodies", multivalent or 0 multispecific fragments constructed by gene fusion (W094/13804; P. Holliger et al, Proc. Natl. Acad. Sci. USA 90 6444-6448, 1993). Fv, scFv or diabody molecules may be stabilised by the incorporation of disulphide bridges linking the VH and VL domains (Y. Reiter et al, Nature Biotech, 14, 25 1239-1245, 1996). Minibodies comprising a scFv joined to a CH3 domain may also be made (S. Hu et al, Cancer Res., 56, 3055-3061, 1996). Where bispecific antibodies are to be used, these may be 30 conventional bispecific antibodies, which can be manufactured in a variety of ways (Holliger, P. and Winter G. Current Opinion Biotechnol. 4, 446-449 (1993)), e.g. prepared chemically or from hybrid hybridomas, or may be any of the bispecific antibody fragments mentioned above. Examples of 44 bispecific antibodies include those of the BiTE technology in which the binding domains of two antibodies with different specificity can be used and directly linked via short flexible peptides. This combines two antibodies on a short single 5 polypeptide chain. Diabodies and scFv can be constructed without an Fc region, using only variable domains, potentially reducing the effects of anti-idiotypic reaction. Bispecific diabodies, as opposed to bispecific whole .0 antibodies, may also be particularly useful because they can be readily constructed and expressed in E.coli. Diabodies (and many other polypeptides such as antibody fragments) of appropriate binding specificities can be readily selected using phage display (W094/13804) from libraries. If one arm .5 of the diabody is to be kept constant, for instance, with a specificity directed against IL-13, then a library can be made where the other arm is varied and an antibody of appropriate specificity selected. Bispecific whole antibodies may be made by knobs-into-holes engineering (J. B. B. Ridgeway et al, .0 Protein Eng., 9, 616-621, 1996). Antigen-binding domain This describes the part of an antibody molecule which comprises the area which specifically binds to and is 25 complementary to part or all of an antigen. Where an antigen is large, an antibody may only bind to a particular part of the antigen, which part is termed an epitope. An antigen binding domain may be provided by one or more antibody variable domains (e.g. a so-called Fd antibody fragment 30 consisting of a VH domain). Preferably, an antigen binding domain comprises an antibody light chain variable region (VL) and an antibody heavy chain variable region (VH). 45 Specific This may be used to refer to the situation in which one member of a specific binding pair will not show any significant binding to molecules other than its specific binding 5 partner(s). The term is also applicable where e.g. an antigen binding domain is specific for a particular epitope which is carried by a number of antigens, in which case the specific binding member carrying the antigen binding domain will be able to bind to the various antigens carrying the epitope. -0 Comprise This is generally used in the sense of include, that is to say permitting the presence of one or more features or components. .5 Isolated This refers to the state in which specific binding members of the invention, or nucleic acid encoding such binding members, will generally be in accordance with the present invention. Isolated members and isolated nucleic acid will be free or .0 substantially free of material with which they are naturally associated such as other polypeptides or nucleic acids with which they are found in their natural environment, or the environment in which they are prepared (e.g. cell culture) when such preparation is by recombinant DNA technology 25 practised in vitro or in vivo. Members and nucleic acid may be formulated with diluents or adjuvants and still for practical purposes be isolated - for example the members will normally be mixed with gelatin or other carriers if used to coat microtitre plates for use in immunoassays, or will be 30 mixed with pharmaceutically acceptable carriers or diluents when used in diagnosis or therapy. Specific binding members may be glycosylated, either naturally or by systems of heterologous eukaryotic cells (e.g. CHO or NSO (ECACC 46 85110503) cells, or they may be (for example if produced by expression in a prokaryotic cell) unglycosylated. Naturally occurring IL-13 5 This generally refers to a state in which the IL-13 protein or fragments thereof may occur. Naturally occurring IL-13 means IL-13 protein which is naturally produced by a cell, without prior introduction of encoding nucleic acid using recombinant technology. Thus, naturally occurring IL-13 may be as 10 produced naturally by for example CD4+ T cells and/or as isolated from a mammal, e.g. human, non-human primate, rodent such as rat or mouse. Recombinant IL-13 L5 This refers to a state in which the IL-13 protein or fragments thereof may occur. Recombinant IL-13 means IL-13 protein or fragments thereof produced by recombinant DNA in a heterologous host. Recombinant IL-13 may differ from naturally occurring IL-13 by glycosylation. ?0 Recombinant proteins expressed in prokaryotic bacterial expression systems are not glycosylated while those expressed in eukaryotic systems such as mammalian or insect cells are glycosylated. Proteins expressed in insect cells however 25 differ in glycosylation from proteins expressed in mammalian cells. By "substantially as set out" it is meant that the relevant CDR or VH or VL domain of the invention will be either 30 identical or highly similar to the specified regions of which the sequence is set out herein. By "highly similar" it is contemplated that from 1 to 5, preferably from 1 to 4 such as 1 to 3 or 1 or 2, or 3 or 4, amino acid substitutions may be made in the CDR and/or VH or VL domain. 47 The structure for carrying a CDR or a set of CDR's of the invention will generally be of an antibody heavy or light chain sequence or substantial portion thereof in which the CDR 5 or set of CDR's is located at a location corresponding to the CDR or set of CDR's of naturally occurring VH and VL antibody variable domains encoded by rearranged immunoglobulin genes. The structures and locations of immunoglobulin variable domains may be determined by reference to (Kabat, E.A. et al, LO Sequences of Proteins of Immunological Interest. 4th Edition. US Department of Health and Human Services. 1987, and updates thereof, now available on the Internet (http://immuno.bme.nwu.edu or find "Kabat" using any search engine). -5 CDR's can also be carried by other scaffolds such as fibronectin or cytochrome B [115, 116]. Preferably, a CDR amino acid sequence substantially as set out .0 herein is carried as a CDR in a human variable domain or a substantial portion thereof. The HCDR3 sequences substantially as set out herein represent preferred embodiments of the present invention and it is preferred that each of these is carried as a HCDR3 in a human heavy chain 25 variable domain or a substantial portion thereof. Variable domains employed in the invention may be obtained from any germ-line or rearranged human variable domain, or may be a synthetic variable domain based on consensus sequences of 30 known human variable domains. A CDR sequence of the invention (e.g. CDR3) may be introduced into a repertoire of variable domains lacking a CDR (e.g. CDR3), using recombinant DNA technology. 48 For example, Marks et al (Bio/Technology, 1992, 10:779-783) describe methods of producing repertoires of antibody variable domains in which consensus primers directed at or adjacent to the 5' end of the variable domain area are used in conjunction 5 with consensus primers to the third framework region of human VH genes to provide a repertoire of VH variable domains lacking a CDR3. Marks et al further describe how this repertoire may be combined with a CDR3 of a particular antibody. Using analogous techniques, the CDR3-derived .0 sequences of the present invention may be shuffled with repertoires of VH or VL domains lacking a CDR3, and the shuffled complete VH or VL domains combined with a cognate VL or VH domain to provide specific binding members of the invention. The repertoire may then be displayed in a suitable .5 host system such as the phage display system of W092/01047 or any of a subsequent large body of literature, including Kay, B.K., Winter, J., and McCafferty, J. (1996) Phage Display of Peptides and Proteins: A Laboratory Manual, San Diego: Academic Press, so that suitable specific binding members may .0 be selected. A repertoire may consist of from anything from 104 individual members upwards, for example from 106 to 108 or 1010 members. Other suitable host systems include yeast display, bacterial display, T7 display, ribosome display and so on. For a review of ribosome display for see Lowe D and 25 Jermutus L, 2004, Curr. Pharm, Biotech, 517-27, also W092/01047. Analogous shuffling or combinatorial techniques are also disclosed by Stemmer (Nature, 1994, 370:389-391), who 30 describes the technique in relation to a P-lactamase gene but observes that the approach may be used for the generation of antibodies. 49 A further alternative is to generate novel VH or VL regions carrying CDR-derived sequences of the invention using random mutagenesis of one or more selected VH and/or VL genes to generate mutations within the entire variable domain. Such a 5 technique is described by Gram et al (1992, Proc. Natl. Acad. Sci., USA, 89:3576-3580), who used error-prone PCR. In preferred embodiments one or two amino acid substitutions are made within a set of HCDR's and/or LCDR's. LO Another method which may be used is to direct mutagenesis to CDR regions of VH or VL genes. Such techniques are disclosed by Barbas et al, (1994, Proc. Natl. Acad. Sci., USA, 91:3809 3813) and Schier et al (1996, J. Mol. Biol. 263:551-567). L5 All the above described techniques are known as such in the art and in themselves do not form part of the present invention. The skilled person will be able to use such techniques to provide specific binding members of the invention using routine methodology in the art. 20 A further aspect of the invention provides a method for obtaining an antibody antigen binding domain specific for IL 13 antigen, the method comprising providing by way of addition, deletion, substitution or insertion of one or more 25 amino acids in the amino acid sequence of a VH domain set out herein a VH domain which is an amino acid sequence variant of the VH domain, optionally combining the VH domain thus provided with one or more VL domains, and testing the VH domain or VH/VL combination or combinations to identify a 30 specific binding member or an antibody antigen binding domain specific for IL-13 antigen and optionally with one or more preferred properties, preferably ability to neutralize IL-13 activity. Said VL domain may have an amino acid sequence which is substantially as set out herein. 50 An analogous method may be employed in which one or more sequence variants of a VL domain disclosed herein are combined with one or more VH domains. 5 In a preferred embodiment, BAK502G9 VH domain (SEQ ID NO: 15 may be subject to mutation to provide one or more VH domain amino acid sequence variants, and/or BAK502G9 VL (SEQ ID NO: 16). .0 A further aspect of the invention provides a method of preparing a specific binding member specific for IL-13 antigen, which method comprises: (a) providing a starting repertoire of nucleic acids .5 encoding a VH domain which either include a CDR3 to be replaced or lack a CDR3 encoding region; (b) combining said repertoire with a donor nucleic acid encoding an amino acid sequence substantially as set out herein for a VH CDR3 such that said donor nucleic acid is .0 inserted into the CDR3 region in the repertoire, so as to provide a product repertoire of nucleic acids encoding a VH domain; (c) expressing the nucleic acids of said product repertoire; 25 (d) selecting a specific binding member specific for a IL-13; and (e) recovering said specific binding member or nucleic acid encoding it. 30 Again, an analogous method may be employed in which a VL CDR3 of the invention is combined with a repertoire of nucleic acids encoding a VL domain which either include a CDR3 to be replaced or lack a CDR3 encoding region. 51 Similarly, one or more, or all three CDRs may be grafted into a repertoire of VH or VL domains which are then screened for a specific binding member or specific binding members specific for IL-13. 5 In a preferred embodiment, one or more of BAK502G9 HCDR1 (SEQ ID NO: 7), HCDR2 (SEQ ID NO: 8) and HCDR3 (SEQ ID NO: 9), or the BAK502G9 set of HCDR's, may be employed, and/or one or more of BAK502G9 LCDR1 (SEQ ID NO: 10), LCDR2 (SEQ ID NO: 11), .0 or the BAK502G9 set of LCDR's. A substantial portion of an immunoglobulin variable domain will comprise at least the three CDR regions, together with their intervening framework regions. Preferably, the portion .5 will also include at least about 50% of either or both of the first and fourth framework regions, the 50% being the C terminal 50% of the first framework region and the N-terminal 50% of the fourth framework region. Additional residues at the N-terminal or C-terminal end of the substantial part of '0 the variable domain may be those not normally associated with naturally occurring variable domain regions. For example, construction of specific binding members of the present invention made by recombinant DNA techniques may result in the introduction of N- or C-terminal residues encoded by linkers 25 introduced to facilitate cloning or other manipulation steps. Other manipulation steps include the introduction of linkers to join variable domains of the invention to further protein sequences including immunoglobulin heavy chains, other variable domains (for example in the production of diabodies) 30 or protein labels as discussed in more detail elsewhere herein. Although in a preferred aspect of the invention specific binding members comprising a pair of VH and VL domains are 52 preferred, single binding domains based on either VH or VL domain sequences form further aspects of the invention. It is known that single immunoglobulin domains, especially VH domains, are capable of binding target antigens in a specific 5 manner. In the case of either of the single specific binding domains, these domains may be used to screen for complementary domains capable of forming a two-domain specific binding member able L0 to bind IL-13. This may be achieved by phage display screening methods using the so-called hierarchical dual combinatorial approach as disclosed in W092/01047, in which an individual colony .5 containing either an H or L chain clone is used to infect a complete library of clones encoding the other chain (L or H) and the resulting two-chain specific binding member is selected in accordance with phage display techniques such as those described in that reference. This technique is also !0 disclosed in Marks et al, ibid. Specific binding members of the present invention may further comprise antibody constant regions or parts thereof. For example, a VL domain may be attached at its C-terminal end to 25 antibody light chain constant domains including human CK or Ck chains, preferably C4 chains. Similarly, a specific binding member based on a VH domain may be attached at its C-terminal end to all or part (e.g. a CHl domain) of an immunoglobulin heavy chain derived from any antibody isotype, e.g. IgG, IgA, 30 IgE and IgM and any of the isotype sub-classes, particularly IgGl and IgG4. IgG4 is preferred. IgG4 is preferred because it does not bind complement and does not create effector functions. Any synthetic or other constant region variant that has these properties and stabilizes variable regions is 53 also preferred for use in embodiments of the present invention. Specific binding members of the invention may be labelled with 5 a detectable or functional label. Detectable labels include radiolabels such as ' 31 I or 99 Tc, which may be attached to antibodies of the invention using conventional chemistry known in the art of antibody imaging. Labels also include enzyme labels such as horseradish peroxidase. Labels further include [0 chemical moieties such as biotin which may be detected via binding to a specific cognate detectable moiety, e.g. labelled avidin. Specific binding members of the present invention are designed .5 to be used in methods of diagnosis or treatment in human or animal subjects, preferably human. Accordingly, further aspects of the invention provide methods of treatment comprising administration of a specific binding !0 member as provided, pharmaceutical compositions comprising such a specific binding member, and use of such a specific binding member in the manufacture of a medicament for administration, for example in a method of making a medicament or pharmaceutical composition comprising formulating the 25 specific binding member with a pharmaceutically acceptable excipient. Clinical indications in which an anti-IL-13 antibody may be used to provide therapeutic benefit include asthma, atopic 30 dermatitis, allergic rhinitis, fibrosis, chronic obstructive pulmonary disease, inflammatory bowel disease, scleroderma and Hodgkin's lymphoma . As already explained, anti-IL-13 treatment is effective for all these diseases. 54 Anti-IL-13 treatment may be given orally, by injection (for example, subcutaneously, intravenously, intraperitoneal or intramuscularly), by inhalation, or topically (for example intraocular, intranasal, rectal, into wounds, on skin). The 5 route of administration can be determined by the physicochemical characteristics of the treatment, by special considerations for the disease or by the requirement to optimise efficacy or to minimise side-effects. LO It is envisaged that anti-IL-13 treatment will not be restricted to use in the clinic. Therefore, subcutaneous injection using a needle free device is also preferred. Combination treatments may be used to provide significant .5 synergistic effects, particularly the combination of an anti IL-13 specific binding member with one or more other drugs. A specific binding member according to the present invention may be provided in combination or addition to short or long acting beta agonists, corticosteroids, cromoglycate, leukotriene 20 (receptor) antagonists, methyl xanthines and their derivatives, IL-4 inhibitors, muscarinic receptor antagonists, IgE inhibitors, histaminic inhibitors, IL-5 inhibitors, eotaxin/CCR3 inhibitors, PDE4 inhibitors, TGF-beta antagonists, interferon-gamma, perfenidone, chemotherapeutic 25 agents and immunotherapeutic agents. Combination treatment with one or more short or long acting beta agonists, corticosteroids, cromoglycate, leukotriene (receptor) antagonists, xanthines, IgE inhibitors, IL-4 30 inhibitors, IL-5 inhibitors, eotaxin/CCR3 inhibitors, PDE4 inhibitors may be employed for treatment of asthma. Antibodies of the present invention can also be used in combination with corticosteroids, anti-metabolites, antagonists of TGF-beta and its downstream signalling pathway, for treatment of fibrosis. 55 Combination therapy of these antibodies with PDE4 inhibitors, xanthines and their derivatives, muscarinic receptor antagonists, short and long beta antagonists can be useful for treating chronic obstructive pulmonary disease. Similar 5 consideration of combinations apply to the use of anti-IL-13 treatment for atopic dermatitis, allergic rhinitis, chronic obstructive pulmonary disease, inflammatory bowel disease, scleroderma and Hodgkin's lymphoma. .0 In accordance with the present invention, compositions provided may be administered to individuals. Administration is preferably in a "therapeutically effective amount", this being sufficient to show benefit to a patient. Such benefit may be at least amelioration of at least one symptom. The S actual amount administered, and rate and time-course of administration, will depend on the nature and severity of what is being treated. Prescription of treatment, e.g. decisions on dosage etc, is within the responsibility of general practitioners and other medical doctors. Appropriate doses of .0 antibody are well known in the art; see Ledermann J.A. et al. (1991) Int. J. Cancer 47: 659-664; Bagshawe K.D. et al. (1991) Antibody, Immunoconjugates and Radiopharmaceuticals 4: 915 922. 25 The precise dose will depend upon a number of factors, including whether the antibody is for diagnosis or for treatment, the size and location of the area to be treated, the precise nature of the antibody (e.g. whole antibody, fragment or diabody), and the nature of any detectable label 30 or other molecule attached to the antibody. A typical antibody dose will be in the range 100ptg to 1 gm for systemic applications, and lpg to 1mg for topical applications. Typically, the antibody will be a whole antibody, preferably the IgG4 isotype. This is a dose for a single treatment of an 56 adult patient, which may be proportionally adjusted for children and infants, and also adjusted for other antibody formats in proportion to molecular weight. Treatments may be repeated at daily, twice-weekly, weekly or monthly intervals, 5 at the discretion of the physician. In preferred embodiments of the present invention, treatment is periodic, and the period between administrations is about two weeks or more, preferably about three weeks or more, more preferably about four weeks or more, or about once a month. 10 Specific binding members of the present invention will usually be administered in the form of a pharmaceutical composition, which may comprise at least one component in addition to the specific binding member. L5 Thus pharmaceutical compositions according to the present invention, and for use in accordance with the present invention, may comprise, in addition to active ingredient, a pharmaceutically acceptable excipient, carrier, buffer, ?0 stabiliser or other materials well known to those skilled in the art. Such materials should be non-toxic and should not interfere with the efficacy of the active ingredient. The precise nature of the carrier or other material will depend on the route of administration, which may be oral, or by 25 injection, e.g. intravenous. Pharmaceutical compositions for oral administration may be in tablet, capsule, powder or liquid form. A tablet may comprise a solid carrier such as gelatin or an adjuvant. Liquid 30 pharmaceutical compositions generally comprise a liquid carrier such as water, petroleum, animal or vegetable oils, mineral oil or synthetic oil. Physiological saline solution, dextrose or other saccharide solution or glycols such as 57 ethylene glycol, propylene glycol or polyethylene glycol may be included. For intravenous injection, or injection at the site of 5 affliction, the active ingredient will be in the form of a parenterally acceptable aqueous solution which is pyrogen-free and has suitable pH, isotonicity and stability. Those of relevant skill in the art are well able to prepare suitable solutions using, for example, isotonic vehicles such as Sodium LO Chloride Injection, Ringer's Injection, Lactated Ringer's Injection. Preservatives, stabilisers, buffers, antioxidants and/or other additives may be included, as required. A composition may be administered alone or in combination with L5 other treatments, either simultaneously or sequentially dependent upon the condition to be treated. Specific binding members of the present invention may be formulated in liquid or solid forms depending on the O physicochemical properties of the molecule and the route of delivery. Formulations may include excipients, or combinations of excipients, for example: sugars, amino acids and surfactants. Liquid formulations may include a wide range of antibody concentrations and pH. Solid formulations may be 25 produced by lyophilisation, spray drying, or drying by supercritical fluid technology, for example. Formulations of anti-IL-13 will depend upon the intended route of delivery: for example, formulations for pulmonary delivery may consist of particles with physical properties that ensure penetration 30 into the deep lung upon inhalation; topical formulations may include viscosity modifying agents, which prolong the time that the drug is resident at the site of action. 58 The present invention provides a method comprising causing or allowing binding of a specific binding member as provided herein to IL-13. As noted, such binding may take place in vivo, e.g. following administration of a specific binding 5 member, or nucleic acid encoding a specific binding member, or it may take place in vitro, for example in ELISA, Western blotting, immunocytochemistry, immuno-precipitation, affinity chromatography, or cell based assays such as a TF-1 assay. 10 The amount of binding of specific binding member to IL-13 may be determined. Quantitation may be related to the amount of the antigen in a test sample, which may be of diagnostic interest. L5 A kit comprising a specific binding member or antibody molecule according to any aspect or embodiment of the present invention is also provided as an aspect of the present invention. In a kit of the invention, the specific binding member or antibody molecule may be labelled to allow its ?0 reactivity in a sample to be determined, e.g. as described further below. Components of a kit are generally sterile and in sealed vials or other containers. Kits may be employed in diagnostic analysis or other methods for which antibody molecules are useful. A kit may contain instructions for use 25 of the components in a method, e.g. a method in accordance with the present invention. Ancillary materials to assist in or to enable performing such a method may be included within a kit of the invention. 30 The reactivities of antibodies in a sample may be determined by any appropriate means. Radioimmunoassay (RIA) is one possibility. Radioactive labelled antigen is mixed with unlabelled antigen (the test sample) and allowed to bind to the antibody. Bound antigen is physically separated from 59 unbound antigen and the amount of radioactive antigen bound to the antibody determined. The more antigen there is in the test sample the less radioactive antigen will bind to the antibody. A competitive binding assay may also be used with 5 non-radioactive antigen, using antigen or an analogue linked to a reporter molecule. The reporter molecule may be a fluorochrome, phosphor or laser dye with spectrally isolated absorption or emission characteristics. Suitable fluorochromes include fluorescein, rhodamine, phycoerythrin LO and Texas Red. Suitable chromogenic dyes include diaminobenzidine. Other reporters include macromolecular colloidal particles or particulate material such as latex beads that are coloured, .5 magnetic or paramagnetic, and biologically or chemically active agents that can directly or indirectly cause detectable signals to be visually observed, electronically detected or otherwise recorded. These molecules may be enzymes which catalyse reactions that develop or change colours or cause !0 changes in electrical properties, for example. They may be molecularly excitable, such that electronic transitions between energy states result in characteristic spectral absorptions or emissions. They may include chemical entities used in conjunction with biosensors. Biotin/avidin or 25 biotin/streptavidin and alkaline phosphatase detection systems may be employed. The signals generated by individual antibody-reporter conjugates may be used to derive quantifiable absolute or 30 relative data of the relevant antibody binding in samples (normal and test). The present invention also provides the use of a specific binding member as above for measuring antigen levels in a 60 competition assay, that is to say a method of measuring the level of antigen in a sample by employing a specific binding member as provided by the present invention in a competition assay. This may be where the physical separation of bound 5 from unbound antigen is not required. Linking a reporter molecule to the specific binding member so that a physical or optical change occurs on binding is one possibility. The reporter molecule may directly or indirectly generate detectable, and preferably measurable, signals. The linkage LO of reporter molecules may be directly or indirectly, covalently, e.g. via a peptide bond or non-covalently. Linkage via a peptide bond may be as a result of recombinant expression of a gene fusion encoding antibody and reporter molecule. L5 The present invention also provides for measuring levels of antigen directly, by employing a specific binding member according to the invention for example in a biosensor system. O The mode of determining binding is not a feature of the present invention and those skilled in the art are able to choose a suitable mode according to their preference and general knowledge. 25 As noted, in various aspects and embodiments, the present invention extends to a specific binding member which competes for binding to IL-13 with any specific binding member defined herein, e.g. BAK502G9 IgG4. Competition between binding members may be assayed easily in vitro, for example by tagging 30 a specific reporter molecule to one binding member which can be detected in the presence of other untagged binding member(s), to enable identification of specific binding members which bind the same epitope or an overlapping epitope. 61 Competition may be determined for example using ELISA in which IL-13 is immobilised to a plate and a first tagged binding member along with one or more other untagged binding members is added to the plate. Presence of an untagged binding member 5 that competes with the tagged binding member is observed by a decrease in the signal emitted by the tagged binding member. In testing for competition a peptide fragment of the antigen may be employed, especially a peptide including an epitope of LO interest. A peptide having the epitope sequence plus one or more amino acids at either end may be used. Such a peptide may be said to "consist essentially" of the specified sequence. Specific binding members according to the present invention may be such that their binding for antigen is [5 inhibited by a peptide with or including the sequence given. In testing for this, a peptide with either sequence plus one or more amino acids may be used. !0 Specific binding members which bind a specific peptide may be isolated for example from a phage display library by panning with the peptide(s). The present invention further provides an isolated nucleic 25 acid encoding a specific binding member of the present invention. Nucleic acid may include DNA and/or RNA. In a preferred aspect, the present invention provides a nucleic acid which codes for a CDR or set of CDR's or VH domain or VL domain or antibody antigen-binding site or antibody molecule, 30 e.g. scFv or IgG4, of the invention as defined above. The present invention also provides constructs in the form of plasmids, vectors, transcription or expression cassettes which comprise at least one polynucleotide as above. 62 The present invention also provides a recombinant host cell which comprises one or more constructs as above. A nucleic acid encoding any CDR or set of CDR's or VH domain or VL 5 domain or antibody antigen-binding site or antibody molecule, e.g. scFv or IgG4 as provided, itself forms an aspect of the present invention, as does a method of production of the encoded product, which method comprises expression from encoding nucleic acid therefor. Expression may conveniently .0 be achieved by culturing under appropriate conditions recombinant host cells containing the nucleic acid. Following production by expression a VH or VL domain, or specific binding member may be isolated and/or purified using any suitable technique, then used as appropriate. .5 Specific binding members, VH and/or VL domains, and encoding nucleic acid molecules and vectors according to the present invention may be provided isolated and/or purified, e.g. from their natural environment, in substantially pure or !0 homogeneous form, or, in the case of nucleic acid, free or substantially free of nucleic acid or genes origin other than the sequence encoding a polypeptide with the required function. Nucleic acid according to the present invention may comprise DNA or RNA and may be wholly or partially synthetic. 25 Reference to a nucleotide sequence as set out herein encompasses a DNA molecule with the specified sequence, and encompasses a RNA molecule with the specified sequence in which U is substituted for T, unless context requires otherwise. 30 Systems for cloning and expression of a polypeptide in a variety of different host cells are well known. Suitable host cells include bacteria, mammalian cells, plant cells, yeast and baculovirus systems and transgenic plants and animals. 63 Mammalian cell lines available in the art for expression of a heterologous polypeptide include Chinese hamster ovary (CHO) cells, HeLa cells, baby hamster kidney cells, NSO mouse melanoma cells, YB2/0 rat myeloma cells, human embryonic 5 kidney cells, human embryonic retina cells and many others. A common, preferred bacterial host is E. coli. The expression of antibodies and antibody fragments in prokaryotic cells such as E. coli is well established in the .0 art. For a review, see for example Pldckthun, A. Bio/Technology 9: 545-551 (1991). Expression in eukaryotic cells in culture is also available to those skilled in the art as an option for production of a specific binding memberfor example Chadd HE and Chamow SM (2001) 110 Current Opinion in .5 Biotechnology 12: 188-194, Andersen DC and Krummen L (2002) Current Opinion in Biotechnology 13: 117, Larrick JW and Thomas DW (2001) Current opinion in Biotechnology 12:411-418. Suitable vectors can be chosen or constructed, containing .0 appropriate regulatory sequences, including promoter sequences, terminator sequences, polyadenylation sequences, enhancer sequences, marker genes and other sequences as appropriate. Vectors may be plasmids, viral e.g. 'phage, or phagemid, as appropriate. For further details see, for 25 example, Molecular Cloning: a Laboratory Manual: 3rd edition, Sambrook and Russell, 2001, Cold Spring Harbor Laboratory Press. Many known techniques and protocols for manipulation of nucleic acid, for example in preparation of nucleic acid constructs, mutagenesis, sequencing, introduction of DNA into 30 cells and gene expression, and analysis of proteins, are described in detail in Current Protocols in Molecular Biology, Second Edition, Ausubel et al. eds., John Wiley & Sons, 1988, Short Protocols in Molecular Biology: A Compendium of Methods from Current Protocols in Molecular Biology, Ausubel et al. 64 eds., John Wiley & Sons, 4 th edition 1999. The disclosures of Sambrook et al. and Ausubel et al. (both) are incorporated herein by reference. 5 Thus, a further aspect of the present invention provides a host cell containing nucleic acid as disclosed herein. Such a host cell may be in vitro and may be in culture. Such a host cell may be in vivo. In vivo presence of the host cell may allow intracellular expression of the specific binding members LO of the present invention as "intrabodies" or intracellular antibodies. Intrabodies may be used for gene therapy [112]. A still further aspect provides a method comprising introducing such nucleic acid into a host cell. The L5 introduction may employ any available technique. For eukaryotic cells, suitable techniques may include calcium phosphate transfection, DEAE-Dextran, electroporation, liposome-mediated transfection and transduction using retrovirus or other virus, e.g. vaccinia or, for insect cells, 20 baculovirus. Introducing nucleic acid in the host cell, in particular a eukaryotic cell may use a viral or a plasmid based system. The plasmid system may be maintained episomally or may incorporated into the host cell or into an artificial chromosome [110,111]. Incorporation may be either by random or 25 targeted integration of one or more copies at single or multiple loci. For bacterial cells, suitable techniques may include calcium chloride transformation, electroporation and transfection using bacteriophage. 30 The introduction may be followed by causing or allowing expression from the nucleic acid, e.g. by culturing host cells under conditions for expression of the gene. 65 In one embodiment, the nucleic acid of the invention is integrated into the genome (e.g. chromosome) of the host cell. Integration may be promoted by inclusion of sequences which promote recombination with the genome, in accordance with 5 standard techniques. The present invention also provides a method which comprises using a construct as stated above in an expression system in order to express a specific binding member or polypeptide as .0 above. Aspects and embodiments of the present invention will now be illustrated by way of example with reference to the following experimentation. .5 EXAMPLE 1 Isolation of anti-IL-13 scFv ScFv antibody repertoire .0 A large single chain Fv (scFv) human antibody library derived from spleen lymphocytes from 20 donors and cloned into a phagemid vector was used for selections [66]. Selection of scFv 25 ScFv which recognised IL-13 were isolated from phage display libraries in a series of repeated selection cycles on recombinant bacterially derived human or murine IL-13 (Peprotech) essentially as described in [67]. In brief, following incubation with the library, the immobilised 30 antigen, which had been pre-coupled to paramagnetic beads, and bound phage were recovered by magnetic separation whilst unbound phage were washed away. Bound phage was then rescued as described by Vaughan et al [67] and the selection process repeated. Different solid surfaces and capture methods were 66 used at different rounds of selection to reduce non-specific binding. Antigen was either covalently coupled to beads (Dynabeads M-270 carboxylic acid) or modified by biotinylation prior to secondary capture by streptavidin-coated beads 5 (Dynabeads M-280) according to manufacturer's protocols (Dynal). A representative proportion of clones from the output of selection rounds were subjected to DNA sequencing as described in Vaughan et al [67] and Osbourn et al [70]. Unique clones were assessed for their ability to neutralise .0 IL-13 as purified scFv preparations in IL-13 dependent cell proliferation assays. Ribosome display libraries were created and screened for scFv that specifically recognised recombinant, bacterially derived .5 human or murine IL-13 (Peprotech), essentially as described in Hanes et al [113]. Initially the BAK278D6 lead clone from the initial selections was converted to ribosome display format, and this template was subsequently used for library creation. On the DNA level, a T7 promoter was added at the 5'-end for .0 efficient transcription to mRNA. On the mRNA level, the construct contained a prokaryotic ribosome-binding site (Shine-Dalgarno sequence). At the 3' end of the single chain, the stop codon was removed and a portion of gIII (gene III) was added to act as a spacer [113]. 25 Ribosome display libraries derived from BAK278D6 were created by mutagenesis of antibody complementarity determining regions (CDRs) where PCR reactions were performed with non-proof reading Taq polymerase. Affinity-based selections were 30 performed whereby, following incubation with the library, the biotinylated human-IL-13 was captured by streptavidin-coated paramagnetic beads (Dynal M280) and bound tertiary complexes (mRNA-ribosome-scFv-IL-13) were recovered by magnetic separation whilst unbound complexes were washed away. The 67 mRNA encoding the bound scFvs were then recovered by RT-PCR as described in Hanes et al [113] and the selection process repeated with decreasing concentrations (100nM - 100pM over 5 rounds) of biotinylated human IL-13 present during the 5 selection. Error-prone PCR was also used to further increase library size. Three intensities of error were employed (2.0, 3.5 and 7.2 mutations per 1,000 bp after a standard PCR reaction, as 0 described in manufacturer's protocol (Clontech)) during the selection regime. Initial error prone PCR reactions took place before round one selections commenced at 100nM. A subsequent round of error prone PCR was performed before round three selections at 10nM biotinylated human-IL-13. As above, a 5 representative proportion of clones from the output of selection rounds were subjected to DNA sequencing as described in Vaughan et al [67) and Osbourn et al [70]. Unique clones were assessed for their ability to neutralise IL-13 as purified scFv preparations in IL-13 dependent cell 0 proliferation assays. EXAMPLE 2 Neutralisation potency of anti-IL-13 scFv in the IL-13 dependent TF-1 cell proliferation assay 25 The neutralisation potency of purified scFv preparations against human and murine IL-13 bioactivity was assessed using TF-1 cell proliferation assay. Purified scFv preparations were prepared as described in Example 3 of WO01/66754. Protein 30 concentrations of purified scFv preparations were determined using the BCA method (Pierce). TF-1 is a human premyeloid cell line established from a patient with erythroleukemia [68]. The TF-1 cell line is factor dependent for survival and proliferation. In this respect TF-1 cells responded to either 68 human or urine IL-13 [69] and were maintained in media containing human GM-CSF (4 ng/ml, R&D Systems). Inhibition of IL-13 dependent proliferation was determined by measuring the reduction in incorporation of tritiated thymidine into the 5 newly synthesized DNA of dividing cells. TF-1 cell assay protocol TF-1 cells were obtained from R&D Systems and maintained according to supplied protocols. Assay media comprised RPMI .0 1640 with GLUTAMAX I (Invitrogen) containing 5% foetal bovine serum (JRH) and 1% sodium pyruvate (Sigma). Prior to each assay, TF-1 cells were pelleted by centrifugation at 300 x g for 5 mins, the media removed by aspiration and the cells resuspended in assay media. This process was repeated twice .5 with cells resuspended at a final concentration of 105 cells/ml in assay media. Test solutions of antibody (in triplicate) were diluted to the desired concentration in assay media. An irrelevant antibody not directed at IL-13 was used as a negative control. Recombinant bacterially derived human or 0 murine IL-13 (Peprotech) was added to a final concentration of 50 ng/ml when mixed with the appropriate test antibody in a total volume of 100 pl/well in a 96 well assay plate. The concentration of IL-13 used in the assay was selected as the dose that at final assay concentration gave approximately 80% 25 of the maximal proliferative response. All samples were incubated for 30 minutes at room temperature. 100 p1l of resuspended cells were then added to each assay point to give a total assay volume of 200 pl/well. Assay plates were incubated for 72 hours at 370C under 5% C02. 25 pl of tritiated 30 thymidine (10 ptCi/ml, NEN) was then added to each assay point and assay plates were returned to the incubator for a further 4 hours. Cells were harvested on glass fibre filter plates (Perkin Elmer) using a cell harvester. Thymidine incorporation 69 was determined using a Packard TopCount microplate liquid scintillation counter. Data were analysed using Graphpad Prism software. 5 Results Despite alternating selection cycles between human and murine antigen no cross-reactive neutralising antibodies were obtained. Two distinct anti-human and one anti-murine IL-13 neutralising scFvs were obtained from selections. BAK278D6 (VH .0 SEQ ID NO: 13; VL SEQ ID NO: 14) and BAK167A11 (VH SEQ ID NO: 23; VL SEQ ID NO: 24) recognised human IL-13 whilst BAK209Bll (VH SEQ ID NO: 25; VL SEQ ID NO: 26) recognised murine IL-13. BAK278D6 (Figure 2) and BAK167All (Figure 1) as scFv neutralised 25 ng/ml human IL-13 with an IC 50 of 44nM and lllnM .5 respectively. BAK209Bll (Figure 3) as a scFv neutralised 25 ng/ml murine IL-13 with an IC 50 of 185nM. EXAMPLE 3 Neutralisation potency of lead clones from targeted .0 optimisation of heavy chain CDR3 of parental clones in the IL 13 dependent TF-1 cell proliferation assay Osbourn et al. [70] have demonstrated that targeted mutagenesis of residues within heavy chain CDR3 can 25 significantly improve the affinity of antibodies. Selections were performed as described in Example 1, on scFv repertoires in which residues within the heavy chain CDR3 of BAK278D6 (SEQ ID NO: 6) BAK167All (SEQ ID NO: 57) had been randomised by mutagenesis. Unique clones from the selection output were 30 identified by DNA sequencing and their neutralising potency assessed as scFv in the TF-1 cell proliferation assay, as described in Example 2. 70 Results Significant gains in potency were achieved for both lineages. The most potent clones from the BAK167All lineage were BAK615E3, BAK612B5 and BAK582F7 which as scFv had IC50 of 3nM 5 (Figure 1), 6.6nM, 6.65nM respectively against 25ng/ml human IL-13 in TF-1 cell proliferation assay. From the BAK278D6 lineage, the most potent clone was BAK502G9, which as scFv had IC50 of 8nM against 25 ng/ml human IL-13 in the TF-1 cell proliferation assay (Figure 2). LO EXAMPLE 4 Neutralisation potency of BAK167A11 and BAK278D6 lineages against non-human primate IL-13 and an IL-13 variant associated with asthma in the TF-1 factor dependent cell .5 proliferation assay Neither of the BAK167All and BAK278D6 human IL-13 neutralising lineages were murine cross-reactive. The inventors therefore decided on the following criteria for the lineage selected for !0 further optimisation and clinical development: should preferably be cross-reactive with non-human primate IL-13 and should recognise a variant of IL-13, in which arginine at amino acid at position 130 is substituted for by glutamine (R130Q). This variant has been genetically associated with 25 asthma and other allergic diseases [37, 39, 41, 71]. Cross reactivity was determined by the ability of purified scFv preparations to bind non-human primate IL-13 and IL-13 variant by surface plasmon resonance (BIAcore) analysis. Functional activity was determined using the TF-1 cell proliferation 30 assay. Production of wild-type, variant and non-human primate IL-13 A cDNA for wild-type human IL-13 was obtained from InvivoGen and modified by site-directed mutagenesis (Stratagene 71 Quikchange@ kit) to yield a cDNA encoding variant IL-13. The coding sequence for both rhesus and cynomolgus monkey IL-13 was obtained by PCR on genomic DNA template using degenerate primers based on the human IL-13 sequence. Both non-human 5 primate (rhesus and cynomolgus) sequences were identical to each other but differed from human IL-13 by seven amino acids (Figure 19). Recombinant wild type, variant and non-human primate IL-13 were subsequently expressed using the baculovirus expression system (Invitrogen). Expression .0 constructs added a carboxyl terminus affinity tag to the expressed protein that allowed purification from insect cell conditioned media to near homogeneity. Qualitative binding assay using BlAcore .5 The binding affinity of purified scFv preparations to non human primate, variant and wild type IL-13 was determined by surface plasmon resonance measurements using a BIAcore 2000 Biosensor (BIAcore AB) as described in Karlsson et al [72]. In brief, IL-13 was coupled to CM5 sensorchips using an amine .0 coupling kit (BIAcore) at a surface density of approximately 200Ru and three concentrations of test scFv (approximately 350nM, 175nM and 88nM) in HBS-EP buffer passed over the sensor chip surface. The resulting sensorgrams were evaluated using BIA evaluation 3.1 software to provide relative binding data. 25 TF-1 assay protocol The assay was performed essentially as described in Example 2 with the following modifications: non-human primate IL-13, 30 human variant IL-13 (R130Q) and wild type human IL-13 were used at concentrations of 50 ng/ml, 25 ng/ml and 25 ng/ml respectively. 72 Results BIAcore binding assay data suggested that BAK278D6 but not BAK167Al1 lineage had the required cross-reactivity profile for further therapeutic development (Table 2). This finding 5 was supported by bioassay data demonstrating that BAK278D6 (Figure 4) and BAK502G9 (Figure 6) were able to neutralise human IL-13, the human IL-13 (R130Q) variant and non-human primate IL-13 in the TF-1 cell proliferation assay with near equivalent potency. In contrast, although BAK615E3 (VH SEQ ID .0 NO: 33; VL SEQ ID NO: 34) had a significantly increased potency against human IL-13 over its parent BAK167A11 (VH SEQ ID NO: 23; VL SEQ ID NO: 24) in the TF-1 cell proliferation assay (Figure 1), neither clone bound non-human primate or variant IL-13 in the BIAcore binding assay. .5 Germlining framework regions of BAK278D6 and BAK502G9 The derived amino acid sequence of BAK278D6 VH (SEQ ID NO: 13) and VL (SEQ ID NO: 14) were aligned to the known human germline sequences in the VBASE database [73) and the closest .0 germline identified by sequence similarity. The closest germline for the VH domain of BAK278D6 (SEQ ID NO: 14) and its derivatives, was identified as DPl4, a member of the VH1 family. The BAK278D6 VH has 9 changes from the DP14 germline within framework regions. The closest germline for the VL of 25 BAK278D6 was identified as VX3 3h. The BAK278D6 VL domain (SEQ ID NO: 14) has only 5 changes from the germline within framework regions. Framework regions of BAK278D6 and its derivatives were returned to germline by site directed mutagenesis (Stratagene Quikchange kit) to identically match 30 native human antibodies. EXAMPLE 5 Neutralisation potency of lead clones from targeted optimisation of heavy chain CDR1 and heavy chain CDR2 73 sequences of BAK502G9 in the human IL-13 dependent TF-1 cell proliferation assay A second phase of optimisation was performed using BAK502G9 5 sequence, with germlined framework regions, as a template. Selections were performed essentially as described in Example 1 on scFv repertories in which either residues within the heavy chain CDR1 or heavy chain CDR2 of BAK502G9 had been randomised by mutagenesis. Unique clones from the selection .0 output were identified by DNA sequencing and their neutralising potency assessed as purified scFv preparations in the TF-1 cell proliferation assay as described in Example 2. Vectors were constructed for the most potent scFv clones to allow re-expression as whole human IgG4 antibody as described .5 by Persic et al. (1997 Gene 187; 9-18) with a few modifications. An oriP fragment was included in the vectors to facilitate use with HEK-EBNA 293 cells and to allow episomal replication. The VH variable domain was cloned into the polylinker between the secretion leader sequence and the human 0 gamma 4 constant domain of the expression vector pEU8.1(+). The VL variable domain was cloned into the polylinker between the secretion leader sequence and the human lambda constant domain of the expression vector pEU4.1(-). 25 Whole antibody was purified from conditioned media from EBNA 293 cells co-transfected with constructs expressing heavy and light chains by protein A affinity chromatography (Amersham Pharmacia). The purified antibody preparations were sterile filtered and stored at 40C in phosphate buffered saline (PBS) 30 prior to evaluation. Protein concentration was determined by measuring absorbance at 280nm using the BCA method (Pierce). Reformatted human IgG4 whole antibodies were compared to commercially available anti-human IL-13 antibodies in the TF-1 proliferation assay described in Example 2. 74 Results As demonstrated in Figure 5, the commercial antibody B-B13, (mouse IgG1 -Euroclone 5) was shown to be significantly more potent against human IL-13 than the commercial antibody JES10 5 5A2 (rat IgG1 - Biosource) with IC5o of 1021pM and 47lpM respectively. Eight clones, namely, BAK1111D10, BAK1166GO2, BAK1167FO2, BAK1167FO4, BAK1183H4, BAK1184C8, BAK1185E1, BAK1185F8, derived from BAK502G9 (and so "BAK502G9 lineage"), in which the heavy chain CDR1 or CDR2 had been targeted, .0 showed improved potency as scFv over the commercial antibodies. These improvements were maintained on conversion to whole antibody human IgG4. Each of these VH and VL domains individually and in the respective pairings of these claims represents an aspect or embodiment of the present invention, .5 as do specific binding members for IL-13 that comprise one or more of them, also specific binding members comprising one or more CDR's from the BAK502G9 lineage clones, preferably a VH domain comprising a BAK502G9 lineage set of HCDR's and/or a VL domain comprising a BAK502G9 lineage set of LCDR's. These may .0 be employed in any and all aspects of the invention as disclosed elsewhere herein. Derivatives of BAK502G9 as whole antibodies (IgG4) had an IC50 ranging from 244pM to 283pM. BAK502G9 as a whole antibody IgG4 had an IC0o of 384pM. In summary, major improvements in potency could be obtained by 25 targeting heavy chain CDR1 (SEQ ID NO:7) or CDR2 (SEQ ID NO: 8) of BAK502G9. Statistical comparisons to B-B13 were made using an ANOVA followed by a Dunnett's post test analysis (InStat software). 30 Further characterisation Selected anti-human antibodies from the BAK278D6 lineage underwent further characterisation to determine their specificity. These included BAK502G9 (VH SEQ ID NO: 15; VL SEQ ID NO: 16) and its derivatives BAK1167F2 (VH SEQ ID NO: 35; VL 75 SEQ ID NO: 36) and BAK1183H4 (VH SEQ ID NO: 37; VL SEQ ID NO: 38), which are representative examples of clones with modifications to heavy chain CDR1 and heavy chain CDR2 of BAK502G9 respectively. 5 EXAMPLE 6 Neutralisation potency of lead clones from targeted optimisation of heavy chain CDR1 and heavy chain CDR2 sequences of BAK502G9 against non-human primate IL-13 and an .0 IL-13 variant associated with asthma in the TF-1 factor dependent cell proliferation assay Cross-reactivity of anti-human IL-13 antibodies was determined by their ability to inhibit non-human primate IL-13 and IL-13 .5 variant mediated TF-1 cell proliferation as described in Example 4. Results Optimised anti-human IL-13 antibodies BAK1167F2 (VH SEQ ID NO: .0 35; VL SEQ ID NO: 36) and BAKll83H4 (VH SEQ ID NO: 37; VL SEQ ID NO: 38) maintained the specificity of their parent BAK502G9 (VH SEQ ID NO: 15; VL SEQ ID NO: 16) (Figure 6). Potency gains against wild type IL-13 were reflected in their ability to neutralise non-human primate IL-13 and an IL-13 variant with 25 substantially equivalent potency. The IC 50 for BAK502G9 against human, human variant and non-human primate IL-13 were 1.4nM, 1.9nM and 2.OnM respectively. The IC 50 for BAKll67F2 against human, human variant and non-human primate IL-13 were 1.OnM, 1.lnM and 1.3nM respectively. The IC50 for BAK1l83H4 against 30 human, human variant and non-human primate IL-13 were 0.9nM, 1.OnM and 1.6nM respectively. These clones are suitable for therapeutic use. 76 EXAMPLE 7 Neutralising potency of lead anti-human IL-13 antibodies against native human IL-13 in HDLM-2 cell proliferation assay 5 The human IL-13 sequence has four potential N-glycosylation sites. The inventors have demonstrated the ability of BAK278D6 and its derivatives to neutralise recombinant IL-13 expressed either in bacterial or baculovirus expression systems. Although, there is evidence that many processing events known LO in mammalian systems do also occur in insects there are key differences in protein glycosylation, particularly N glycosylation [74]. The inventors investigated the ability of BAK278D6 derivatives .5 to neutralise native IL-13 released from human cells. HDLM-2 cells were isolated by Drexler et al [75] from a patient with Hodgkin's disease. Skinnider et al [76] demonstrated that HDLM-2 cell proliferation was in part 0 dependent on autocrine and paracrine release of IL-13. Lead anti-human IL-13 antibodies were assessed for their ability to inhibit HDLM-2 cell proliferation mediated by the release of native (or naturally occurring) IL-13. 25 HDLM-2 cell assay protocol HDLM-2 cells were obtained from the Deutsche Sammlung von Mikroorganismen und Zellkulturen (DSMZ) and maintained according to supplied protocols. Assay media comprised RPI 1640 with Glutamax I (Invitrogen) containing 20% foetal bovine 30 serum. Prior to each assay, the cells were pelleted by centrifugation at 300x g for 5 min, the media removed by aspiration and the cells resuspended in fresh media. This process was repeated three times and the cells were finally resuspended to a final concentration of 2 x 105 cells/ml in 77 assay media. 50pl of resuspended cells were added to each assay point in a 96 well assay plate. Test solutions of antibodies (in triplicate) were diluted to the desired concentration in assay media. An irrelevant isotype antibody 5 not directed at IL-13 was used as a negative control. The appropriate test antibody in a total volume of 50il / well were added to the cells, each assay point giving a total assay volume of 100ptl / well. Assay plates were incubated for 72 hours at 370C under 5% C02. 25 il of tritiated thymidine (10 LO pCi/ml, NEN) was then added to each assay point and assay plates were returned to the incubator for a further 4 hours. Cells were harvested on glass fibre filter plates (Perkin Elmer) using a cell harvester. Thymidine incorporation was determined using a Packard TopCount microplate liquid L5 scintillation counter. Data were analysed using Graphpad Prism software. Results As demonstrated in Figure 7, BAK502G9 (VH SEQ ID NO: 15; VL 20 SEQ ID NO: 16), and its derivatives BAKll83H4 (VH SEQ ID NO: 37; VL SEQ ID NO: 38) and BAKll67F2 (VH SEQ ID NO: 35; VL SEQ ID NO: 36) were able to cause a dose dependent inhibition of cell proliferation with relative potencies similar to those observed in other bioassays. IC50 for BAK502G9, BAKll83H4, 25 BAKll67F2 as human IgG4 were 4.6nM, 3.5nM and 1.lnM respectively. IC5o for the commercial antibodies JES10-5A2 and B-B13 were 10.7nM and 16.7nM respectively. EXAMPLE 8 30 Neutralising potency of lead anti-human IL-13 antibodies against IL-13 dependent responses in disease relevant primary cells 78 Secondary bioassays were performed using primary cells and readouts more relevant to airway disease. These included eotaxin release from normal human lung fibroblasts (NHLF) and vascular adhesion molecule 1 (VCAM-1) upregulation on the 5 surface of human umbilical vein endothelial cells (HUVEC). Both IL-13 dependent responses could contribute to eosinophil recruitment, a feature of the asthma phenotype [92]. NHLF assay protocol .0 IL-13 has been shown to cause ectaxin release from lung fibroblasts[77] [78] [79]. Factor dependent eotaxin release from NHLF was determined by ELISA. NHLF were obtained from Biowhittaker and maintained according .5 to supplied protocols. Assay media was FGM-2 (Biowhittaker). Test solutions of antibody (in triplicate) were diluted to the desired concentration in assay media. An irrelevant antibody not directed at IL-13 was used as a negative control. Recombinant bacterially-derived human IL-13 (Peprotech) was .0 subsequently added to a final concentration of 10 ng/ml when mixed with the appropriate test antibody in a total volume of 200 pl. The concentration of IL-13 used in the assay was selected as the dose that gave an approximately 80% of the maximal response. All samples were incubated for 30 minutes at 25 room temperature. Assay samples were then added to NHLF that had been preseeded at a density of 1 x 104 cells per well in 96-well assay plates. Assay plates were incubated at 37*C for 16-24 hours at 37 0 C under 5% C0 2 . Assay plates were centrifuged at 300 x g for 5 minutes to pellet detached cells. Eotaxin 30 levels in the supernatant were determined by ELISA using reagents and methods described by the manufacturer (R&D Systems). Data were analysed using Graphpad Prism software. 79 Results BAK278D6 lineage clones were able to inhibit human IL-13 dependent eotaxin release from NHLF. Relative potency was similar to that observed in the TF-1 cell proliferation assay 5 (Figure 8). BAK502G9 (VH SEQ ID NO: 15; VL SEQ ID NO: 16), BAK1183H4 (VH SEQ ID NO: 37; VL SEQ ID NO: 38), BAK1167F2 (VH SEQ ID NO: 35; VL SEQ ID NO: 36) had IC50 of 207pM, 118pM and 69pM respectively against 10 ng/ml human IL-13. Commercial antibodies JES10-5A2 and B-B13 had IC50 of 623pM and 219pM .0 respectively. HUVEC assay protocol IL-13 has been shown to upregulate expression of VCAM-1 on cell surface of HUVECs [80, 81]. Factor dependent VCAM-l .5 expression was determined by detection of upregulation of VCAM-l receptor cellular expression using a time-resolved fluorescence read out. HUVEC were obtained from Biowhittaker and maintained according :0 to supplied protocols. Assay media was EGM-2 (Biowhittaker). Test solutions of antibody (in triplicate) were diluted to the desired concentration in assay media. An irrelevant antibody not directed at IL-13 was used as a negative control. Recombinant bacterially derived human IL-13 (Peprotech) was 25 added to a final concentration of 10 ng/ml when mixed with the appropriate test antibody in a total volume of 200 pl. The concentration of IL-13 used in the assay was selected as the dose that gave approximately 80% of the maximal response. All samples were incubated for 30 minutes at room temperature. 30 Assay samples were then added to HUVEC that had been preseeded at 4 x 104 cells per well in 96-well assay plates. Assay plates were incubated at 370C for 16-20 hours under 5% C02. Assay media was then removed by aspiration and replaced with blocking solution (PBS containing 4% dried Marvel@ milk 80 powder). Assay plates were incubated at room temperature for 1 hour at room temperature. Wells were washed three times with PBST Tween before 100 pl (1:500 dilution in PBST/l% Marvel®) of biotinylated anti-VCAM-1 antibody (Serotec) was added to 5 each well. Assay plates were incubated at room temperature for 1 hour. Wells were washed three times with Delfia wash buffer (Perkin Elmer) before 100 pl of Europium-labelled Streptavidin or anti-murine IgG1 (1:1000 dilution in Delfia assay buffer, Perkin Elmer) was added to each well. Assay plates were then .0 incubated at RT for 1 hour. Wells were washed 7 times with Delfia wash buffer (Perkin Elmer). Finally, 100pl of enhancement solution (Perkin Elmer) was added to each well and fluorescence intensity was determined using the Wallac 1420 VICTOR2 plate reader (Standard Europium protocol). Data were .5 analysed using Graphpad Prism software. Results Typical data for BAK502G9 (VH SEQ ID NO: 15; VL SEQ ID NO: 16), BAKl183H4 (VH SEQ ID NO: 37; VL SEQ ID NO: 38), BAKll67F2 .0 (VH SEQ ID NO: 35; VL SEQ ID NO: 36) as whole antibody human IgG4 are shown in Figure 9. Relative potency was similar to the observed in the TF-1 cell proliferation assay. IC 50 for BAK502G9, BAKll83H4 and BAKll67F2 were 235pM, 58pM and 55pM respectively against lOng/ml human IL-13. 25 EXAMPLE 9 Neutralisation potency of anti-IL-13 antibodies against IL-1Q8 and IL-4 dependent VCAM-1 upregulation 30 The specificity of the BAK278D6 lineage of clones was assessed in a modification of the HUVEC bioassay. Together with IL-13, both IL-4 and IL-10 have been shown to upregulate expression of VCAM-1 on cell surface of HUVECs [80, 81]. 81 HUVEC assay protocol The assay was performed essentially as described in Example 5 with the following modifications. Recombinant human IL-1 and IL-4 (R&D Systems) were used in place of human IL-13 at 0.5 5 ng/ml and 1 ng/ml respectively and represented the dose that gave approximately 80% of the maximal response. Results None of the clones evaluated from the BAK278D6 lineage LO neutralised VCAM-l upregulation in response to either human IL-10 or IL-4 and thus demonstrated specificity for IL-13 (Figure 10). IL-4 is most closely related to IL-13, sharing 30% sequence identity at the amino acid level [82]. .5 EXAMPLE 10 Neutralisation potency of BAK2O9Bll as a human IgG4 in a murine IL-13 dependent murine B9 cell proliferation assay BAK209Bll, identified as an anti-murine IL-13 neutralising .0 clone as a scFv as described in Example 1, was reformatted as a whole antibody human IgG4 as described in Example 5 and its potency evaluated in the murine IL-13 dependent B9 cell proliferation assay. B9 is a murine B-cell hybridoma cell line [83]. B9 is factor dependent for survival and proliferation. 25 In this respect B cells respond to murine IL-13 and are maintained in media containing human IL-6 (50pg/ml, R&D Systems). Inhibition of murine IL-13 dependent proliferation was determined by measuring the reduction in incorporation of tritiated thymidine into the newly synthesized DNA of dividing 30 cells. B9 cell assay protocol B9 cells were obtained from European Collection of Animal Cell Culture ECACC and maintained according to supplied protocols. 82 The assay was performed essentially as described for the TF-1 assay in Example 2 but with the following modifications. Assay media comprised RPMI-1640 with GLUTAMAX I (Invitrogen) containing 5% foetal bovine serum (Hyclone) and 50ptM 2 5 mercaptoethanol (Invitrogen). Recombinant bacterially derived murine IL-13 (Peprotech) replaced human IL-13 with a final assay concentration of lng/ml. Results LO BAK209Bll (VH SEQ ID NO: 25; VL SEQ ID NO: 26) as a human IgG4 neutralised 1 ng/ml urine IL-13 with an IC 50 of 776pM in the B9 assay (Figure 11). BAK209Bll therefore represents a useful tool to investigate the role of IL-13 in murine models of disease. This is clearly demonstrated in Example 12, which L5 demonstrates the efficacy of BAK209B1l in a murine model of acute pulmonary inflammation. EXAMPLE 11 Affinity determination of anti-IL-13 antibodies by BlAcore ?0 analysis The affinity of BAK502G9 (VH SEQ ID NO: 15; VL SEQ ID NO: 16), BAKll67F2 (VH SEQ ID NO: 35; VL SEQ ID NO: 36) and BAKll83H4 (VH SEQ ID NO: 37; VL SEQ ID NO: 38) for human IL-13 and 25 BAK209Bll (VH SEQ ID NO: 25; VL SEQ ID NO: 26) for murine IL 13 as human IgG4 were determined by surface plasmon resonance measurements using a BIAcore 2000 Biosensor (BIAcore AB) essentially as described in [72]. In brief, antibodies were coupled to CM5 sensorchips using an amine coupling kit 30 (BIAcore) at a surface density of approximately 500Ru and a serial dilution of IL-13 (between 50nM to 0.78nM) in HBS-EP buffer was passed over the sensorchip surface. The resulting sensorgrams were evaluated using BIA evaluation 3.1 software to provide kinetic data. 83 Results BAK502G9, BAKll67F2 and BAKll83H4 IgG4 bound human IL-13 with high affinity with Kd of 178 pM, 136pM and 81pM respectively corresponding to their relative potency in cell based assays. 5 BAK209B11 bound murine IL-13 with affinity of 5.lnM (Table 3). EXAMPLE 12 Efficacy of BAK209B11 in a murine model of acute allergic 10 pulmonary inflammation Murine model of acute allergic pulmonary inflammation The effect of BAK209B1l (VH SEQ ID NO: 25; VL SEQ ID NO: 26), an anti-murine IL-13 neutralising human IgG4 antibody, was L5 investigated in a murine of acute allergic pulmonary inflammation. This model was performed essentially as described by Riffo-Vasquez et al [84] and is characterised at its endpoint by increased bronchial alveolar lavage (BAL) IL 13 (Figure 12), cellular infiltration into the lung and BAL !0 (Figure 13), increased serum IgE levels and airways hyperresponsiveness (AHR). Model protocol Female Balb/C mice (Charles River UK) were treated with either 25 anti-murine IL-13 antibody BAK209B1l (at 12, 36, 119 or 357 pg doses) or an isotype matched control antibody (357 pig dose). On days 0 and 7, mice in each group were sensitised by intraperitoneal injection of l0pg of ovalbumin (Ova) in 0.2 ml of the vehicle (saline containing 2% A1 2 0 3 (Rehydragel) as an 30 adjuvant). A separate control group of non-sensitised mice received an equal volume of the vehicle. Mice were challenged with ovalbumin on days 14, 15 and 16. Ovalbumin was diluted to 1% (w/v) in sterile saline prior to nebulisation. All inhalation challenges were administered in a Plexiglas 84 exposure chamber. Ova was aerosolised using a deVilbiss Ultraneb 2000 nebuliser (Sunrise Medical) in a series of three exposures of 20 minutes separated by 1 hour intervals. 5 BAK209B11 or an irrelevant human IgG4 were administered intravenously, 1 day prior to first challenge and then 2 hours prior to each subsequent challenge (4 doses in total). The model ended at day 17, 24 hours post final challenge. Blood (serum) and BAL were collected. Serum was assayed for total LO IgE. BAL was obtained by injecting 3 aliquots of saline (0.3ml, 0.3ml and 0.4ml) and pooling samples. Total leukocytes and differential cell counts were obtained from BAL cells. Results .5 Ovalbumin challenge of sensitised mice caused a significant (p<0.05) increase in total BAL cell recruitment over non sensitised but challenged animals. This recruitment was dose dependently inhibited by BAK209B1l; significant (p<0.05) inhibition was seen with 36pg BAK209Bl1, but not control .0 antibody (Figure 13). Similar effects were also seen on eosinophils (Figure 14) and neutrophils (Figure 15) with significant (p<0.05) inhibition of cellular influx at a minimum BAK209B11 dose of 36ptg. This inhibition was not seen with the control antibody. Lymphocytes were also induced in 25 sensitised but not non-sensitised mice upon challenge. This induction was dose-dependently inhibited by BAK209B11, with maximal inhibition seen with 36ptg BAK209B11. Control antibody had no effect (Figure 16). Although monocyte/macrophages were not induced in sensitised animals when compared to non 30 sensitised animals, background levels were depressed by 36pig BAK209B1l, but not by control antibody (Figure 17). Serum IgE levels were significantly increased in sensitised animals when compared to non-sensitised after challenge (p<0.05). This 85 increase was decreased after treatment with 36tg BAK209Bl1 but not by the control antibody. In summary, systemic administration of BAK209Bll, a murine IL 5 13 neutralising antibody, but not control antibody inhibited inflammatory cell influx and the upregulation of serum IgE levels caused by sensitisation and subsequent challenge with ovalbumin in a murine model of allergic inflammation. L0 Examples 13 to 20 are prophetic. EXAMPLE 13 Efficacy of BAK209Bll in the Lloyd murine model of acute pulmonary inflammation -5 Murine model of acute allergic pulmonary inflammation The effect of BAK209Bll (VH SEQ ID NO: 25; VL SEQ ID NO: 26), an anti murine IL-13 neutralising antibody, was investigated in a second murine model of acute allergic pulmonary inflammation. !0 This model was performed essentially as described by McMillan et al. [85] and is characterised at its endpoint by increased BAL and lung tissue IL-13, cellular infiltration into the lung and BAL, increased serum IgE levels and airways hyperresponsiveness (AHR). 25 Model protocol Female Balb/C mice (Charles River UK) were administered with various doses of anti-murine IL-13 antibody BAK209Bll or an isotype matched control antibody, as follows. On days 0 and 30 12, mice in each group were sensitised (SN) by intraperitoneal injection of lOgg of ovalbumin (Ova) in 0.2 ml of the vehicle (saline containing 2mg Al(OH) 3 as an adjuvant [calculated as described in Example 12]). A separate control group of non sensitised mice (NS) received an equal volume of the vehicle. 86 Mice were challenged with ovalbumin for 20 minutes on days 19, 20, 21, 22, 23 and 24. Ovalbumin was diluted to 5% (w/v) in saline prior to nebulisation. All inhalation challenges were administered in a Plexiglas exposure chamber. Ova was 5 aerosolised using a deVilbiss Ultraneb 2000 nebuliser (Sunrise Medical). On days 18,19,20,21,22,23 and 24 mice were administered with various intraperitoneal doses (237ptg, 23.7ptg or 2.37pg; denoted in figure 21 by H,M and L) of anti-murine IL-13 antibody BAK209B11 muIgGl or an isotype matched control .0 antibody (237pg). Airway function was assessed on days 0 and 25 by increasing methacholine challenges and monitored using conscious plethysmography (Buxco). PC 5 o (concentration of methacholine required to increase baseline PenH by 50%) was estimated for individual mice at both day 0 and day 25 from 4 .5 parameter unfixed curve fitting of methacholine dose-response curves. The model ended at day 25, 24 hours post final challenge. Blood, serum, BAL and lung tissue were collected. 0 Results Lung function was evaluated for individual animals at day 0 (pre-treatment) and at day 25 (post-challenge) and was quantitated by calculating PC 5 o values (concentration of 25 methacholine required to increase baseline PenH by 50%) (Figure 21A). An individuals airways hyperresponsiveness (AHR) was determined by the change in log PC 50 at day 25 versus day 0 (log day 25 PC 5 o - log day 0 PC 5 o) . This delta logPC 5 o was the primary endpoint of the study; PC 5 o data log 30 transformed because of requirements of endpoint ANOVA. Individual changes were averaged within groups to generate group average delta log PC 50 (as shown in Figure 21B) 87 Ovalbumin challenge of sensitised mice caused a significant AHR compared to non-sensitised and challenged mice (p<0.01). BAK209Bll caused a clear and dose-dependent decrease in AHR whereas the control antibody had no effect. 5 EXAMPLE 14 Efficacy of BAK209B11 in the Gerard murine model of acute pulmonary inflammation LO Murine model of acute allergic pulmonary inflammation The effect of BAK209Bll (VH SEQ ID NO: 25; VL SEQ ID NO: 26), an anti-murine IL-13 neutralising human IgG4 antibody, was investigated in a third murine model of acute allergic pulmonary inflammation. This model was performed essentially .5 as described by Humbles et al. [86] and is characterised at its endpoint by increased BAL and lung tissue IL-13, cellular infiltration into the lung and BAL, increased serum IgE levels and airways hyperresponsiveness (AHR). !0 Model protocol Female Balb/C mice (Charles River UK) were administered with various doses of anti-murine IL-13 antibody BAK209B1l or an isotype matched control antibody. On days 0, 7 and 14, mice in each group were sensitised (SN) by intraperitoneal injection 25 of 10ptg of ovalbumin (Ova) in 0.2 ml of the vehicle (saline containing 1.125mg Al(OH) 3 as an adjuvant [calculated as described in Example 12]). A separate control group of non sensitised mice (NS) received an equal volume of the vehicle. Mice were challenged with ovalbumin for 20 minutes on days 21, 30 22, 23 and 24. Ovalbumin was diluted to 5% (w/v) in saline prior to nebulisation. All inhalation challenges were administered in a Plexiglas exposure chamber. Ova was aerosolised using a deVilbiss Ultraneb 2000 nebuliser (Sunrise Medical). 88 The model ended at day 25, 24 hours post challenge. Blood, serum, BAL and lung tissue were collected. EXAMPLE 15 5 Efficacy of BAK2O9B1l in the Lloyd chronic model of pulmonary inflammation Murine model of chronic allergic pulmonary inflammation The effect of BAK209Bll (VH SEQ ID NO: 25; VL SEQ ID NO: 26), an LO anti murine IL-13 neutralising human IgG4 antibody, was investigated in a model of chronic allergic pulmonary inflammation. This model was performed essentially as described by Temelkovski et al. [87] and is characterised at its endpoint by cellular infiltration into the lung and BAL, [5 increased serum IgE levels and airways hyperresponsiveness (AHR). Model protocol Female Balb/C mice (Charles River UK) were dosed with various !0 doses of anti-murine IL-13 antibody BAK209Bl1 or an isotype matched control antibody. On days 0 and 11, mice in each group were sensitised (SN) by intraperitoneal injection of 10ptg of ovalbumin (Ova) in 0.2 ml of the vehicle (saline containing 2mg Al(OH) 3 as an adjuvant [calculated as described in Example 25 12]). A separate control group of non-sensitised mice (NS) received an equal volume of the vehicle. Mice were challenged with ovalbumin for 20 minutes on days 18, 19, 20, 21, 22, 23, 28, 30, 32, 35, 37, 39, 42, 44, 46, 49 and 51. Ovalbumin was diluted to 5% (w/v) in saline prior to nebulisation. All 30 inhalation challenges were administered in a Plexiglas exposure chamber. Ova was aerosolised using a deVilbiss Ultraneb 2000 nebuliser (Sunrise Medical). 89 The model ended at day 52, 24 hours post challenge. Blood, serum, BAL and lung tissue were collected. EXAMPLE 16 5 Efficacy of anti-human IL-13 antibodies against exogenous human IL-13 administered to the murine air pouch model The effect of anti-human IL-13 antibodies on the pro inflammatory action of human IL-13 was investigated in a basic LO murine model. This model was performed essentially as described by Edwards et al [93] and was characterised at its endpoint by cellular infiltration into the airpouch. Model protocol .5 An air pouch was created on the back of female Balb/C mice by subcutaneous injection of 2.5mL of sterile air at day 0. The air pouch was reinflated with another 2.5mL sterile air at day 3. 2ig huIL-13 in 0.75% CMC was injected directly into the pouch at day 6. 24 hours later the mice were killed and the !0 air pouch lavaged with lmL heparinised saline. Antibody treatments were either given with the huIL-13 (into the pouch) or given systemically. Results 25 Human IL-13, injected into the airpouch (i.po.), caused a significantly increased infiltration of total leukocytes (p<0.01) and eosinophils (p<0.01) at 24 hours post-challenge versus vehicle (0.75% carboxymethyl cellulose (CMC) in saline i.po.) treated mice. 30 Locally administered BAK502G9 (200mg, 20mg or 2mg intrapouch) significantly and dose-dependently inhibited the total leukocyte (p<0.01) and eosinophil (p<0.
0 1) infiltration into the air pouch caused by 2ig huIL-13 in 0.75% CMC. 90 Systemically administered BAK209B1l (30mg/kg, 10mg/kg and 1mg/kg) also signficantly and dose-dependently inhibited the total leukocyte (p<0.01) and eosinophil (p<0.01) infiltration 5 into the air pouch caused by 2pg huIL-13 in 0.75% CMC. EXAMPLE 17 Generation of human IL-13 knock-in / murine IL-13 knock out transgenic mice for the purposes of evaluating the efficacy of .0 anti-human IL-13 antibodies in models of pulmonary allergic inflammation The present inventors have generated mice which express human, rather than murine IL-13 by gene targeting. The mouse IL-13 .5 gene has been replaced from start to stop codon with the relevant portion of the human IL-13 gene. This mouse strain expresses human IL-13, rather than mouse IL-13, in response to the same stimuli as in the wild-type mouse, as the endogenous IL-13 promoter and IL-13 pA tail remaining unchanged. It has 10 been shown that human IL-13 can bind to and signal through mouse IL-13 receptors to generate the same physiological consequences as signalling caused by mouse IL-13 ligating mouse IL-13 receptors. For example exogenous human IL-13 caused inflammatory cell recruitment into the murine air pouch 25 (Figure 18). These transgenic animals allow us to evaluate non-murine cross reactive anti-human IL-13 antibodies in established murine models of disease. This mouse has been used in the acute allergic airway 30 inflammation models (as described in examples 18 and 19) and chronic allergic airway inflammation models (as described in Example 20) allowing the evaluation of anti-human IL-13 antibody pharmacology in allergic airway disease. 91 EXAMPLE 18 Efficacy of anti-human IL-13 antibodies in the huIL-13 transgenic Lloyd murine model of acute pulmonary inflammation 5 Murine model of acute allergic pulmonary inflammation The effect of anti human IL-13 neutralising human IgG4 antibodies were investigated in a murine model of acute allergic pulmonary inflammation using the transgenic mice .0 generated in example 17. This model was performed essentially as described by McMillan et al. [85] and example 13. The model was characterised at its endpoint by increased BAL and lung tissue IL-13, cellular infiltration into the lung and BAL, increased serum IgE levels and airways .5 hyperresponsiveness (AHR). Model protocol The protocol for this model was as described in Example 13 except that anti-human IL-13 antibodies were dosed instead of :0 BAK209B1l. EXAMPLE 19 Efficacy of anti-human IL-13 antibodies in the huIL-13 transgenic Gerard murine model of acute pulmonary inflammation 25 Murine model of acute allergic pulmonary inflammation The effect of anti human IL-13 neutralising human IgG4 antibodies were investigated in another murine model of acute 30 allergic pulmonary inflammation using the transgenic mice generated in example 17. This model was performed essentially as described by Humbles et al, [86] and in example 14. The model is characterised at its endpoint by increased BAL and lung tissue IL-13, cellular infiltration into the lung and 92 BAL, increased serum IgE levels and airways hyperresponsiveness (AHR). Model protocol 5 The protocol for this model was as described in Example 14 except that anti-human IL-13 antibodies were dosed instead of BAK209B11. EXAMPLE 20 .0 Efficacy of anti-human IL-13 antibodies in the huIL-13 transgenic Lloyd chronic model of pulmonary inflammation The effect of anti human IL-13 neutralising human IgG4 antibodies were investigated in a model of chronic allergic .5 pulmonary inflammation using the transgenic mice generated in example 17. This model was performed essentially as described by Temelkovski et al. [87) and in Example 15 and is characterised at its endpoint by cellular infiltration into the lung and BAL, increased serum IgE levels and airways .0 hyperresponsiveness (AHR). Model protocol The protocol for this model was as described in Example 15 except that anti-human IL-13 antibodies were dosed instead of 25 BAK209Bll EXAMPLE 21 Pharmacokinetics and pharmacodynamics of anti-human IL-13 antibodies in Ascaris.suum-allergic cynomolgus monkeys 30 The pharmacokinetics and pharmacodynamics of 502G9 were evaluated in 4 allergic but non-challenged cynomolgus primates (2 male/2 female) after a single 10mg/kg i.v bolus dose. The experiment ran for 29 days. The antibody's pharmacokinetic 93 parameters were determined from a geomean average serum-drug concentration curve and are detailed below in Table 4. In the same study serum IgE concentrations were also followed 5 using a human IgE ELISA kit (Bethyl laboratories, USA). Results Serum IgE concentrations were significantly reduced after a single 10mg/kg i.v bolus dose of BAK502G9, from 100 % control .0 levels (predose) to 66 ± 10% of control values (p<0.05), at 4 and 5 days after dosing. This lowering of serum IgE concentration recovered to 88 ± 8 % of control levels by day 22 (see Figure 20). Again these data were derived by normalising each animals serum IgE concentration to predose .5 levels, where predose concentrations was 100%, and then averaging the curves from the 4 animals tested. The two male monkeys had relatively low predose total serum IgE (60ng/mL and 67ng/mL). These IgE levels did not change in .0 a fashion suggesting a trend following treatment with BAK502G9 (Figure 20B). The two female monkeys had relatively high predose total serum IgE (1209ng/mL and 449ng/mL). These IgE levels were decreased following treatment with BAK502G9, maximally by 60% at 7 days, and returning to approximately 25 predose levels by 28 days post-administration (Figure 20B). These data provide indication that BAK502G9 lowers serum IgE concentrations in animals with relatively high circulating IgE of IgE. 30 EXAMPLE 22 Efficacy of anti-human IL-13 antibodies in cynomolgus models of dermal allergy The effects of anti-human IL-13 neutralising human IgG4 94 antibodies were investigated in a primate model of acute allergic dermal inflammation. This model was performed by injecting human IL-13 and A.suum antigen intradermally into cynomolgus monkeys. 24-96h later, dermal biopsies and serum 5 samples were taken. The model was characterised at its endpoint by cellular infiltration into the skin. EXAMPLE 23 Efficacy of anti-human IL-13 antibodies in cynomolgus models .0 of pulmonary allergy The effect of anti human IL-13 neutralising human IgG4 antibodies were investigated in a primate model of acute allergic pulmonary inflammation. This model was performed by .5 exposing a.suum-allergic cynomolgus primates to nebulised a.suum antigen, thereby generating an allergic reaction. This allergy was characterized at its end point by cellular infiltration into the lung and BAL, increased serum IgE levels and airways hyper-responsiveness. 0 Pharmacodynamics were additionally evaluated ex vivo using a flow cytometric method. CD23 is the high affinity IgE receptor and can be expressed on peripheral human blood mononuclear cells. CD23 expression can be induced, in terms 25 of the number of cells expressing CD23 and also in how much CD23 each cell expresses by both IL-13 and IL-4. The IL-13, but not IL-4, mediated response can be inhibited by anti-human IL-13 antibodies. 30 Animals were preselected for entry into this 2-phase study on the basis of previously established AHR following nebulised antigen (ascaris suum extract) challenge. In phase I airway function was assessed during intravenous histamine challenge on days 1 and 11. PC 30 , the histamine dose required to 95 generate a 30% increase in lung resistance (RL) above baseline, was determined from each histamine dose-response curve. On days 9 and 10, animals were challenged with individually tailored doses of nebulised antigen previously 5 shown to generate a 40% increase in RL as well as a 35% decrease in dynamic compliance (CDYN). Historically in this model, a greater RL has been observed following the second challenge with a given allergen dose than the first; this is antigen priming. The two antigen challenges caused AHR, as .0 measured by an increased area under the histamine dose response curve and/or a fall in PC 3 0, and BAL, as well as eosinophilia at day 11 compared to day 1. Animals displaying an AHR-phenotype were selected to enter phase II. .5 Phase II was run exactly as phase I except that all animals received a 30mg/kg BAK502G9 infusion on days 1, 5 and 9. The effects of BAK502G9 were assessed by comparing the changes seen in phase II with changes seen in phase I for individual animals. .0 Blood, serum, BAL and lung tissue were colleted. Serum IgE levels were monitored by ELISA. Serum from BAK502G9 treated cynomolgus monkeys was shown to inhibit the expression of CD23 on human peripheral blood mononuclear cells induced by IL-13 25 but not IL-4. The magnitude of this inhibition was consistent with the serum BAK502G9 levels predicted by PK ELISA. Results BAK502G9 significantly inhibited AHR as measured by RL AUC 30 (p<0.05) (Figure 26A; Table 7). An inhibitory effect of BAK502G9 on AHR, as measured by PC 3 o, was observed but did not reach statistical significance (Figure 26B; Table 7). BAK502G9 also significantly inhibited both antigen priming (p<0.01) (Figure 26C; Table 7) and BAL inflammation. BAK502G9 96 significantly inhibited total cell (p<0.05) and eosinophil (p<0.05) but not macrophage, lymphocyte or mast cell influx into the BAL (Figure 26D; Table 7). 5 EXAMPLE 24 Efficacy of anti-human IL-13 antibodies against the asthmatic phenotype that develops when human IL-13 is administered to the mouse lung 0 Murine model of airways hyperresponsiveness The efficacy of the anti-human IL-13 neutralising antibody BAK502G9, against the development of airways hyper responsiveness (AHR) following administration of human IL-13 to the mouse lung was investigated. This model was performed 5 essentially as described by Yang et al [119) with the exception that human IL-13 was used in place of murine IL-13. Model protocol To develop the phenotype, male BALB/c mice were exposed to two 0 doses of human IL-13 separated by a 48-hour interval. In brief, mice were anaesthetised with an intravenous injection of 100pl saffan solution (1:4 diluted in water). Mice were intubated with a 22-gauge catheter needle, through which human recombinant IL-13 (25 pg dissolved in 20pl phosphate-buffered 25 saline (PBS)) or vehicle control (PBS) was instilled. Airway function was assessed 24 hours after the last administration of IL-13 by increasing methacholine challenges and monitored using conscious plethysmography (Buxco) . PC 200 (concentration of methacholine required to increase baseline penH by 200%) 30 was determined from 4 parameter unfixed curve fitting of methacholine dose-response curves. Antibody treatments were administered by intra-peritoneal injection 24 hours prior to the each dose of IL-13. 97 Results Intratracheal installation of human IL-13 into naive wild-type mice resulted in development of significant (p<0.05) airways hyperresponsiveness relative to control animals as determined 5 by PC 2 oo methacholine concentrations. Systemically administered BAK502G9 (lmg/kg) significantly (p<0.01) inhibited the development of AHR whereas the null control antibody had no effect (Figure 23). 0 EXAMPLE 25 Neutralisation potency of BAK502G9 as a human IgG4 against human IL-13 dependent IgE release from human B cells. 5 B cell switching assay protocol IL-13 has been shown to induce IgE synthesis in human B cells in vitro [120]. Factor dependent IgE release from human B cells was determined by ELISA. The neutralisation potency of BAK502G9 as a human IgG4 was assessed against human IL-13 0 dependent IgE release from human B cells. Peripheral blood mononuclear cells (PBMC) were purified from human buffy coat (Blood Transfusion Service) by centrifugation over a 1.077g/L density gradient. B cells were purified from 25 PBMC with a B cell isolation kit II (Miltenyi Biotec), using reagents and methods described by the manufacturer. Assay media comprised Iscoves modified dulbeccos medium (Life Technologies) containing 10% foetal bovine serum and 20pg/mL human transferrin (Serologicals Proteins Inc). Following 30 purification, B cells were resuspended to a final concentration of 10 6 /mL in assay media. 50pl of resuspended cells were added to each assay point in a 96 well assay plate. 50pl of 4pg/mL of the anti-CD40 antibody EA5 (Biosource) was added to assay wells as appropriate. Test solutions of 98 antibodies (six replicates) were diluted to the desired concentration in assay media. An irrelevant antibody not directed at IL-13 was used as a negative control. 50pl / well of the appropriate test antibody were added to the cells. 5 Recombinant bacterially derived human IL-13 (Peprotech) was subsequently added to a final concentration of 30ng/ml to give a total assay volume of 200pl/well. The concentration of IL 13 used in the assay was selected to give a maximal response. Assay plates were incubated for 14 days at 37 0 c under 5% C02. .0 IgE levels in the supernatant were determined by ELISA using reagents and protocols supplied by the manufacturer (BD Biosciences, Bethyl Laboratories). Data were analysed using Graphpad prism software. 5 Results As demonstrated in Figure 24, BAK502G9 (VH SEQ ID NO: 15; VL SEQ ID NO: 16) was able to inhibit human IL-13 dependant IgE production by human B cells. BAK502G9 as human IgG4 had an IC50 of 1.8nM against 30ng/ml human IL-13. 0 EXAMPLE 26 Efficacy of BAK502G9 against IL-13 mediated potentiation of histamine induced Ca2 signalling in primary human bronchial smooth muscle cells 25 IL-13 has been shown to directly modulate the contractility of airway smooth muscle [121, 122]. Intracellular calcium mobilization is a prerequisite for smooth muscle contraction. Recent studies have shown that IL-13's ability to alter smooth 30 muscle contractility is mediated in part through modulation of contractile agonist induced Ca signaling [123, 124]. The efficacy of BAK502G9, an anti-human IL-13 antibody formatted as an IgG4, against IL-13 mediated alterations in 99 primary human bronchial smooth muscle cells (BSMC) signalling responses to the contractile agonist, histamine, was investigated in a Ca2+ signalling assay. 5 BSMC Ca2+ signalling assay protocol Human primary BSMC, Smooth Muscle Growth Medium-2 (SmGM-2) and Smooth Muscle Basal Medium (SmBM) were obtained from Bio Whittaker. The BSMC were maintained in SmGM-2 according to supplier's recommendations. BSMC were plated at 2 x 10 4 0 cells/well in a 96-well microtitre cell culture plate and were allowed to attach for 24 hours, then re-fed and incubated for a further 24 hours. Prior to the Ca2+ signalling experiment, the BSMC were stimulated with IL-13 (Peprotech) at 50ng/ml final concentration with or without antibody and incubated for 5 18-24 hours. BAK502G9 and an isotype matched irrelevant control monoclonal antibody, CAT-001, were evaluated at a final concentration of lOpg/ml. Changes in intracellular Ca2+ concentrations in response to histamine (Calbiochem), titrated from 20pM, were measured using standard techniques with the o Ca2+ sensitive dye Fluo-4 (Molecular Probes) and a 96-well Fluorescence Imaging Plate Reader (FLIPR) (Molecular Devices). The area under the curve (AUC) of the Ca2+ signalling response to histamine was determined for each cell pre-treatment condition. Data analyses were performed using GraphPad Prism 25 version 4 for Windows (GraphPad Software). Results Pre-incubation of BSMC with IL-13 significantly enhanced Ca2+ signalling in response to histamine. Pre-incubation of 30 BAK502G9 (Figure 25B) (but not an irrelevant isotype control antibody (Figure 25A)) with IL-13 significantly inhibited the potentiation of Ca2+ signalling in response to histamine (Figure 25). 100 EXAMPLE 27 Neutralisation potency of anti-IL-13 antibodies in a human IL 13 dependent PBMC CD23 expression assay 5 The potency of a representative IL-13 antibody was evaluated in the human IL-13 dependent peripheral blood mononuclear cell (PBMC) CD23 expression assay. PBMC respond to both IL-13 and IL-4 by increasing cell surface expression of CD23 [120]. CD23 (FceRII) is the low-affinity receptor for IgE and is LO expressed on a variety of inflammatory cells, including monocytes. Inhibition of human IL-13 dependent CD23 expression upregulation was determined by measuring the reduction in binding of fluorescently labelled CD23 monoclonal antibody to PBMCs by flow cytometry. -5 Assay protocol Human blood was obtained from the Blood Transfusion Service and erythrocytes depleted by 40 minute dextran-T500 (Pharmacia) sedimentation (0.6% final concentration). The .0 leukocyte and platelet rich fraction was then separated by a 20 minute 1137g centrifugation over a discontinuous Percoll gradient of 3mL 64% and 5mL 80% (100% was 9 parts Percoll and 1 part 10x PBS). PBMCs were harvested from the top of the 64% layer, washed and resuspended in assay buffer (Invitrogen RPMI 25 1640, 10% FCS, 200IU/mL penicillin, 100pg/mL streptomycin, 2mM L-Glutamine). The assay was performed in 24 well plates with 2x10 6 cells, ± 80pM recombinant human IL-13 (Peprotech) or 21pM recombinant human IL-4 (R&D Systems), ± BAK502G9 or irrelevant IgG4, in a final volume of 500mcL. Cells were 30 cultured for 48h at 37C before being harvested and stained with CD23-PE (BD Pharmingen) for 20 minutes at 4C. Finally, cells were read on a flow cytometer. CD23 expression was determined by CD23 'score'; percent of CD23 positive cells multiplied by the 'brightness' of the stain (geomean 101 fluorescence). No stimulant CD23 'score' was subtracted and data presented as a percentage of the response to IL-13 alone (100%). Data has been expressed as the mean ± SEM drawn from 4-6 separate experiments, using cells from 4-6 individual 5 donors, performed in triplicate for each point. Results Incubation of PBMC with 8OpM IL-13 or 21pM IL-4 for 48 hours resulted in clear CD23 expression (Figure 27 and Figure 28). .0 BAK502G9 dose-dependently inhibited IL-13-induced CD23 expression with a geometric mean of 120.2pM (Figure 27). In contrast, BAK502G9 was unable to inhibit the CD23 expression induced by 21.4pM IL-4 (n=4 from individual donors, Figure 28). Irrelevant IgG4 did not inhibit either IL-13 or IL-4 .5 dependent CD23 expression on PBMC (Figure 27 and Figure 28). Co-stimulation of PBMC with 80pM IL-13 and 21.4pM IL-4, produced an additive CD23 response. BAK502G9, but not CAT 001, reduced CD23 expression levels to those seen with IL-4 stimulation alone (Figure 28). 0 EXAMPLE 28 Neutralisation potency of a human IL-13 antibody in a human IL-13 dependent eosinophil shape change assay 25 The aims of this study were to evaluate the effect of IL-13 antibodies on eosinophil shape change induced by mediators released from NHLF following stimulation with factors found in the lungs of asthmatics such as IL-13 (125,126], TNF-ax [127], TGF-31 [128]. IL-13 synergises with TNF-x [129] or TGF-P1 30 [130] to induce fibroblasts to produce eotaxin-1, which can then act to directly chemoattract eosinophils. Leukocyte shape change responses are mediated through rearrangements of the cellular cytoskeleton and are essential to the processes of leukocyte migration from the microcirculation into sites of 102 inflammation. Inhibition of IL-13-dependent shape-change inducing factor release by NHLFs was determined by measuring the reduction in eotaxin-1secretion by ELISA and reduction in eosinophil shape change by flow cytometry. 5 Assay protocol NHLF cells were cocultured with media alone or media containing stimulants (9.6nM IL-13, 285.7pM TNF-a (R&D Systems) and 160pM TGF-1 (R&D Systems) in the absence or .0 presence of BAK502G9 (concentration range 875nM - 6.84nM). Cells were then cultured for a further 48h at 370C before the resulting conditioned media was aspirated and stored at -80 0 C. The concentration of eotaxin-1 in conditioned media was assessed using the R&D systems Duoset ELISA system (R&D .5 Systems). Human blood was obtained from the Blood Transfusion Service and erythrocytes depleted by 40 minute dextran-T500 (Pharmacia) sedimentation (0.6% final concentration). The :0 leukocyte and platelet rich fraction was then separated by a 20 minute 1137g centrifugation over a discontinuous Percoll gradient of 3mL 64% and 5mL 80% (100% was 9 parts Percoll and 1 part lOx PBS). Granulocytes were harvested from the 64%:80% interface, washed and resuspended in assay buffer (Sigma PBS, 25 1mM CaCl2, 1mM MgCl2, 10mM HEPES, 10mM D-glucose, 0.1% Sigma BSA, pH 7.3). The assay was performed in FACS tubes with 5x10 5 cells, ± 3nM recombinant human eotaxin-1 (R&D Systems) or conditioned media, in a final volume of 400pL. Cells were incubated for 8.5 minutes at 37C before being transferred to 30 40C and fixed with a fixing agent (CellFix, BD Biosciences) and finally read on a flow cytometer. Eosinophils were identified by their FL-2 autofluorescence and the forward scatter (FSC) parameter read. Eosinophil FSC changed in response to both eotaxin-1 and conditioned media providing 103 measurement of shape change. Tubes were sampled at high flow rate and acquisition was terminated after 1000 eosinophil events or 1 minute, whichever was the sooner. Shape change was calculated as a percentage of the FSC caused by shape 5 change buffer alone (100% blank shape change). Data have been expressed as the mean % blank shape change ± SEM drawn from 4 separate experiments. Each experiment used cells from an individual buffy coat (and hence individual donor), performed in duplicate for each point. .0 Results NHLF cells co-stimulated with 9.6nM IL-13, 285.7pM TNF- a and 160pM TGF-P1 and cultured for 48h secreted 9.6nM eotaxin-1 into the culture media. In contrast, NHLF cells cultured only .5 with maintenance media secreted 0.lnM eotaxin-1 into the culture media. This eotaxin-1 production was IL-13 dependent as IL-13/TNF-a/TGF-p1 co-stimulated NHLF cell eotaxin-1 production was dose-dependently inhibited by BAK502G9 with an
IC
50 of 32.4nM (Figure 29A) .0 The primary aim of this part of the study was to examine eosinophil shape change. The magnitude of eosinophil shape change in response to 3nM eotaxin (positive control) was 122.2±2.1% (n=4). Eotaxin-l induced shape change was 25 completely inhibited by 100nM of an anti-eotaxin antibody CAT 213, mean shape change 101.0±1.0% (n=4). Media from NHLF cells co-stimulated with 9.6nM IL-13, 285.7pM TNF-a and 160pM TGF-Pl and cultured for 48h (conditioned 30 media), induced a clear eosinophil and shape change (Figure 29B). In contrast, media from NHLF cultured for 48h in NHLF maintenance media alone did not induce eosinophil shape change (Figure 29B). 104 The addition of anti-IL-13 antibody BAK502G9 to co-stimulated media prior to NHLF culture, resulted in a dose-dependent inhibition of eosinophil shape change, with a geometric mean
IC
50 of 16.8nM when assayed at 1:16 dilution (Figure 29B). 5 The ability of stimulants (IL-13, TNF-a and TGF-01) not cultured with NHLF cells to induce eosinophil and neutrophil shape change was also investigated. 9.6nM IL-13, 285.7pM TNF a and 160pM TGF-Pl did not induce a clear eosinophil shape .0 change. This suggests that the eosinophil shape change ability of conditioned media develops during NHLF cell culture with the stimulants is not due to any of the stimulants alone or in combination (Figure 29B). .5 Example 29 Mapping of anti-IL-13 antibodies on Human IL-13 The epitope mapping of a representative IL-13 antibody BAK502G9 was performed using a molecular approach and standard .0 peptide excision. Molecular Approach IL-13 chimaeras were engineered, where parts of the human IL 13 sequence were replaced with murine sequence. These chimeras 25 were used in binding studies with representative IL-13 antibodies to help identify the specific epitope. Two panels of IL-13 chimaeras were produced. The first panel contained nine chimaeras (Figure 30) and was used to locate 30 the general position of the epitope. The second panel contained ten chimaeras (Figure 31) and was used to fine map the epitope. 105 The chimaeric IL-13 sequences were assembled using PCR and cloned into a Gateway entry vector, which were then recombined with a destination vector pDEST8 (modified to code for a detection and affinity tag at the C-terminus of the 5 recombinant protein). These expression vectors were used to transform DHl0BacTM chemically competent E coli allowing site specific transposition of tagged chimeric IL-13, into the baculovirus shuttle vector (bacmid). Recombinant bacmid DNA was isolated for each IL-13 chimera and transfected into Sf9 .0 (Spodoptera frugiperda) insect cells using Cellfectin® Reagent. Recombinant baculovirus was harvested 72 hours post transfection and passaged through Sf9 insect cells twice more. Insect 2000-500 ml culture supernatant was purified on an 5 affinity column and eluted material was concentrated from 16 to 1 ml and loaded on a size exclusion Superdex 200 HR10/300GL column for final polishing and buffer exchange. A homogenous competition assay using biotinylated human IL-13, .0 streptavidin-anthophyocynate and Europium labelled BAK502G9 was developed. The assay is as follows: Eu-BAK502G9 binds biotinylated-human IL-13, the complex is then recognised by the streptavidin APC conjugate and when a flash of light is applied the energy is transferred from the APC label to the 25 Europium by proximity, and time resolved florescence can be measured. Competition for this binding is introduced by way of the un-labelled human IL-13 (as control) and the chimeric constructs. This competition is quantified to calculate the relative affinities of the IL-13 mutants for IL-13 antibodies 30 enabling mutations altering binding to be identified. Results Chimeric construct IL13-Helix D (Table 5) was found to be the weakest competitor against biotinylated human IL-13 for 106 binding BAK502G9, indicating that helixD within the IL-13 molecule was involved with BAK502G9 epitope binding (Table 5) Reduced activity was also seen for the 4041 and 3334 mutants where residues 40, 41, and 33, 34 of the parent sequence 5 respectively were changed indicating potential involvement of helixA in the recognition of BAK502G9. The reduced activities of loop3 was discounted as this loop has a reduced number of amino acids in the mutant as compared to the human molecule and is likely to alter the overall structure of the protein. .0 Other reductions in the ability of the chimeric IL-13 molecules to compete for BAK502G9 binding were not considered significant for such amino acid changes. A more targeted set of mutations within helix D (Figure 26) .5 were then tested. Results obtained are demonstrated in Table 6 and are as follows: Results show that chimeric constructs 116117TK (where lysine at position 116 was replaced with threonine and the aspartate !0 at position 117 was replaced with lysine), 123KA (where lysine at position 123 was replaced) and 127RA (where arginine at position 127 was replaced) are least able to compete for binding to BAK502G9 (123KA and 127RA do not compete at 1 pM). Other residues implicated in binding to BAK502G9 due to their 25 reduced effectiveness in the competition assay include the helixD residues 124Q (here lysine has been replaced with glutamine) and 120121SY (a leucine histidine pair has been changed to a serine tyrosine pair). Mutation of leucine at position 58L also reduces binding and analysis of the 3D 30 structures revealed that this residue packs against helixD and may either be directly contacted by BAK502G9 or may affect the alignment of helixD. 107 These experiments demonstrate that residues within helixD are critical for the binding of BAK502G9 to IL-13. In particular the lysine at position 123 and the arginine at position 127 are critical for this binding as mutation to either abolishes 5 binding of BAK502G9. Epitope Excision The epitope mapping of BAK502G9 was also performed using the standard peptide excision procedure. Here IgG is immobilised 10 onto solid phase and allowed to capture the IL-13 ligand. The formed complex is then subject to specific proteolytic digestion, during which accessible peptide bonds are cleaved, however those protected by the IgG: ligand interface remain intact. Thus, a peptide containing the epitope remains bound L5 to the IgG. This can then be desorbed, collected and identified by mass spectrometry (ms). Two complementary techniques were used, the first made use of the Ciphergen ProteinChip Reader MALDI-TOF mass spectrometer, 0 where it was possible to covalently link the IgG to a mass spectrometer chip and then perform the digestion and extraction in-situ. The second technique used biotinylated BAK502G9 linked to streptavidin coated beads and allowed the collection of sufficient peptide for sequence confirmation by 25 tandem mass spectrometry (ms/ms). The two procedures although differing in absolute detail and scale involved essentially the same steps, coupling of the IgG, blocking of unreacted binding sites, washing, ligand 30 capture, removal of unbound ligand, digestion and a final washing step. The MALDI-TOF ms approach made use of proprietary ms chips activated with carbonyldiimidazole that covalently binds to 108 free primary amine groups to which the IgG at 1-2 mg/ml in PBS was coupled to overnight at 40C. The chip was subsequently blocked with an ethanolamine solution at room temperature for 1 hour and then washed extensively with PBS or HBS plus a 5 suitable detergent. A one picomole aliquot of IL-13 was then applied to the chip in either PBS or HBS and allowed to bind to the chemically immobilized IgG for 2 hours at room temperature. This was followed by further washes in PBS or HBS with and without detergent to remove any non-specifically .0 bound IL-13. A solution of trypsin ranging from 200 to 3.lpg/ml in PBS or HBS was then applied to the IgG:ligand complex and digestion allowed to proceed for 30 minutes at room temperature after which the chip was washed in PBS or HBS plus detergent, PBS or HBS and finally water. After .5 application of a suitable MALDI-TOF ms matrix the chip was then be placed directly in the mass spectrometer and analysed. The bead based approach started with the biotinylation of the IgG, using an NHS biotin compound, at a molar ratio of 1 IgG .0 to 4 biotin molecules. Removal of unbound biotin and the by products of the reaction using gel filtration followed this. The biotinylated IgG was then allowed to bind to neutravidin coated agarose beads, where it was attempted to maximize the IgG capture. Aliquots of IgG coated beads were then dispensed 25 into a concentrator spin columns and washed with Dulbecco's PBS + 0.05% Tween 20 followed by resuspension in Dulbecco's PBS + 0.05% Tween 20. A pulse of IL-13 was then applied to the resuspended IgG beads and binding allowed to proceed for 10 minutes after which the liquid phase was removed by 30 centrifugation and the beads washed with Dulbecco's PBS + 0.05% Tween 20 followed by resuspension in Dulbecco's PBS + 0.05% Tween 20. 109 The bead:IgG:ligand complex was then subject to proteolysis with either trypsin or chymotrypsin with incubation at room temperature or 370C. After which the beads were again washed in Dulbecco's PBS + 0.05% Tween 20 followed by a further 5 washes in Dulbecco's PBS without detergent. The beads were then resuspended in a water, acetonitrile, trifluroacetic mix and the supernatant recovered. This was then variously analysed either by MALDI-TOF ms or by reverse phase HPLC mass spectrometry, including tandem (ms/ms) fragmentation using the [0 ThermoQuest LCQ ESI ion-trap mass spectrometer. An attempt was then made to match the resulting fragmentation pattern to the human IL-13 sequence and the separate heavy and light chain sequence of BAK502G9 IgG. .5 During the experimental sequence a number of controls, primarily blank surfaces, IgG only and isotype controls were employed to demonstrate that the identified peptides were derived specifically from IgG captured IL-13 and not a product of BAK502G9 or non-specifically bound IL-13 digestion. 0 Results The experimental series consistently gave single IL-13 specific peptides for each digestion. Data from the LCQ ion trap instrument revealed that the tryptic fragment had a 25 monoisotopic mass of 3258Da (MH+) and the chymotrypsin fragment a monoisotopic mass of 3937Da (MH+). A search of these masses against the appropriate in silico digestion of human IL-13 gave close matches to related 30 peptides in the C-terminal portion of the molecule. 110 Match for trypsin peptide mass: 3258Da At a tolerance of 1000ppm, 3258Da matches to the sequence from 5 aspartic acid at position 106 to the C-terminal asparagine at position 132. There are no other matches at this tolerance. This region is highlighted in bold on the sequence of the precursor form of human IL-13 below. 0 MALLLTTVIALTCLGGFASPGPVPPSTALRELIEELVNITQNQKAPLCNGSMVWSINLTAGM YCAALESLINVSGCSAIEKTQRMLSGFCPHKVSAGQFSSLHVRDTKIEVAQFVKDLLLHLKK LFREGRFN Match for chymotrypsin peptide mass: 3937Da .5 At a tolerance of 1000ppm, 3937Da matches to the sequence from serine at position 99 to the C-terminal asparagine at position 132. This region is highlighted in bold on the sequence of the precursor form of human IL-13 below. '0 MALLLTTVIALTCLGGFASPGPVPPSTALRELIEELVNITQNQKAPLCNGSMVWSINLTAGM YCAALESLINVSGCSAIEKTQRMLSGFCPHKVSAGQFSSLHVRDTKIEVAQFVKDLLLHLKK LFREGRFN 25 Both these matches show that the BAK502G9 IgG retains the C terminal portion of the IL-13 molecule during proteolysis of the antibody:ligand complex. The identity of both peptides was successfully confirmed by 30 the ms/ms, neither of which showed any significant sequence parallels with BAK502G9. The ms/ms fragment map tailored to identify either Y or B ions matched 26 of 104 possible ions in one charge state for the trypsin peptide and 19 of 128 possible ions for the chymotrypsin peptide. A review of all 111 charge states shows identification of 23 of the 27 amino acid residues for the trypsin fragment and 29 of the 33 residues for the chymotrypsin fragment. This is sufficient to confirm identity. 5 The experimental sequence as a whole has identified that part of the BAK502G9 epitope on human IL-13 as lying within the twenty-seven C-terminal amino acid residues. These findings corroborate the finding of the molecular approach detailed .0 above. REFERENCES 1. McKenzie, A.N., et al. J Immunol, 1993. 150(12): p. 5436 44. .5 2. Minty, A., et al. Nature, 1993. 362(6417): p. 248-50. 3. Nakamura, Y., et al. Am J Respir Cell Mol Biol, 1996. 15(5): p. 680-7. 4. Robinson, D.S., et al. N Engl J Med, 1992. 326(5): p. 298-304. 0 5. Walker, C., et al. Am J Respir Crit Care Med, 1994. 150(4): p. 1038-48. 6. Humbert, M., et al. Am J Respir Crit Care Med, 1996. 154(5): p. 1497-504. 7. Corrigan, C.J. and A.B. Kay Int Arch Allergy Appl 25 Immunol, 1991. 94(1-4): p. 270-1. 8. Bentley, A.M., et al. Am J Respir Cell Mol Biol, 1993. 8(1): p. 35-42. 9. Murata, T., et al. Int J Hematol, 1999. 69(1): p. 13-20. 10. Andrews, A.L., et al. J Biol Chem, 2002. 277(48): p. 30 46073-8. 11. Miloux, B., et al. FEBS Lett, 1997. 401(2-3): p. 163-6. 12. Hilton, D.J., et al. Proc Natl Acad Sci U S A, 1996. 93(1) : p. 497-501. 13. Kuperman, D., et al. J Exp Med, 1998. 187(6): p. 939-48. 112 14. Nelms, K., et al. Annu Rev Immunol, 1999. 17: p. 701-38. 15. Zhang, J.G., et al. J Biol Chem, 1997. 272(14): p. 9474 80. 16. Caput, D., et al. J Biol Chem, 1996. 271(28): p. 16921-6. 5 17. Kawakami, K., et al. Blood, 2001. 97(9): p. 2673-9. 18. Wood, N., et al. J Exp Med, 2003. 197(6): p. 703-709. 19. Chiaramonte, M.G., et al. J Exp Med, 2003. 197(6): p. 687-701. 20. Beasley, R., et al. J Allergy Clin Immunol, 2000. 105(2 0 Pt 2): p. S466-72. 21. Peat, J.K. and J. Li J Allergy Clin Immunol, 1999. 103(1 Pt 1): p. 1-10. 22. Society, B.T., British guideline on the management of asthma. Thorax, 2003. 58 Suppl 1: p. il-94. 5 23. GINA, Global Strategy for Asthma Management and Prevention. 2002, National Insitute of Health. 24. Milgrom, H., B. Bender, and F. Wamboldt. Ann Allergy Asthma Immunol, 2002. 88(5): p. 429-31. 25. Fish, L. and C.L. Lung, Adherence to asthma therapy. Ann .0 Allergy Asthma Immunol, 2001. 86(6 Suppl 1): p. 24-30. 26. Bender, B.G. J Allergy Clin Immunol, 2002. 109(6 Suppl): p. S554-9. 27. Wills-Karp, M., et al. Science, 1998. 282(5397): p. 2258 61. 25 28. Grunig, G., et al. Science, 1998. 282(5397): p. 2261-3. 29. Venkayya, R., et al. Am J Respir Cell Mol Biol, 2002. 26(2): p. 202-8. 30. Morse, B., et al. Am J Physiol Lung Cell Mol Physiol, 2002. 282(1): p. L44-9. 30 31. Zhu, Z., et al. J Clin Invest, 1999. 103(6): p. 779-88. 32. Walter, D.M., et al. J Immunol, 2001. 167(8): p. 4668-75. 33. Cohn, L., J.S. Tepper, and K. Bottomly. J Immunol, 1998. 161(8): p. 3813-6. 34. Taube, C., et al. J Immunol, 2002. 169(11): p. 6482-9. 113 35. Yang, E.S., et al. J. Allergy Immunol., 2002. 109: p. A168. 36. Blease, K., et al. J Immunol, 2001. 166(8): p. 5219-24. 37. Heinzmann, A., et al. Hum Mol Genet, 2000. 9(4): p. 549 5 59. 38. Howard, T.D., et al. Am J Hum Genet, 2002. 70(1): p. 230 6. 39. Kauppi, P., et al. Genomics, 2001. 77(1-2): p. 35-42. 40. Graves, P.E., et al. J Allergy Clin Immunol, 2000. .0 105(3): p. 506-13. 41. Arima, K., et al. J Allergy Clin Immunol, 2002. 109(6): p. 980-7. 42. van der Pouw Kraan, T.C., et al. Genes Immun, 1999. 1(1): p. 61-5. .5 43. Humbert, M., et al. J Allergy Clin Immunol, 1997. 99(5): p. 657-65. 44. Kotsimbos, T.C., P. Ernst, and Q.A. Hamid, Proc Assoc Am Physicians, 1996. 108(5): p. 368-73. 45. Komai-Koma, M., F.Y. Liew, and P.C. Wilkinson, J Immunol, .0 1995. 155(3) : p. 1110-6. 46. Naseer, T., et al. Am J Respir Crit Care Med, 1997. 155(3): p. 845-51. 47. Huang, S.K., et al. J Immunol, 1995. 155(5): p. 2688-94. 48. Kroegel, C., et al. Eur Respir J, 1996. 9(5): p. 899-904. 25 49. Ohshima, Y., et al. Pediatr Res, 2002. 51(2): p. 195-200. 50. Hasegawa, M., et al. J Rheumatol, 1997. 24(2): p. 328-32. 51. Hancock, A., et al. Am J Respir Cell Mol Biol, 1998. 18(1): p. 60-5. 52. Lee, C.G., et al. J Exp Med, 2001. 194(6): p. 809-21. 30 53. Jain-Vora, S., et al. Am J Respir Cell Mol Biol, 1997. 17(5): p. 541-51. 54. Fallon, P.G., et al. J Immunol, 2000. 164(5): p. 2585-91. 55. Chiaramonte, M.G., et al. J Clin Invest, 1999. 104(6): p. 777-85. 114 56. Chiaramonte, M.G., et al. Hepatology, 2001. 34(2): p. 273-82. 57. Sluiter, H.J., et al. Eur Respir J, 1991. 4(4): p. 479 89. 5 58. Zheng, T., et al. J Clin Invest, 2000. 106(9): p. 1081 93. 59. Tashkin, D.P., et al., Methacholine reactivity predicts changes in lung function over time in smokers with early chronic obstructive pulmonary disease. The Lung Health 0 Study Research Group. Am J Respir Crit Care Med, 1996. 153(6 Pt 1): p. 1802-11. 60. Van Der Pouw Kraan, T.C., et al. Genes Immun, 2002. 3(7): p. 436-9. 61. Skinnider, B.F., et al. Blood, 2001. 97(1): p. 250-5. 5 62. Kapp, U., et al. J Exp Med, 1999. 189(12): p. 1939-46. 63. Fiumara, P., F. Cabanillas, and A. Younes, Blood, 2001. 98(9): p. 2877-8. 64. Terabe, M., et al. Nat Immunol, 2000. 1(6): p. 515-20. 65. Ahlers, J.D., et al. Proc Natl Acad Sci U S A, 2002. 0 99(20): p. 13020-5. 66. Hutchings, C., Generation of Naive Human Antibody Libraries, in Antibody Engineering, R. Kontermann and S. Dubel, Editors. 2001, Springer Laboratory Manuals, Berlin. p. 93-108. 25 67. Vaughan, T.J., et al. Nat Biotechnol, 1996. 14(3): p. 309-14. 68. Kitamura, T., et al. Blood, 1989. 73(2): p. 375-80. 69. Lefort, S., et al. FEBS Lett, 1995. 366(2-3): p. 122-6. 70. Osbourn, J.K., et al. Immunotechnology, 1996. 2(3): p. 30 181-96. 71. Howard, T.D., et al. Am J Respir Cell Mol Biol, 2001. 25(3): p. 377-84. 72. Karlsson, R., A. Michaelsson, and L. Mattsson, J Immunol Methods, 1991. 145(1-2): p. 229-40. 115 73. Tomlinson, VBASE. 1997, MRC Centre for Protein Engineering, Cambridge, UK. 74. Altmann, F., et al. Glycoconj J, 1999. 16(2): p. 109-23. 75. Drexler, H.G., et al. Leuk Res, 1986. 10(5): p. 487-500. 5 76. Skinnider, B.F., U. Kapp, and T.W. Mak, Leuk Lymphoma, 2002. 43(6): p. 1203-10. 77. Terada, N., et al. Clin Exp Allergy, 2000. 30(3): p. 348 55. 78. Wenzel, S.E., et al. J Immunol, 2002. 169(8): p. 4613-9. .0 79. Richter, A., et al. Am J Respir Cell Mol Biol, 2001. 25(3): p. 385-91. 80. Bochner, B.S., et al. J Immunol, 1995. 154(2): p. 799 803. 81. Kotowicz, K., et al. Int Immunol, 1996. 8(12): p. 1915 .5 25. 82. McKenzie, A.N., et al. Journal of Immunology, 1993. 150(12): p. 5436-44. 83. Bouteiller, C.L., et al. J Immunol Methods, 1995. 181(1): p. 29-36. 0 84. Riffo-Vasquez, Y., et al. Clin Exp Allergy, 2000. 30(5): p. 728-38. 85. McMillan, S.J., et al. J Exp Med, 2002. 195(1): p. 51-7. 86. Humbles, A.A., et al. Proc Natl Acad Sci U S A, 2002. 99(3): p. 1479-84. 25 87. Temelkovski, J., et al. Thorax, 1998. 53(10): p. 849-56. 88. Belvisi, M.G., et al., Pulm Pharmacol Ther, 2001. 14(3): p. 221-7. 30 89. Barnes, P.J., et al. Eur Respir J, 1996. 9(4): p. 636-42. 90. Barnes, P.J., Pharmacol Ther, 2003. 97(1): p. 87-94. 91. Wardlaw, A.J., Clin Med, 2001. 1(3): p. 214-8. 116 92. Edwards, J.C., et al. J Pathol, 1981. 134(2): p. 147-56. 93. McDonough, J.E., et al. W.M. Elliot, and J.C. Hogg. TGF 5 beta Isoform and IL-13 Immunostaining on Lung Tissue from Patients with COPD. in ATS 99th International Conference. 2003. Seattle. 94. Wold, et al. Multivariate data analysis in chemistry. .0 Chemometrics - Mathematics and Statistics in Chemistry (Ed.: B. Kowalski), D. Reidel Publishing Company, Dordrecht, Holland, 1984 (ISBN 90-277-1846-6). 95. Norman et al. Applied Regression Analysis. Wiley .5 Interscience; 3rd edition (April 1998) ISBN: 0471170828 96. Abraham Kandel, Eric Backer. Computer-Assisted Reasoning in Cluster Analysis. Prentice Hall PTR; (May 11, 1995), ISBN: 0133418847 .0 97. Wojtek Krzanowski. Principles of Multivariate Analysis: A User's Perspective (Oxford Statistical Science Series, No 22 (Paper)). Oxford University Press; (December 2000), ISBN: 0198507089 25 98. Ian H. Witten, Eibe Frank. Data Mining: Practical Machine Learning Tools and Techniques with Java Implementations. Morgan Kaufmann; (October 11, 1999), ISBN: 1558605525 30 99. David G. T. Denison (Editor), Christopher C. Holmes, Bani K. Mallick, Adrian F. M. Smith. Bayesian Methods for Nonlinear Classification and Regression (Wiley Series in Probability and Statistics). John Wiley & Sons; (July 2002), ISBN: 0471490369 117 100. Arup K. Ghose, Vellarkad N. Viswanadhan. Combinatorial Library Design and Evaluation Principles, Software, Tools, and Applications in Drug Discovery. ISBN: 0-8247 5 0487-8 101. Chothia C. et al. Journal Molecular Biology (1992) 227, 799-817. 0 102. Al-Lazikani, et al. Journal Molecular Biology (1997) 273(4), 927-948. 103. Chothia, et al. Science, 233,755-758 (1986). 5 104. Whitelegg, N.R.J. and Rees, A.R (2000). Prot. Eng., 13, 819-824. 105. Available from Accelerys Inc. 0 106. Guex, N. and Peitsch, M.C. (1997). Electrophoresis (1997) 18, 2714-2723. 107. Kabat E A et al (1991): Sequences of Proteins of Immunological Interest, 5'h Edition. US Department of 25 Health and Human Services, Public Service, NIH, Washington. 108. Kontermann R and Dubel Stefan; (2001) Antibody Engineering, Springer Laboratory Manuals. 30 109. Mendez et al (1997); Nature Genetics Vol. 2: 146-156. 110. Csonka E et al (2000) Journal of Cell Science, 113: 3207 3216. 118 111. Vanderbyl S et al (2002) Molecular Therapy, 5(5): 10. 112. Marasco WA (1997) Gene Therapy, 4(1): 11. 5 113. Hanes J et al (2000). Methods in Enzymology, Vol 328:24. 114. Li et al (2003). Abstract for poster [605] submitted at The American Thoracis Society Annual Meeting, 2003, L0 Seattle. 115. Koide et al (1998). Journal of Molecular Biology, Vol 284:1141-1151. .5 116. Nygren et al (1997). Current Opinion in Structural Biology, Vol 7:463-469. 117. Heller, F., et al. (2002) Immunity, 17(5):629-38. !0 118. Inoue, S., et al. (1999) Am J Gastroenterol, 94(9):2441 6. 119. Yang, M., et al. Am J Respir Cell Mol Biol. 2001. 25(4): p. 522-30 25 120. Punnonen J., et al 1993. Proc Natl Acad Sci. 90(8):3730 4. 121.Grunstein, M., et al. Am J Physiol Lung Cell Mol Physiol 30 2002. 282: p. L520-L528. 122. Laporte, J., et al. Am J Respir Crit Care Med 2001. 164: p. 141-148. 119 123. Tliba 0., et al. Br J Pharmacol 2003. 140(7): p. 1159 62. 124. Deshpande, D., et al. Am J Respir Cell Mol Biol 2004. 5 31(1): p. 36-42; Epub Feb 5 as doi:10.1165/rcmb.2003-03130C. 125. Humbert et al. 1997. J. Allergy Clin. Immunol., 99:657. 126. Berry, M.A., Parker, D., Neale, N., Woodman, L., Morgan, .0 A. Monk, P.D.. Submitted to J. Allergy Clin Immunol. 127. Obase et al. Ann Allergy Asthma Immunol. 2001; 86(3) :304-10. 5 128. Chu et al. 2000; J. Allergy Clin. Immunol. 106:1115 129. Terada et al. 2000. Clin. Exp. Allergy., 30: 348-55. 130. Wenzel et al. 2000. J. Immunol. 169: 4613-19. '0 120 14 14 ti C4 g ,q 2 0 2 0 0 0 0 a 00 FT-1 I 0 .00 44 - - - - - - I-H+H+fl I I I I 11 CD 0 v 0 tb 10 H H $4 H w N C. x E. N 0 1 1 ta n m F.ImI INI 10 TrT --- :,Pz 0 -424 . 10i .0 Goo: 0; - - - - - - + so r (n m 4 to M mi" Cal 0 wl 01 m w 0 vi m 0 02 0 4n Colo m In 0 w %a SO.,:: 'A 10 vi ;41 to 0 09 .4 0 #A z 14 - - - - - - - -- Tt 09 $4 -1 H r. 0 >IMI M t4 4 99 A I I I pq 1 11-0 C3 H vi eq 1.414 A 94 M 14 910 M H 4 XI 1 .4 VZO 0 IcIm ro a, al >. A. 0 1 91 z 09 is 9c 14 PC .4 64 [4 64 14 Z c Ion n 0 4 C06 r,992IR 02 d? RJR R COLROO- coijolcoo 0! x we S 0 Ct q Irm R R FH-F to to 10 to it 08 > 69 +a 02 Sir. a. :T-FT -- to Q 09 a[ VC ic > --------- rc. .3 tc ---------
----------
cc 04( a 09 LE -TT.T T -GQ F-T m(UH zolt a ---- - ----------- W& Q - - - - -- - - - - - - - - - 90 ot D. CM P. as - - - -- - - -- - - - -- - - - - - soot 0: got oot F -T-T-TT-1-all, .+-+I+. Ll g: T7-7T 69 0 T TM to to ON 99 0 1. 1.411 - - - - - - 09 al 09 p Is z 49 p 02 IQ so 0 V9 m I I F- I J= cloc" CS 0 --- I I F TTF -41 U191010 zq 4 1141 1 1 ANaOa to m I I I I F F " -A 1 -1 RTFTT te .3 -- &C z I I F RRR@IEIR&-RIR@R-MI@IRRRI@PRRR M- 2 o 2 21'' Table 2 Binding specificity of anti-human IL-13 antibodies Human IL-13 Human IL-13 Non-human variant primate IL-13 BAK278D6 BAK502G9 + + + BAK615E3 + 5 Table 3a Kinetic analysis of anti-human IL-13 antibodies IgG Off-rate On-rate KD (s1) (M~ s ) (pM) BAK278D6 7.41e- 3 5.49e 5 13500 BAK502G9 4.09e 4 2.49e 6 178 BAK1167F2 4.05e~ 4 2.99e 6 136 BAK1183H4 3.00e~ 4 3.7e 6 81 0 Table 3b Kinetic analysis of anti-murine IL-13 antibodies IgG Off-rate (s~') On-rate KD (M~1 s~ ) (pM) BAK209B11 1.98e 2 3.9e" 5100 15 123 Table 4 Pharmacokinetics of BAK502G9 in 4 allergic but non-challenged cynomolgus primates (2 male/2 female) after a single 10mg/kg 5 i.v bolus dose over 29 days. BAK502G9 levels in serum were measured by ELISA (mean data). Cmax 349.04 (t=0.25h) (ptg/mL) Vdinf 75.03 <80mL/kg, infers no (mL.kg~ ) tissue binding. Clinf 0.23 (mL. hr .kg- 1 ) AUCinf 42.99 (mg.h.mL 1 ) AUCext 17.34 <30% so clearance and (%) vol. of distribution should be accurate. To.s 223.55 (h) Vdinf = volume of distribution over time 0 - infinity, 10 calculated from the extrapolated AUC. Cling = clearance over time 0 - infinity, calculated from the extrapolated AUC. AUCiaf = area under the curve (measure of total drug exposure) over time 0 - infinity, including an extrapolated term based 15 on the elimination rate constant (k) and the last observed serum drug concentration. AUCet = percentage of the total AUC that is extrapolated. To5 = Drug half-life in the terminal elimination phase. 20 124 Table 5 First set of Chimeric constructs Chimeric constructs IC50 nM BAK502G9 0.17 ± 0.07 loopi 0.71 ± 0.35 hum-flag 1.30 ± 0.18 30R 1.76 ± 0.45 3738VN 1.89 1.9 helixB 2.49 0.88 helixC 4.11 0.70 loop3 5.45 3.96 4041 12.02 ± 1.3 3334 12.17 ± 1.2 helixD 110.07 ± 9.9 5 Table 6 Second Set of Chimeric Constructs Chimeric Constructs IC50 nM BAK502G9 0.11± 0.04 113H 1.6 ± 0.5 128H 1.6± 1.0 119LA 1.96 ± 1.0 130P 2.22 ± 0.8 120121SY 4.73 ± 1.5 58LA 5.2 ± 2.0 124Q 18.7 ± 15.9 116117T 82 ±11.3 123KA none 127 RA none 125 Table 7: Effects of BAK502G9 on various predefined endpoints. Phase I Phase II Parameter N N Endpoint change change AHR (RL AUC) 0.020±0.003 14a 0.004±0.006 14a -0.016±0.006* AHR (PC 3 0 ) -1.343±0.318 1 8 b 1.061±0.244 1 8 b 0.282±0.179 Antigen priming 0.159±0.033 20c 0.033±0.025 20c 0.126t0.043** (RL AUC) ___ ___________ BAL total cells 20.623±3.160 2 1 d 14.597±1.95 2 1 d -6.026±2.194* BAL eosinophils 18.453±3.009 21 d 13.412±1.73 2 1 d -5.041±2.090* BAL mononuclear 2.050±0.438 2 1 d 1.176±0.481 2 1 d -0.874±0.506 cells______ __ 5 21 animals displaying AHR (PC 30 ) in Phase I and an additional animal with an antigen priming phenotype were carried forward for testing in Phase II (22 in total). Not every animal had AHR as measured by both AUC and PC 30 . Only animals which LO displayed AHR in phase I and whose AHR was assessed in both Phase I and Phase II were included in the AHR results. Statistical testing was performed using InStat. Testing was a 2-way student's t-test against the null hypothesis that the endpoint did include the number 0 (i.e. there was no change in .5 phase II compared to phase I); *p<0.05, **p<0.01. Data are shown as arithmetic mean ± SEM (n=14-21). '5 animals were excluded from the AUC analysis as they did not display AHR (increased AUC) in Phase I. 3 further animals were excluded due to a technical failure in 20 Phase II airway function data collection. b 3 animals were excluded from PC 3 0 analysis due to a technical failure in Phase II airway function data collection (same animals as in a). The additional animal with antigen priming phenotype was excluded as it did not display PC 3 o AHR in Phase 25 I. 126 c 2 animals were excluded from the antigen priming analysis as there was a technical failure in Phase I airway function data collection. 5 d 1 animal was excluded from the BAL analysis due to marked BAL inflammation at study initiation. Definition Throughout this specification the word "comprise", or 0 variations such as "comprises" or "comprising", will be understood to imply the inclusion of a stated element, integer or step, or group of elements, integers or steps, but not the exclusion of any other element, integer or step, or group of elements, integers or steps. 5 Prior art references Any discussion of documents, acts, materials, devices, articles or the like which has been included in the present specification is solely for the purpose of providing a context D for the present invention. It is not to be taken as an admission that any or all of these matters form part of the prior art base or were common general knowledge in the field relevant to the present invention as it existed before the priority date of each claim of this application. 127 Antibody domain sequences BAK278D6 HEAVY CHAIN CDR1-SEQ ID NO 1: NYGLS CDR2-SEQ ID NO 2: WISANNGDTNYGQEFQG CDR3-SEQ ID NO 3: DSSSNWARWFFDL BAK278D6 LIGHT CHAIN CDR1-SEQ ID NO 4: GGNNIGSKLVH CDR2-SEQ ID NO 5: DDGDRPS CDR3-SEQ ID NO 6: QVWDTGSDPVV BAK502G9 HEAVY CHAIN CDR1-SEQ ID NO 7: NYGLS CDR2-SEQ ID NO 8: WISANNGDTNYGQEFQG CDR3-SEQ ID NO 9: DSSSSWARWFFDL LIGHT CHAIN CDR1-SEQ ID NO 10: GGNIIGSKLVH CDR2-SEQ ID NO 11: DDGDRPS CDR3-SEQ ID NO 12: QVWDTGSDPVV BAK278D6 HEAVY CHAIN DOMAIN SEQ ID NO 13: EVQLVQSGAEVKKPGASVKVSCKASGYTFRNYGLSWVRQAPGQGLEWMGWISANNGDTN YGQEFQGRITMTTETSTNTAHMELRSLRSDDTAVYYCVRDSSSNWARWFFDLWGKGTMV TVSS BAK278D6 LIGHT CHAIN DOMAIN SEQ ID NO 14: SYVLTQPPSVSVAPGQTARIPCGGNNIGSKLVHWYQQKPGQAPVLVVYDDGDRPSGIPE RFSGSNSGNTATLTISRIDAGDEADYYCQVWDTGSDPVVFGGGTKLTVL 128 BAK502G9 HEAVY CHAIN DOMAIN SEQ ID NO 15: QVQLVQSGAEVKKPGASVKVSCKASGYTFTNYGLSWVRQAPGQGLEWMGWISANNGDTN YGQEFQGRVTMTTDTSTSTAYMELRSLRSDDTAVYYCARDSSSSWARWFFDLWGRGTLV TVSS BAK502G9 LIGHT CHAIN DOMAIN SEQ ID NO 16: SYVLTQPPSVSVAPGKTARITCGGNIIGSKLVHWYQQKPGQAPVLVIYDDGDRPSGIPE RFSGSNSGNTATLTISRVEAGDEADYYCQVWDTGSDPVVFGGGTKLTVL BAK278D6 HEAVY CHAIN FR1-SEQ ID NO 17: EVQLVQSGAEVKKPGASVKVSCKASGYTFR FR2-SEQ ID NO 18: WVRQAPGQGLEWMG FR3-SEQ ID NO 19: RITMTTETSTNTAHMELRSLRSDDTAVYYCVR BAK278D6 LIGHT CHAIN FR1-SEQ ID NO 20: SYVLTQPPSVSVAPGQTARIPC FR2-SEQ ID NO 21: WYQQKPGQAPVLVVY FR3-SEQ ID NO 22: GIPERFSGSNSGNTATLTISRIDAGDEADYYC BAK167A1 HEAVY CHAIN DOMAIN SEQ ID NO 23: EVQLLESGGGLVQPGGSLRLSCAASGFTFSSYAMSWVRQAPGKGLEWVSAISGSGGSTY YADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARVGAAGEGYYGYWGRGTLVTV SS BAK167A11 LIGHT CHAIN DOMAIN SEQ ID NO 24: NFMLTQPHSVSESPGKTVTISCTRSSGSIASNYVQWYQQRPGSAPTTVIYDDNQRPSGV PDRFSGSIDSSSNSASLTISGLKTEDEADYYCQSYDSNNDVFGGGTKVTVL 129 BAK209B11 HEAVY CHAIN DOMAIN SEQ ID NO 25: QVQLQESGGGGLVQPGGSLRLSCAASGFTFSSYGMSWVRQAPGKGLEWVSSISASGDST FYADSVKGRFTISRDNNKNMVFLQVNSLRADDTAVYFCAKDWSQWLVGDAFDVWGRGTT VTVSS BAK209B1l LIGHT CHAIN DOMAIN SEQ ID NO 26: DIQLTQSPSTLSASVGDRVTITCRASQSVSLWVAWYQQRPGKAPKLLIYDGSTLQSGVP ARFSGSGSGTEFTLTISSLQPDDFATYYCQQYKTFSTFGQGTKVEIKRA BAK502G9 HEAVY CHAIN FR1-SEQ ID NO 27: QVQLVQSGAEVKKPGASVKVSCKASGYTFT FR2-SEQ ID NO 28: WYRQAPGQGLEWMG FR3-SEQ ID NO 29: RVTMTTDTSTSTAYMELRSLRSDDTAVYYCAR BAK502G9 LIGHT CHAIN FR1-SEQ ID NO 30: SYVLTQPPSVSVAPGKTARITC FR2-SEQ ID NO 31: WYQQKPGQAPVLVIY FR3-SEQ ID NO 32: GIPERFSGSNSGNTATLTISRVEAGDEADYYC BAK615E3 HEAVY CHAIN DOMAIN SEQ ID NO 33: EVQLLESGGGLVQPGGSLRLSCAASGFTFSSYAMSWVRQAPGKGLEWVSAISGSGGSTY YADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARVGKATTEEGYYGYWGRGTLV TVSS BAK615E3 LIGHT CHAIN DOMAIN SEQ ID NO 34: NFMLTQPHSVSESPGKTVTISCTRSSGSIASNYVQWYQQRPGSAPTTVIYDDNQRPSGV PDRFSGSIDSSSNSASLTISGLKTEDEADYYCQSYDSNNDVFGGGTKVTVL 130 BAKll67F2 HEAVY CHAIN DOMAIN SEQ ID NO 35: QVQLVQSGAEVKKPGASVKVSCKASGYTFEQTGVSWVRQAPGQGLEWMGWISANNGDTN YGQEFQGRVTMTTDTSTSTAYMELRSLRSDDTAVYYCARDSSSSWARWFFDLWGRGTLV TVSS BAKl167F2 LIGHT CHAIN DOMAIN SEQ ID NO 36: SYVLTQPPSVSVAPGKTARITCGGNIIGSKLVHWYQQKPGQAPVLVIYDDGDRPSGIPE RFSGSNSGNTATLTISRVEAGDEADYYCQVWDTGSDPVVFGGGTKLTVL BAK1183H4 HEAVY CHAIN DOMAIN SEQ ID NO 37: QVQLVQSGAEVKKPGASVKVSCKASGYTFTNYGLSWVRQAPGQGLEWMGWINYDGGNTQ YGQEFQGRVTMTTDTSTSTAYMELRSLRSDDTAVYYCARDSSSSWARWFFDLWGRGTLV TVSS BAK1183H4 LIGHT CHAIN DOMAIN SEQ ID NO 38: SYVLTQPPSVSVAPGKTARITCGGNIIGSKLVHWYQQKPGQAPVLVIYDDGDRPSGIPE RFSGSNSGNTATLTISRVEAGDEADYYCQVWDTGSDPVVFGGGTKLTVL BAK1105H3 HEAVY CHAIN DOMAIN SEQ ID NO 39: QVQLVQSGAEVKKPGASVKVSCKASGYTFTNYGLSWVRQAPGQGLEWMGWISGLNGETL YGQEFQGRVTMTTDTSTSTAYMELRSLRSDDTAVYYCARDSSSSWARWFFDLWGRGTLV TVSS 131 BAK1105H3 LIGHT CHAIN DOMAIN SEQ ID NO 40: SYVLTQPPSVSVAPGKTARITCGGNIIGSKLVHWYQQKPGQAPVLVIYDDGDRPSGIPE RFSGSNSGNTATLTISRVEAGDEADYYCQVWDTGSDPVVFGGGTKLTVL BAK111lD10 HEAVY CHAIN DOMAIN SEQ ID NO 41: QVQLVQSGAEVKKPGASVKVSCKASGYTFTNYGLSWVRQAPGQGLEWMGWIATPDGQTS YGQEFQGRVTMTTDTSTSTAYMELRSLRSDDTAVYYCARDSNSSWARWFFDLWGRGTLV TVSS BAKllllD10 LIGHT CHAIN DOMAIN SEQ ID NO 42: SYVLTQPPSVSVAPGKTARITCGGNIIGSKLVHWYQQKPGQAPVLVIYDDGDRPSGIPE RFSGSNSGNTATLTISRVEAGDEADYYCQVWDTGSDPVVFGGGTKLTVL BAK1167F4 HEAVY CHAIN DOMAIN SEQ ID NO 43: QVQLVQSGAEVKKPGASVKVSCKASGYTFIDTGVSWVRQAPGQGLEWMGWISANNGDTN YGQEFQGRVTMTTDTSTSTAYMELRSLRSDDTAVYYCARDSSSSWARWFFDLWGRGTLV TVSS BAK1167F4 LIGHT CHAIN DOMAIN SEQ ID NO 44: SYVLTQPPSVSVAPGKTARITCGGNIIGSKLVHWYQQKPGQAPVLVIYDDGDRPSGIPE RFSGSNSGNTATLTISRVEAGDEADYYCQVWDTGSDPVVFGGGTKLTVL BAK1184C8 HEAVY CHAIN DOMAIN SEQ ID NO 45: QVQLVQSGAEVKKPGASVKVSCKASGYTFTNYGLSWVRQAPGQGLEWMGWISGSNGYTS YGQEFQGRVTMTTDTSTSTAYMELRSLRSDDTAVYYCARDSSSSWARWFFDLWGRGTLV TVSS 132 BAK1184C8 LIGHT CHAIN DOMAIN SEQ ID NO 46: SYVLTQPPSVSVAPGKTARITCGGNIIGSKLVHWYQQKPGQAPVLVIYDDGDRPSGIPE RFSGSNSGNTATLTISRVEAGDEADYYCQVWDTGSDPVVFGGGTKLTVL BAK1185E1 HEAVY CHAIN DOMAIN SEQ ID NO 47: QVQLVQSGAEVKKPGASVKVSCKASGYTFTNYGLSWVRQAPGQGLEWMGWINDATGDTQ YGQEFQGRVTMTTDTSTSTAYMELRSLRSDDTAVYYCARDSSSSWARWFFDLWGRGTLV TVSS BAK1185E1 LIGHT CHAIN DOMAIN SEQ ID NO 48: SYVLTQPPSVSVAPGKTARITCGGNIIGSKLVHWYQQKPGQAPVLVIYDDGDRPSGIPE RFSGSNSGNTATLTISRVEAGDEADYYCQVWDTGSDPVVFGGGTKLTVL BAK1185F8 HEAVY CHAIN DOMAIN SEQ ID NO 49: QVQLVQSGAEVKKPGASVKVSCKASGYTFTDYGLSWVRQAPGQGLEWMGWIRNIDGYTI YGQEFQGRVTMTTDTSTSTAYMELRSLRSDDTAVYYCARDSSSSWARWFFDLWGRGTLV TVSS BAKll85F8 LIGHT CHAIN DOMAIN SEQ ID NO 50: SYVLTQPPSVSVAPGKTARITCGGNIIGSKLVHWYQQKPGQAPVLVIYDDGDRPSGIPE RFSGSNSGNTATLTISRVEAGDEADYYCQVWDTGSDPVVFGGGTKLTVL BAKll87B4 HEAVY CHAIN DOMAIN SEQ ID NO 51: QVQLVQSGAEVKKPGASVKVSCKASGYTFTNYGLSWVRQAPGQGLEWMGWIDDDSGTTI YGQEFQGRVTMTTDTSTSTAYMELRSLRSDDTAVYYCARDSSSSWARWFFDLWGRGTLV TVSS 133 BAK1187B4 LIGHT CHAIN DOMAIN SEQ ID NO 52: SYVLTQPPSVSVAPGKTARITCGGNIIGSKLVHWYQQKPGQAPVLVIYDDGDRPSGIPE RFSGSNSGNTATLTISRVEAGDEADYYCQVWDTGSDPVVFGGGTKLTVL BAK1166G2 HEAVY CHAIN DOMAIN SEQ ID NO 53: QVQLVQSGAEVKKPGASVKVSCKASGYTFANTGISWVRQAPGQGLEWMGWISANNGDTN YGQEFQGRVTMTTDTSTSTAYMELRSLRSDDTAVYYCARDSSSSWARWFFDLWGRGTLV TVSS BAK1166G2 LIGHT CHAIN DOMAIN SEQ ID NO 54: SYVLTQPPSVSVAPGKTARITCGGNIIGSKLVHWYQQKPGQAPVLVIYDDGDRPSGIPE RFSGSNSGNTATLTISRVEAGDEADYYCQVWDTGSDPVVFGGGTKLTVL BAK167A11 HEAVY CHAIN CDR1-SEQ ID NO 55: SYAMS CDR2-SEQ ID NO 56: AISGSGGSTYYADSVKG CDR3-SEQ ID NO 57: VGAAGEGYYGY BAK167A11 LIGHT CHAIN CDR1-SEQ ID NO 58: TRSSGSIASNYVQ CDR2-SEQ ID NO 59: DDNQRPS CDR3-SEQ ID NO 60: QSYDSNNDV BAK1167F2 HEAVY CHAIN CDR1-SEQ ID NO 61: QTGVS CDR2-SEQ ID NO 62: WISANNGDTNYGQEFQG CDR3-SEQ ID NO 63: DSSSSWARWFFDL 134 BAK1167F2 LIGHT CHAIN CDR1-SEQ ID NO 64: GGNIIGSKLVH CDR2-SEQ ID NO 65: DDGDRPS CDR3-SEQ ID NO 66: QVWDTGSDPVV BAK1166G2 HEAVY CHAIN CDR1-SEQ ID NO 67: NTGIS CDR2-SEQ ID NO 68: WISANNGDTNYGQEFQG CDR3-SEQ ID NO 69: DSSSSWARWFFDL BAK1166G2 LIGHT CHAIN CDR1-SEQ ID NO 70: GGNIIGSKLVH CDR2-SEQ ID NO 71: DDGDRPS CDR3-SEQ ID NO 72: QVWDTGSDPVV BAK1184C8 HEAVY CHAIN CDR1-SEQ ID NO 73: NYGLS CDR2-SEQ ID NO 74: WISGSNGYTSYGQEFQG CDR3-SEQ ID NO 75: DSSSSWARWFFDL BAK1184C8 LIGHT CHAIN CDR1-SEQ ID NO 76: GGNIIGSKLVH CDR2-SEQ ID NO 77: DDGDRPS CDR3-SEQ ID NO 78: QVWDTGSDPVV BAK1185E1 HEAVY CHAIN CDR1-SEQ ID NO 79: NYGLS CDR2-SEQ ID NO 80: WINDATGDTQYGQEFQG CDR3-SEQ ID NO 81: DSSSSWARWFFDL 135 BAK1185E1 LIGHT CHAIN CDR1-SEQ ID NO 82: GGNIIGSKLVH CDR2-SEQ ID NO 83: DDGDRPS CDR3-SEQ ID NO 84: QVWDTGSDPVV BAK1167F4 HEAVY CHAIN CDR1-SEQ ID NO 85: DTGVS CDR2-SEQ ID NO 86: WISANNGDTNYGQEFQG CDR3-SEQ ID NO 87: DSSSSWARWFFDL BAK1167F4 LIGHT CHAIN CDR1-SEQ ID NO 88: GGNIIGSKLVH CDR2-SEQ ID NO 89: DDGDRPS CDR3-SEQ ID NO 90: QVWDTGSDPVV BAKllllD10 HEAVY CHAIN CDR1-SEQ ID NO 91: NYGLS CDR2-SEQ ID NO 92: WIATPDGQTSYGQEFQG CDR3-SEQ ID NO 93: DSNSSWARWFFDL BAKllllD10 LIGHT CHAIN CDR1-SEQ ID NO 94: GGNIIGSKLVH CDR2-SEQ ID NO 95: DDGDRPS CDR3-SEQ ID NO 96: QVWDTGSDPVV BAK1183H4 HEAVY CHAIN CDR1-SEQ ID NO 97: NYGLS CDR2-SEQ ID NO 98: WINYDGGNTQYGQEFQG CDR3-SEQ ID NO 99: DSSSSWARWFFDL 136 BAK1183H4 LIGHT CHAIN CDR1-SEQ ID NO 100: GGNIIGSKLVH CDR2-SEQ ID NO 101: DDGDRPS CDR3-SEQ ID NO 102: QVWDTGSDPVV BAK1185H8 HEAVY CHAIN CDR1-SEQ ID NO 103: DYGLS CDR2-SEQ ID NO 104: WRINDGYTIYGQEFQG CDR3-SEQ ID NO 105: DSSSSWARWFFDL BAK1185H8 LIGHT CHAIN CDR1-SEQ ID NO 106: GGNIIGSKLVH CDR2-SEQ ID NO 107: DDGDRPS CDR3-SEQ ID NO 108: QVWDTGSDPVV BAK278D6 HEAVY CHAIN-SEQ ID NO: 109 CAGGTGCAGCTGGTGCAGTCTGGGGCTGAGGTGAAGAAGCCTGGGGCCTCAGTGAAGGT CTCCTGCAAGGCTTCTGGTTACACCTTTACAAATTATGGTCTCAGCTGGGTGCGACAGG CCCCTGGACAAGGGCTTGAGTGGATGGGATGGATCAGCGCTAATAATGGCGACACAAAT TATGGACAGGAATTCCAGGGCAGAGTCACCATGACCACAGATACATCCACGAGCACAGC CTACATGGAGTTGAGGAGCCTGAGATCTGACGACACGGCCGTTTATTACTGTGCGAGAG ACTCCAGCAGCAACTGGGCCCGCTGGTTTTTCGATCTCTGGGGCCGGGGGACACTGGTC ACCGTCTCCTCA BAK278D6 LIGHT CHAIN-SEQ ID NO: 110 TCCTATGTGCTGACTCAGCCACCCTCGGTGTCAGTGGCCCCAGGTAAGACGGCCAGGAT TACCTGTGGGGGAAACAACATTGGAAGTAAACTTGTACACTGGTACCAGCAGAAGCCAG GCCAGGCCCCTGTGCTGGTCATCTATGATGATGGCGACCGGCCCTCAGGGATCCCTGAG CGATTCTCTGGCTCCAACTCTGGGAACACGGCCACCCTGACCATCAGCAGGGTCGAGGC CGGGGATGAGGCCGACTATTATTGTCAGGTGTGGGATACTGGTAGTGATCCCGTGGTAT TCGGCGGAGGGACCAAGCTGACCGTCCTAGGT 137 BAK502G9 HEAVY CHAIN-SEQ ID NO: 111 CAGGTCCAGCTGGTGCAGTCTGGGGCTGAGGTGAAGAAGCCTGGGGCCTCAGTGAAGGT CTCCTGCAAGGCTTCTGGTTACACCTTTACAAATTATGGTCTCAGCTGGGTGCGACAGG CCCCTGGACAAGGGCTTGAGTGGATGGGATGGATCAGCGCTAATAATGGCGACACAAAT TATGGACAGGAATTCCAGGGCAGAGTCACCATGACCACAGATACATCCACGAGCACAGC CTACATGGAGTTGAGGAGCCTGAGATCTGACGACACGGCCGTTTATTACTGTGCGAGAG ACTCCAGCAGCAGCTGGGCCCGCTGGTTTTTCGATCTCTGGGGCCGGGGGACACTGGTC ACCGTCTCCTCA BAK502G9 LIGHT CHAIN-SEQ ID NO: 112 TCCTATGTGCTGACTCAGCCACCCTCGGTGTCAGTGGCCCCAGGAAAGACGGCCAGGAT TACCTGTGGGGGAAACATCATTGGAAGTAAACTTGTACACTGGTACCAGCAGAAGCCAG GCCAGGCCCCTGTGCTGGTCATCTATGATGATGGCGACCGGCCCTCAGGGATCCCTGAG CGATTCTCTGGCTCCAACTCTGGGAACACGGCCACCCTGACCATCAGCAGGGTCGAGGC CGGGGATGAGGCCGACTATTATTGTCAGGTGTGGGATACTGGTAGTGATCCCGTGGTAT TCGGCGGAGGGACCAAGCTGACCGTCCTAGGT BAK1105HO3 HEAVY CHAIN-SEQ ID NO: 113 CAGGTCCAGCTGGTGCAGTCTGGGGCTGAGGTGAAGAAGCCTGGGGCCTCAGTGAAGGT CTCCTGCAAGGCTTCTGGTTACACCTTTACAAATTATGGTCTCAGCTGGGTGCGACAGG CCCCTGGACAAGGGCTTGAGTGGATGGGATGGATCTCCGGCTTGAACGGCGAGACATTG TATGGACAGGAATTCCAGGGCAGAGTCACCATGACCACAGATACATCCACGAGCACAGC CTACATGGAGTTGAGGAGCCTGAGATCTGACGACACGGCCGTTTATTACTGTGCGAGAG ACTCCAGCAGCAGCTGGGCCCGCTGGTTTTTCGATCTCTGGGGCCGGGGGACACTGGTC ACCGTCTCCTCA BAK1105H03 LIGHT CHAIN-SEQ ID NO: 114 TCCTATGTGCTGACTCAGCCACCCTCGGTGTCAGTGGCCCCAGGAAAGACGGCCAGGAT TACCTGTGGGGGAAACATCATTGGAAGTAAACTTGTACACTGGTACCAGCAGAAGCCAG GCCAGGCCCCTGTGCTGGTCATCTATGATGATGGCGACCGGCCCTCAGGGATCCCTGAG CGATTCTCTGGCTCCAACTCTGGGAACACGGCCACCCTGACCATCAGCAGGGTCGAGGC CGGGGATGAGGCCGACTATTATTGTCAGGTGTGGGATACTGGTAGTGATCCCGTGGTAT TCGGCGGAGGGACCAAGCTGACCGTCCTAGGT 138 BAKllllD10 HEAVY CHAIN-SEQ ID NO: 115 CAGGTGCAGCTGGTGCAGTCTGGGGCTGAGGTGAAGAAGCCTGGGGCCTCAGTGAAGGT CTCCTGCAAGGCTTCTGGTTACACCTTTACAAATTATGGTCTCAGCTGGGTGCGACAGG CCCCTGGACAAGGGCTTGAGTGGATGGGATGGATCGCAACCCCAGACGGCCAGACAAGC TATGGACAGGAATTCCAGGGCAGAGTCACCATGACCACAGATACATCCACGAGCACAGC CTACATGGAGTTGAGGAGCCTGAGATCTGACGACACGGCCGTTTATTACTGTGCGAGAG ACTCCAACAGCAGCTGGGCCCGCTGGTTTTTCGATCTCTGGGGCCGGGGGACACTGGTC ACCGTCTCCTCA BAKllllD10 LIGHT CHAIN-SEQ ID NO: 116 TCCTATGTGCTGACTCAGCCACCCTCGGTGTCAGTGGCCCCAGGAAAGACGGCCAGGAT TACCTGTGGGGGAAACATCATTGGAAGTAAACTTGTACACTGGTACCAGCAGAAGCCAG GCCAGGCCCCTGTGCTGGTCATCTATGATGATGGCGACCGGCCCTCAGGGATCCCTGAG CGATTCTCTGGCTCCAACTCTGGGAACACGGCCACCCTGACCATCAGCAGGGTCGAGGC CGGGGATGAGGCCGACTATTATTGTCAGGTGTGGGATACTGGTAGTGATCCCGTGGTAT TCGGCGGAGGGACCAAGCTGACCGTCCTAGGT BAK 1167F2 HEAVY CHAIN-SEQ ID NO: 117 CAAGTGCAGCTGGTGCAGTCTGGGGCTGAGGTGAAGAAGCCTGGGGCCTCAGTGAAGGT CTCCTGCAAGGCTTCTGGTTACACCTTTGAGCAGACCGGCGTCTCCTGGGTGCGACAGG CCCCTGGACAAGGGCTTGAGTGGATGGGATGGATCAGCGCTAATAATGGCGACACAAAT TATGGACAGGAATTCCAGGGCAGAGTCACCATGACCACAGATACATCCACGAGCACAGC CTACATGGAGTTGAGGAGCCTGAGATCTGACGACACGGCCGTTTATTACTGTGCGAGAG ACTCCAGCAGCAGCTGGGCCCGCTGGTTTTTCGATCTCTGGGGCCGGGGGACACTGGTC ACCGTCTCCTCA BAK 1167F2 LIGHT CHAIN-SEQ ID NO: 118 TCCTATGTGCTGACTCAGCCACCCTCGGTGTCAGTGGCCCCAGGAAAGACGGCCAGGAT TACCTGTGGGGGAAACATCATTGGAAGTAAACTTGTACACTGGTACCAGCAGAAGCCAG GCCAGGCCCCTGTGCTGGTCATCTATGATGATGGCGACCGGCCCTCAGGGATCCCTGAG CGATTCTCTGGCTCCAACTCTGGGAACACGGCCACCCTGACCATCAGCAGGGTCGAGGC CGGGGATGAGGCCGACTATTATTGTCAGGTGTGGGATACTGGTAGTGATCCCGTGGTAT TCGGCGGAGGGACCAAGCTGACCGTCCTAGGT 139 BAK 1167F04 HEAVY CHAIN-SEQ ID NO: 119 CAGGTGCAGCTGGTGCAGTCTGGGGCTGAGGTGAAGAAGCCTGGGGCCTCAGTGAAGGT CTCCTGCAAGGCTTCTGGTTACACCTTTATCGACACCGGGGTCTCCTGGGTGCGACAGG CCCCTGGACAAGGGCTTGAGTGGATGGGATGGATCAGCGCTAATAATGGCGACACAAAT TATGGACAGGAATTCCAGGGCAGAGTCACCATGACCACAGATACATCCACGAGCACAGC CTACATGGAGTTGAGGAGCCTGAGATCTGACGACACGGCCGTTTATTACTGTGCGAGAG ACTCCAGCAGCAGCTGGGCCCGCTGGTTTTTCGATCTCTGGGGCCGGGGGACACTGGTC ACCGTCTCCTCA BAK 1167F04 LIGHT CHAIN-SEQ ID NO: 120 TCCTATGTGCTGACTCAGCCACCCTCGGTGTCAGTGGCCCCAGGAAAGACGGCCAGGAT TACCTGTGGGGGAAACATCATTGGAAGTAAACTTGTACACTGGTACCAGCAGAAGCCAG GCCAGGCCCCTGTGCTGGTCATCTATGATGATGGCGACCGGCCCTCAGGGATCCCTGAG CGATTCTCTGGCTCCAACTCTGGGAACACGGCCACCCTGACCATCAGCAGGGTCGAGGC CGGGGATGAGGCCGACTATTATTGTCAGGTGTGGGATACTGGTAGTGATCCCGTGGTAT TCGGCGGAGGGACCAAGCTGACCGTCCTAGGT BAK 1183H4 HEAVY CHAIN-SEQ ID NO: 121 CAGGTGCAGCTGGTGCAGTCTGGGGCTGAGGTGAAGAAGCCTGGGGCCTCAGTGAAGGT CTCCTGCAAGGCTTCTGGTTACACCTTTACAAATTATGGTCTCAGCTGGGTGCGACAGG CCCCTGGACAAGGGCTTGAGTGGATGGGATGGATCAACTACGACGGCGGCAACACACAG TATGGACAGGAATTCCAGGGCAGAGTCACCATGACCACAGATACATCCACGAGCACAGC CTACATGGAGTTGAGGAGCCTGAGATCTGACGACACGGCCGTTTATTACTGTGCGAGAG ACTCCAGCAGCAGCTGGGCCCGCTGGTTTTTCGATCTCTGGGGCCGGGGGACACTGGTC ACCGTCTCCTCA BAK 1183H4 LIGHT CHAIN-SEQ ID NO: 122 TCCTATGTGCTGACTCAGCCACCCTCGGTGTCAGTGGCCCCAGGAAAGACGGCCAGGAT TACCTGTGGGGGAAACATCATTGGAAGTAAACTTGTACACTGGTACCAGCAGAAGCCAG GCCAGGCCCCTGTGCTGGTCATCTATGATGATGGCGACCGGCCCTCAGGGATCCCTGAG CGATTCTCTGGCTCCAACTCTGGGAACACGGCCACCCTGACCATCAGCAGGGTCGAGGC CGGGGATGAGGCCGACTATTATTGTCAGGTGTGGGATACTGGTAGTGATCCCGTGGTAT TCGGCGGAGGGACCAAGCTGACCGTCCTAGGT 140 BAK1184C8 HEAVY CHAIN-SEQ ID NO: 123 CAGGTGCAGCTGGTGCAGTCTGGGGCTGAGGTGAAGAAGCCTGGGGCCTCAGTGAAGGT CTCCTGCAAGGCTTCTGGTTACACCTTTACAAATTATGGTCTCAGCTGGGTGCGACAGG CCCCTGGACAAGGGCTTGAGTGGATGGGATGGATCAGCGGGAGCAACGGCTACACATCT TATGGACAGGAATTCCAGGGCAGAGTCACCATGACCACAGATACGTCCACGAGCACAGC CTACATGGAGTTGAGGAGCCTGAGATCTGACGACACGGCCGTTTATTACTGTGCGAGAG ACTCCAGCAGCAGCTGGGCCCGCTGGTTTTTCGATCTCTGGGGCCGGGGGACACTGGTC ACCGTCTCCTCA BAK1184C8 LIGHT CHAIN-SEQ ID NO: 124 TCCTATGTGCTGACTCAGCCACCCTCGGTGTCAGTGGCCCCAGGAAAGACGGCCAGGAT TACCTGTGGGGGAAACATCATTGGAAGTAAACTTGTACACTGGTACCAGCAGAAGCCAG GCCAGGCCCCTGTGCTGGTCATCTATGATGATGGCGACCGGCCCTCAGGGATCCCTGAG CGATTCTCTGGCTCCAACTCTGGGAACACGGCCACCCTGACCATCAGCAGGGTCGAGGC CGGGGATGAGGCCGACTATTATTGTCAGGTGTGGGATACTGGTAGTGATCCCGTGGTAT TCGGCGGAGGGACCAAGCTGACCGTCCTAGGT BAK1185E1 HEAVY CHAIN-SEQ ID NO: 125 CAGGTGCAGCTGGTGCAGTCTGGGGCTGAGGTGAAGAAGCCTGGGGCCTCAGTGAAGGT CTCCTGCAAGGCTTCTGGTTACACCTTTACAAATTATGGTCTCAGCTGGGTGCGACAGG CCCCTGGACAAGGGCTTGAGTGGATGGGATGGATCAACGACGCCACCGGCGACACACAG TATGGACAGGAATTCCAGGGCAGAGTCACCATGACCACAGATACATCCACGAGCACAGC CTACATGGAGTTGAGGAGCCTGAGATCTGACGACACGGCCGTTTATTACTGTGCGAGAG ACTCCAGCAGCAGCTGGGCCCGCTGGTTTTTCGATCTCTGGGGCCGGGGGACACTGGTC ACCGTCTCCTCA BAK1185E1 LIGHT CHAIN-SEQ ID NO: 126 TCCTATGTGCTGACTCAGCCACCCTCGGTGTCAGTGGCCCCAGGAAAGACGGCCAGGAT TACCTGTGGGGGAAACATCATTGGAAGTAAACTTGTACACTGGTACCAGCAGAAGCCAG GCCAGGCCCCTGTGCTGGTCATCTATGATGATGGCGACCGGCCCTCAGGGATCCCTGAG CGATTCTCTGGCTCCAACTCTGGGAACACGGCCACCCTGACCATCAGCAGGGTCGAGGC CGGGGATGAGGCCGACTATTATTGTCAGGTGTGGGATACTGGTAGTGATCCCGTGGTAT TCGGCGGAGGGACCAAGCTGACCGTCCTAGGT 141 BAK1185F8 HEAVY CHAIN-SEQ ID NO: 127 CAGGTGCAGCTGGTGCAGTCTGGGGCTGAGGTGAAGAAGCCTGGGGCCTCAGTGAAGGT CTCCTGCAAGGCTTCTGGTTACACCTTTACAGATTATGGTCTCAGCTGGGTGCGACAGG CCCCTGGACAAGGGCTAGAGTGGATGGGATGGATCCGCAACATCGACGGCTACACAATT TATGGACAGGAATTCCAGGGCAGAGTCACCATGACCACAGATACATCCACGAGCACAGC CTACATGGAGTTGAGGAGCCTGAGATCTGACGACACGGCCGTTTATTACTGTGCGAGAG ACTCCAGCAGCAGCTGGGCCCGCTGGTTTTTCGATCTCTGGGGCCGGGGGACACTGGTC ACCGTCTCCTCA BAK1185F8 LIGHT CHAIN-SEQ ID NO: 128 TCCTATGTGCTGACTCAGCCACCCTCGGTGTCAGTGGCCCCAGGAAAGACGGCCAGGAT TACCTGTGGGGGAAACATCATTGGAAGTAAACTTGTACACTGGTACCAGCAGAAGCCAG GCCAGGCCCCTGTGCTGGTCATCTATGATGATGGCGACCGGCCCTCAGGGATCCCTGAG CGATTCTCTGGCTCCAACTCTGGGAACACGGCCACCCTGACCATCAGCAGGGTCGAGGC CGGGGATGAGGCCGACTATTATTGTCAGGTGTGGGATACTGGTAGTGATCCCGTGGTAT TCGGCGGAGGGACCAAGCTGACCGTCCTAGGT BAK1187B4 HEAVY CHAIN-SEQ ID. NO: 129 CAGGTCCAGCTGGTGCAGTCTGGGGCTGAGGTGAAGAAGCCTGGGGCCTCAGTGAAGGT CTCCTGCAAGGCTTCTGGTTACACCTTTACAAATTATGGTCTCAGCTGGGTGCGACAGG CCCCTGGACAAGGGCTTGAGTGGATGGGATGGATCGACGACGACAGCGGCACGACAATA TATGGACAGGAATTCCAGGGCAGAGTCACCATGACCACAGATACATCCACGAGCACAGC CTACATGGAGTTGAGGAGCCTGAGATCTGACGACACGGCCGTTTATTACTGTGCGAGAG ACTCCAGCAGCAGCTGGGCCCGCTGGTTTTTCGATCTCTGGGGCCGGGGGACACTGGTC ACCGTCTCCTCA BAK1187B4 LIGHT CHAIN-SEQ ID NO: 130 TCCTATGTGCTGACTCAGCCACCCTCGGTGTCAGTGGCCCCAGGAAAGACGGCCAGGAT TACCTGTGGGGGAAACATCATTGGAAGTAAACTTGTACACTGGTACCAGCAGAAGCCAG GCCAGGCCCCTGTGCTGGTCATCTATGATGATGGCGACCGGCCCTCAGGGATCCCTGAG CGATTCTCTGGCTCCAACTCTGGGAACACGGCCACCCTGACCATCAGCAGGGTCGAGGC CGGGGATGAGGCCGACTATTATTGTCAGGTGTGGGATACTGGTAGTGATCCCGTGGTAT TCGGCGGAGGGACCAAGCTGACCGTCCTAGGT 142 BAKll66G02 HEAVY CHAIN-SEQ ID NO: 131 CAAGTGCAGTTGGTGCAGTCTGGGGCTGAGGTGAAGAAGCCTGGGGCCTCAGTGAAGGT CTCCTGCAAGGCTTCTGGTTACACCTTTGCGAACACCGGGATCTCGTGGGTGCGACAGG CCCCTGGACAAGGGCTTGAGTGGATGGGATGGATCAGCGCTAATAATGGCGACACAAAT TATGGACAGGAATTCCAGGGCAGAGTCACCATGACCACAGATACATCCACGAGCACAGC CTACATGGAGTTGAGGAGCCTGAGATCTGACGACACGGCCGTTTATTACTGTGCGAGAG ACTCCAGCAGCAGCTGGGCCCGCTGGTTTTTCGATCTCTGGGGTCGGGGGACACTGGTC ACCGTCTCCTCA BAKll66G02 LIGHT CHAIN-SEQ ID NO: 132 TCCTATGTGCTGACTCAGCCACCCTCGGTGTCAGTGGCCCCAGGAAAGACGGCCAGGAT TACCTGTGGGGGAAACATCATTGGAAGTAAACTTGTACACTGGTACCAGCAGAAGCCAG GCCAGGCCCCTGTGCTGGTCATCTATGATGATGGCGACCGGCCCTCAGGGATCCCTGAG CGATTCTCTGGCTCCAACTCTGGGAACACGGCCACCCTGACCATCAGCAGGGTCGAGGC CGGGGATGAGGCCGACTATTATTGTCAGGTGTGGGATACTGGTAGTGATCCCGTGGTAT TCGGCGGAGGGACCAAGCTGACCGTCCTAGGT BAK165E7 HEAVY CHAIN-SEQ ID NO: 133 EVQLVQSGAEVKKPGASVKVSCKASGYTFTNYGLSWVRQAPGQGLEWMGWISANNGETN YGQEFQGRVTMTTETPTNTAHMELRSLTSDDTAVYYCVRDSSSNWARWYFDLWGQGTLV TVSS BAK165E7 LIGHT CHAIN-SEQ ID NO: 134 SYVLTQPPSVSVAPGQTARIPCGGNNIGSKLVHWYQQKPGQAPVLVVYDDGDRPSGIPE RFSGSNSGNTATLTISRIDAGDEADYYCQVWDTGSDPVVFGGGTKLTVLG BAK165E7 HEAVY CHAIN CDR1-SEQ ID NO: 135 NYGLS CDR2-SEQ ID NO: 136 WISANNGETNYGQEFQG CDR3-SEQ ID NO: 137 DSSSNWARWYFDL BAK165E7 LIGHT CHAIN CDR1-SEQ ID NO: 138 GGNNIGSKLVH 143 CDR2-SEQ ID NO: 139 DDGDRPS CDR3-SEQ ID NO: 140 QVWDTGSDPVV BAK582F7 HEAVY CHAIN CDR1-SEQ ID NO 141: SYAMS CDR2-SEQ ID NO 142: AISGSGGSTYYADSVKG CDR3-SEQ ID NO 143: VGAAGEGYYGY BAK582F7 LIGHT CHAIN CDR1-SEQ ID NO 144: TRSSGSIASNYVE CDR2-SEQ ID NO 145: DDNQRPS CDR3-SEQ ID NO 146: QSYDSNNDV BAK612B5 HEAVY CHAIN CDR1-SEQ ID NO 147: SYAMS CDR2-SEQ ID NO 148: AISGSGGSTYYADSVKG CDR3-SEQ ID NO 149: VGRATTDEGYYGY BAK612B5 LIGHT CHAIN CDR1-SEQ ID NO 150: TRSSGSIASNYVQ CDR2-SEQ ID NO 151: DDNQRPS CDR3-SEQ ID NO 152: QSYDSNNDV BAK615E3 HEAVY CHAIN CDR1-SEQ ID NO 153: SYAMS CDR2-SEQ ID NO 154: AISGSGGSTYYADSVKG CDR3-SEQ ID NO 155: VGKATTEEGYY BAK615E3 LIGHT CHAIN CDR1-SEQ ID NO 156: TRSSGSIASNYVQ CDR2-SEQ ID NO 157: DDNQRPS CDR3-SEQ ID NO 158: QSYDSNNDV 144 BAK0278D6 HEAVY CHAIN CDR1-SEQ ID NO 159: AATTATGGTCTCAGC CDR2-SEQ ID NO 160: TGGATCAGCGCTAATAATGGCGACACAAATTAT GGACAGGAATTCCAGGGC CDR3-SEQ ID NO 161: GACTCCAGCAGCAACTGGGCCCGCTGGTTTTTC GATCTC BAK278D6 LIGHT CHAIN CDR1-SEQ ID NO 162: GGGGGAAACAACATTGGAAGTAAACTTGTACAC CDR2-SEQ ID NO 163: GATGATGGCGACCGGCCCTCA CDR3-SEQ ID NO 164: CAGGTGTGGGATACTGGTAGTGATCCCGTGGTA BAK502G9 HEAVY CHAIN CDR1-SEQ ID NO 165: AATTATGGTCTCAGC CDR2-SEQ ID NO 166: TGGATCAGCGCTAATAATGGCGACACAAATTATGGACA GGAATTCCAGGGC CDR3-SEQ ID NO 167: GACTCCAGCAGCAGCTGGGCCCGCTGGTTTTTCGATCTC BAK502G9 LIGHT CHAIN CDR1-SEQ ID NO 168: GGGGGAAACATCATTGGAAGTAAACTTGTACAC CDR2-SEQ ID NO 169: GATGATGGCGACCGGCCCTCA CDR3-SEQ ID NO 170: CAGGTGTGGGATACTGGTAGTGATCCCGTGGTA CH Domains-SEQ ID NO: 171 ASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQS SGLYSLSSVVTVPSSSLGTKTYTCNVDHKPSNTKVDKRVESKYGPPCPSCPAPEFLGGP SVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKPREEQFN STYRVVSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQE EMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSRLTVDKS RWQEGNVFSCSVMHEALHNHYTQKSLSLSLGK CL Domain-SEQ ID NO: 172 QPKAAPSVTLFPPSSEELQANKATLVCLISDFYPGAVTVAWKADSSPVKAGVETTTPSK QSNNKYAASSYLSLTPEQWKSHRSYSCQVTHEGSTVEKTVAPTECS 145

Claims (36)

1. An isolated specific binding member that binds a primate IL-13, wherein said binding member comprises a set of CDR's comprising amino acid sequences HCDR1, HCDR2, HCDR3, LCDR1, LCDR2 and LCDR3, said set of CDR's being selected from the sets of CDR's set forth in Table 1 other than the following: (a) a BAK278D6 set of CDR's, wherein the HCDR1 has the amino acid sequence of SEQ ID NO: 1, the HCDR2 has the amino acid sequence of SEQ ID NO: 2, the HCDR3 has the amino acid sequence of SEQ ID NO: 3, the LCDR1 has the amino acid sequence of SEQ ID NO: 4, the LCDR2 has the amino acid sequence of SEQ ID NO: 5, and the LCDR3 has the amino acid sequence of SEQ ID NO: 6; or (b) a BAK502G9 set of HCDR's, wherein the HCDR1 has the amino acid sequence of SEQ ID NO: 7, the HCDR2 has the amino acid sequence of SEQ ID NO: 8, the HCDR3 has the amino acid sequence of SEQ ID NO: 9, the LCDR1 has the amino acid sequence of SEQ ID NO: 10, the LCDR2 has the amino acid sequence of SEQ ID NO: 11, and the LCDR3 has the amino acid sequence of SEQ ID NO: 12, wherein an antibody antigen-binding site of said binding member comprises: (a) a human antibody VH domain comprising said amino acid sequences HCDR1, HCDR2 and HCDR3; and (b) a human antibody VL domain comprising said amino acid sequences LCDR1, LCDR2 and LCDR3.
2. The isolated specific binding member according to claim 1, wherein the set of CDR's comprises one or two amino acid substitutions in an amino acid sequence selected from HCDR1, HCDR2, HCDR3, LCDR1, LCDR2 and LCDR3 when compared with the 146 corresponding amino acid sequence of a BAK278D6 set of CDR's using the standard numbering of Kabat.
3. The isolated specific binding member according to claim 2, wherein the one or two amino acid substitutions are at one or two residue positions within one or two CDR's of the BAK278D6 set of CDR's selected from the group consisting of: (a) residue position 31 and/or 32 and/or 34 in HCDR1; and/or (b) residue position 52and/or 52A and/or 53 and/or 54 and/or 56 and/or 58 and/or 60 and/or 61 and/or 62 and/or 64 and/or 65 in HCDR2; and/or (c) residue position 96 and/or 97 and/or 98 and/or99 and/or 101 in HCDR3; and/or (d) residue position 26 and/or 27 and/or 28 and/or 30 and/or 31 in LCDR1; and/or (e) residue position 56 in LCDR2; and/or (f) residue position 95A and/or 97 in LCDR3.
4. The isolated specific binding member according to claim 3, wherein the one or two substitutions are selected from the group consisting of: (a) a substitution of a residue at position 31 of HCDR1 for a residue selected from the group consisting of Q, D, L, G and E; and/or (b) a substitution of a residue at position 32 of HCDR1 for T; and/or (c) a substitution of a residue at position 34 of HCDR1 for a residue selected from the group consisting of V, I and F; and/or (d) a substitution of a residue at position 52 of HCDR2 for a residue selected from the group consisting of D, N, A, R, G and E; and/or 147 (e) a substitution of a residue at position 52A of HCDR2 for a residue selected from the group consisting of D, G, T, P, N and Y; and/or (f) a substitution of a residue at position 53 of HCDR2 for a residue selected from the group consisting of D, L, A, P, T, S, T and R; and/or (g) a substitution of a residue at position 54 of HCDR2 for a residue selected from the group consisting of S, T, D, G, K and I; and/or (h) a substitution of a residue at position 56 of HCDR2 for a residue selected from the group consisting of T, E, Q, L, Y, N, V, A, M and G; (i) a substitution of a residue at position 58 of HCDR2 for a residue selected from the group consisting of I, L, Q, S, M, H, D and K; and/or (j) a substitution of a residue at position 60 of HCDR2 for R; and/or (k) a substitution of a residue at position 61 of HCDR2 for R; and/or (1) a substitution of a residue at position 62 of HCDR2 for K or G; and/or (m) a substitution of a residue at position 64 of HCDR2 for R; and/or (n) a substitution of a residue at position 65 of HCDR2 for K; and/or (o) a substitution of a residue at position 96 of HCDR3 for R or D; and/or (p) a substitution of a residue at position 97 of HCDR3 for a residue selected from the group consisting of N, D, T and P; and/or (q) a substitution of a residue at position 98 of HCDR3 for R; and/or 148 (r) a substitution of a residue at position 99 of HCDR3 for a residue selected from the group consisting of S, A, I, R, P and K; and/or (s) a substitution of a residue at position 101 of HCDR3 for Y; and/or (t) a substitution of a residue at position 26 of LCDR1 for D or S; (u) a substitution of a residue at position 27 of LCDR1 for a residue selected from the group consisting of I, L, M, C, V, K, Y, F, R, T, S, A, H and G; and/or (v) a substitution of a residue at position 28 of LCDR1 for V; and/or (w) a substitution of a residue at position 30 of LCDR1 for G; and/or (x) a substitution of a residue at position 31 of LCDR1 for R; and/or (y) a substitution of a residue at position 56 of LCDR2 for T; and/or (z) a substitution of a residue at position 95A of LCDR3 for N; and/or (a') a substitution of a residue at position 97 of LCDR3 for I.
5. The isolated specific binding member according to claim 4 comprising: (a) a substitution of a residue at position 99 of HCDR3 for a residue selected from the group consisting of S, A, I, R, P and K; and/or (b) a substitution of a residue at position 27 of LCDR1 for a residue selected from the group consisting of I, L, M, C, V, K, Y, F, R, T, S, A, H and G. 149
6. The isolated specific binding member according to claim 4 or 5 comprising: (a) a substitution of a N residue at position 99 of HCDR3 for S; and/or (b) a substitution of a N residue at position 27 of LCDR1 for I.
7. The isolated specific binding member according to any one of claims 1 to 6 wherein HCDR1, HCDR2 and HCDR3 of the VH domain are within a germ-line framework and/or LCDR1, LCDR2 and LCDR3 of the VL domain are within a germ-line framework.
8. The isolated specific binding member according to claim 7, wherein the HCDR1, HCDR2 and HCDR3 of the VH domain are within germ-line framework VH1 DP-14.
9. The isolated specific binding member according to claim 7 or 8, wherein the LCDR1, LCDR2 and LCDR3 of the VL domain are within germ-line framework VL X3-3H.
10. The isolated specific binding member according to any one of claims 1 to 9, wherein said binding member binds a human IL 13 variant in which arginine at position 130 is replaced by glutamine.
11. The isolated specific binding member according to any one of claims 1 to 10, wherein the primate is a non-human primate.
12. The isolated specific binding member according to claim 11, wherein the non-human primate is rhesus or cynomolgus. 12. The specific binding member according to any one of claims 1 to 11, wherein said binding member binds IL-13 with an 150 affinity equal to or better than the affinity of an IL-13 antigen-binding site formed by the BAK502G9 VH domain (SEQ ID NO: 15) and the BAK502G9 VL domain (SEQ ID NO: 16) as determined under the same conditions.
13. The specific binding member according to any one of claims 1 to 12, wherein said binding member neutralizes human IL-13.
14. The specific binding member according to claim 13, wherein the specific binding member neutralizes human IL-13 with a potency equal to or better than the potency of a IL-13 antigen binding site formed by the BAK502G9 VH domain (SEQ ID NO: 15) and the BAK502G9 VL domain (SEQ ID NO: 16) as determined under the same conditions.
15. The specific binding member according to any one of claims 1 to 14, wherein said binding member comprises an scFv antibody molecule, an antibody constant region, or a whole antibody.
16. The specific binding member according to claim 15, wherein the whole antibody is an IgG4 isotype.
17. An isolated antibody VH domain of the specific binding member according to any one of claims 1 to 16.
18. An isolated antibody VL domain of the specific binding member according to any one of claims 1 to 16.
19. A composition comprising the specific binding member according to any one of claims 1 to 16 or the antibody VH domain according to claim 17 or the antibody VL domain according to claim 18 and at least one additional component. 151
20. The composition according to claim 19, wherein the at least one additional component comprises a pharmaceutically acceptable excipient, vehicle or carrier.
21. An isolated nucleic acid which comprises a nucleotide sequence encoding the specific binding member according to any one of claims 1 to 16 or the antibody VH domain according to claim 17 or the antibody VL domain according to claim 18.
22. A host cell transformed with the nucleic acid according to claim 21.
23. A method of producing a specific binding member or antibody VH domain or antibody VL domain, the method comprising culturing the host cell according to claim 22 under conditions sufficient to produce said specific binding member or antibody VH domain or antibody VL domain.
24. The method according to claim 23 further comprising isolating and/or purifying the specific binding member or antibody VH domain or antibody VL domain from the host cell.
25. The method according to claim 23 or 24 further comprising formulating the specific binding member or antibody VH domain or antibody VL domain into a composition, such that said composition comprises said specific binding member or antibody VH domain or antibody VL domain and at least one additional component.
26. A method for producing an antibody antigen-binding domain specific for human IL-13, the method comprising providing a VH domain comprising HCDR 1, HCDR2 and HCDR3, by way of addition, deletion, substitution or insertion of one 152 or more amino acids in the amino acid sequence of a parent VH domain comprising HCDR 1, HCDR2 and HCDR3; optionally, combining the provided VH domain with one or more VL domains to thereby provide one or more VH/VL combinations; and testing the provided VH domain or the one or more VH/VL combinations to thereby identify an antibody antigen binding domain specific for human IL-13, wherein the provided VH domain comprises HCDR1, HCDR2 and HCDR3 selected from Table 1 other than a VH domain comprising SEQ ID NOS: 1 and 2 and 3, or a VH domain comprising SEQ ID NOS: 7 and 8 and 9.
27. The method according to claim 26, wherein the parent VH domain comprises an amino acid sequence selected from the group consisting of SEQ ID NO: 13 and SEQ ID NO: 15.
28. The method according to claim 26 or 27, further comprising providing a VL domain comprising LCDR 1, LCDR2 and LCDR3, by way of addition, deletion, substitution or insertion of one or more amino acids in the amino acid sequence of a parent VL domain comprising LCDR 1, LCDR2 and LCDR3.
29. The method according to claim 28 wherein the parent VL domain comprises an amino acid sequence selected from the group consisting of SEQ ID NO: 14 and SEQ ID NO: 16.
30. A method for producing an antibody antigen-binding domain specific for human IL-13, the method comprising providing a VL domain comprising LCDR 1, LCDR2 and LCDR3, by way of addition, deletion, substitution or insertion of one or more amino acids in the amino acid sequence of a parent VL domain comprising LCDR 1, LCDR2 and LCDR3; 153 optionally, combining the provided VL domain with one or more VH domains to thereby provide one or more VH/VL combinations; and testing the provided VL domain or the one or more VH/VL combinations to thereby identify an antibody antigen binding domain specific for human IL-13, wherein the provided VL domain comprises LCDR1, LCDR2 and LCDR3 selected from Table 1 other than a VH domain comprising SEQ ID NOS: 4 and 5 and 6, or a VH domain comprising SEQ ID NOS: 10 and 11 and 12.
31. The method according to claim 30 wherein the parent VL domain comprises an amino acid sequence selected from the group consisting of SEQ ID NO: 14 and SEQ ID NO: 16.
32. The method according to claim 30 or 31, further comprising providing a VH domain comprising HCDR 1, HCDR2 and HCDR3, by way of addition, deletion, substitution or insertion of one or more amino acids in the amino acid sequence of a parent VH domain comprising HCDR 1, HCDR2 and HCDR3.
33. A method for producing an antibody antigen-binding domain specific for human IL-13, the method comprising providing a VH domain comprising HCDR 1, HCDR2 and HCDR3, by way of addition, deletion, substitution or insertion of one or more amino acids in the amino acid sequence of a parent VH domain comprising HCDR 1, HCDR2 and HCDR3, wherein the parent VH domain is selected from the group consisting of: (a) a VH domain comprising a HCDR1 sequence set forth in SEQ ID NO: 55, a HCDR2 sequence set forth in SEQ ID NO: 56, and a HCDR3 sequence set forth in SEQ ID NO: 57; (b) a VH domain comprising a HCDR1 sequence set forth in SEQ ID NO: 153, a HCDR2 sequence set forth in SEQ 154 ID NO: 154, and a HCDR3 sequence set forth in SEQ ID NO: 155; (c) a VH domain comprising a HCDR1 sequence set forth in SEQ ID NO: 141, a HCDR2 sequence set forth in SEQ ID NO: 142, and a HCDR3 sequence set forth in SEQ ID NO: 143; and (d) a VH domain comprising a HCDR1 sequence set forth in SEQ ID NO: 147, a HCDR2 sequence set forth in SEQ ID NO: 148, and a HCDR3 sequence set forth in SEQ ID NO: 149; optionally combining the VH domain thus provided with one or more VL domains to provide one or more VH/VL combinations; and testing the provided VH domain or the VH/VL combination(s) to identify an antibody antigen binding domain specific for human IL-13.
34. The method according to claim 33, wherein the parent VH domain amino acid sequence is selected from the group consisting of SEQ ID NO: 55 and SEQ ID NO: 33.
35. The method according to claim 33 or 34, further comprising providing a VL domain comprising LCDR 1, LCDR2 and LCDR3 by way of addition, deletion, substitution or insertion of one or more amino acids in the amino acid sequence of a parent VL domain comprising LCDR 1, LCDR2 and LCDR3, wherein the parent VL domain is selected from the group consisting of: (a) a VL domain comprising a LCDR1 sequence set forth in SEQ ID NO: 58, a LCDR2 sequence set forth in SEQ ID NO: 59, and a LCDR3 sequence set forth in SEQ ID NO: 60; (b) a VL domain comprising a LCDR1 sequence set forth in SEQ ID NO: 156, a LCDR2 sequence set forth in SEQ ID 155 NO: 157, and a LCDR3 sequence set forth in SEQ ID NO: 158; (c) a VL domain comprising a LCDR1 sequence set forth in SEQ ID NO: 144, a LCDR2 sequence set forth in SEQ ID NO: 145, and a LCDR3 sequence set forth in SEQ ID NO: 146; and (a) a VL domain comprising a LCDR1 sequence set forth in SEQ ID NO: 150, a LCDR2 sequence set forth in SEQ ID NO: 151, and a LCDR3 sequence set forth in SEQ ID NO:
152. 36. The method according to claim 35, wherein the parent VL domain comprises an amino acid sequence selected from the group consisting of SEQ ID NO: 24 and SEQ ID NO: 34. 37. The method according to any one of claims 23 to 36, wherein providing a VH domain and/or a VL domain comprises performing CDR mutagenesis on the parent VH domain and/or the parent VL domain or nucleic acid encoding the parent VH domain and/or the parent VL domain. 38. A method of producing a specific binding member that binds human IL-13, wherein the method comprises: providing one or more starting nucleic acids, each encoding a VH domain or a part thereof comprising CDR1 and/or CDR2 and/or CDR3; combining a starting nucleic acid with one or more donor nucleic acid to thereby insert the donor nucleic acid(s) into the starting nucleic acid or into the HCDR1 and/or HCDR2 and/or HCDR3 thereof wherein donor nucleic acid(s) is(are) inserted into the CDR1 and/or CDR2 and/or CDR3 region of the starting nucleic acid so as to produce a product repertoire of nucleic acids each encoding a VH domain or a part thereof comprising 156 HCDR1 and/or HCDR2 and/or HCDR3, said VH domain being other than a VH domain comprising SEQ ID NOS: 1 and 2 and 3 or a VH domain comprising SEQ ID NOS: 7 and 8 and 9; expressing the nucleic acids of the product repertoire to thereby produce VH domain(s); optionally, combining said produced VH domains with one or more VL domains; selecting a specific binding member specific for human IL 13, which specific binding member comprises a produced VH domain and optionally a VL domain; and recovering said specific binding member or nucleic acid encoding the specific binding member. 39. The method according to claim 38, wherein said method further comprises producing a donor nucleic acid by mutation of nucleic acid(s) encoding HCDR1 and/or HCDR2 and/or HCDR3. 40. The method according to claim 39, wherein a donor nucleic acid is produced by mutation of nucleic acid(s) encoding HCDR1 and/or HCDR2. 41. The method according to claim 39, wherein a donor nucleic acid is produced by mutation of nucleic acid encoding HCDR3. 42. The method according to any one of claims 37 to 41, comprising performing random mutagenesis to produce nucleic acid(s) encoding the VH domain or HCDR1 and/or HCDR2 and/or HCDR3 thereof and/or to produce nucleic acid(s) encoding the VL domain or LCDR1 and/or LCDR2 and/or LCDR3 thereof. 43. The method according to any one of claims 23 to 42, wherein said method further comprises attaching a VH domain to an antibody constant region. 157 44. The method according to any one of claims 23 to 43, whrein said method further comprises providing a VH domain and/or the VL domain within an IgG, scFv or Fab antibody molecule. 45. The method according to claim 44, wherein said method further comprises providing an IgG, scFv or Fab antibody molecule comprising the produced specific binding member. 46. The method according to any one of claims 23 to 45, wherein said method further comprises testing the antibody antigen binding domain specific for human IL-13 and/or testing the specific binding member that binds human IL-13 for an ability to neutralize human IL-13, and identifying an antibody antigen binding domain or specific binding member that binds to human IL-13 and/or neutralizes human IL-13. 47. The method according to claim 46, wherein said method further comprises testing the antibody antigen-binding domain specific for human IL-13 and testing the specific binding member that binds human IL-13 for an ability to neutralize human IL-13, and identifying an antibody antigen-binding domain or specific binding member that binds to human IL-13 and neutralizes human IL-13. 48. The method according to claim 47, wherein the identified antibody antigen-binding domain or specific binding member that binds to human IL-13 and neutralizes human IL-13 is an scFv antibody molecule or an Fab antibody molecule. 49. The method according to claim 48, wherein wherein the identified antibody antigen-binding domain or specific binding member that binds to human IL-13 and neutralizes human IL-13 is 158 an scFv antibody molecule, and wherein said method further comprises providing a VH domain and/or VL domain of said scFv molecule antibody molecule in a whole antibody. 50. The method according to claim 48, wherein the identified antibody antigen-binding domain or specific binding member that binds to human IL-13 and neutralizes human IL-13 is an Fab antibody molecule, and wherein said method further comprises providing a VH domain and/or VL domain of said Fab antibody molecule in a whole antibody.. 51. The method according to any one of claims 23 to 50, wherein said method further comprises formulating the antibody antigen-binding domain or specific binding member that binds IL-13,or VH domain and/or VL domain thereof into a composition comprising at least one additional component. 52. The method according to any one of claims 23 to 50, wherein said method further comprises binding the antibody antigen-binding domain or specific binding member that binds IL-13 or VH domain and/or VL domain thereof or composition comprising same to IL-13 or a fragment of IL-13. 53. A specific binding member when produced by performing the method according to any one of claims 23 to 52. 54. A composition comprising the specific binding member according to claim 53 and a pharmaceutically acceptable excipient, vehicle or carrier. 55. A method comprising binding the specific binding member according to any one of claims 1 to 16 or 53 or the composition 159 according to any one of claims 19, 20 or 54 to human IL-13 or a fragment of human IL-13. 56. The method according to claim 55, wherein said method further comprises determining a level of binding of the specific binding member to IL-13 or a fragment of IL-13. 57. Use of the specific binding member according to any one of claims 1 to 16 or 53 in the manufacture of a medicament for treatment of a disease or disorder selected from the group consisting of asthma, atopic dermatitis, allergic rhinitis, fibrosis, inflammatory bowel disease and Hodgkin's lymphoma. 58. A method of treatment of a disease or disorder selected from the group consisting of asthma, atopic dermatitis, allergic rhinitis, fibrosis and Hodgkin's lymphoma, the method comprising administering the specific binding member according to any one of claims 1 to 16 or 53 or the composition according to any one of claims 19, 20 or 54 to a patient with the disease or disorder or at risk of developing the disease or disorder. 59. An isolated specific binding member for human IL-13, comprising an antibody antigen-binding domain site which is composed of a human antibody VH domain and a human antibody VL domain and which comprises a set of CDR's, HCDR1, HCDR2, HCDR3, LCDR1, LCDR2 and LCDR3, wherein the VH domain comprises HCDR1, HCDR2 and HCDR3 and the VL domain comprises LCDR1, LCDR2 and LCDR3, wherein: (a) HCDR1 is of amino acid sequence other than SEQ ID NO: 1 or 7 having the formula HXi HX 2 G HX 3 S wherein: 160 HXi is selected from the group consisting of N, Q, D, L, G and E; HX 2 is selected from the group consisting of Y and T; and HX 3 is selected from the group consisting of V, I, F and L; (b) HCDR2 is of amino acid sequence other than SEQ ID NO: 2 or 8 having the formula W I HX 4 HX 5 HX 6 HX 7 G HX 8 T HX 9 Y HX 10 HXii HX 12 F HX 13 HX 14 wherein: HX 4 is selected from the group consisting of S, D, N, A, R, G and E; HX 5 is selected from the group consisting of A, D, G, T, P, N and Y; HX 6 is selected from the group consisting of N, D, L, A, P, T, S, I and R; HX, is selected from the group consisting of N, S, T, D, G, K and I; HX 8 is selected from the group consisting of D, T, E, Q, L, Y, N, V, A, M and G; HX 9 is selected from the group consisting of N, I, L, Q, S, M, H, D and K; HX 10 is selected from the group consisting of G and R; HXii is selected from the group consisting of Q and R; HX 12 is selected from the group consisting of E, K and G; HX 13 is selected from the group consisting of Q and R; and HX 14 is selected from the group consisting of G and K; (c) HCDR3 is of amino acid sequence other than SEQ ID NO: 3 or 9 having the formula D HX 15 HX 16 HX 17 HX 18 W A R W HX 19 F HX 2 0 L wherein: 161 HX 15 is selected from the group consisting of S, R and D; HX 16 is selected from the group consisting of S, N, D, T and P; HX 1 is selected from the group consisting of S and R; HX 18 is selected from the group consisting of S, N, A, I, R, P and K; HX 19 is selected from the group consisting of F and Y; HX 20 is selected from the group consisting of D and Y; (d) LCDR1 is of amino acid sequence other than SEQ ID NO: 4 or 10 having the formula G G LXi LX 2 LX 3 G LX 4 LX 5 L V H wherein: LXi is selected from the group consisting of N, D and S; LX 2 is selected from the group consisting of N, I, L, M, C, V, K, Y, F, R, T, S, A, H and G; LX 3 is selected from the group consisting of I and V; LX 4 is selected from the group consisting of S and G; and LX 5 is selected from the group consisting of K and R; (e) LCDR2 is of amino acid sequence other than SEQ ID NO: 5 or 11 having the formula D D G D R P LX 6 wherein: LX6 is selected from the group consisting of S and T; and (f) LCDR3 is of amino acid sequence other than SEQ ID NO: 6 or 12 having the formula Q V W D T G S LX 7 P V LX 8 wherein: LX, is selected from the group consisting of D and N; and 162 LX8 is selected from the group consisting of V and I. 60. The isolated specific binding member according to claim 59, wherein: HXi is selected from the group consisting of D and N; HX 2 is Y; HX 3 is L; HX 4 is selected from the group consisting of S and G; HX 5 is selected from the group consisting of T and A; HX 6 is N; HX 7 is selected from the group consisting of N and I, HX 8 is D; HX 9 is selected from the group consisting of N, D and K; HXio is G; HX 12 is selected from the group consisting of E and G, HX 13 is Q; HX 19 is F; LXi is selected from the group consisting of N and S; LX 2 is selected from the group consisting of N, Y, T, S, and I; LX 6 is S; and LX 7 is D. 61. The isolated specific binding member according to claim 59, wherein: HXi is selected from the group consisting of N and D; HX 2 is Y; HX 3 is L; HX 4 is selected from the group consisting of S and G; HX 5 is selected from the consisting of A and T; HX 6 is N; HX 7 is N; HX 8 is selected from the group consisting of D and G; 163 HX 9 is selected from the group consisting of I, S, N and D; HXii is Q; HX 12 is E and K; HX 14 is G; HX 15 is S; HX 16 is selected from the group consisting of S and N; HX 17 is S; HX 18 is selected from the group consisting of S and N; HX 19 is F; HX 2 0 is D; LXi is selected from the group consisting of N and D; LX 3 is I; and LX 8 is V. 62. The isolated specific binding member according to claim 59, wherein: HX, is selected from the group consisting of N, S, T, D, G and K; HX 8 is selected from the group consisting of D, T, E, Q, L, Y, N, V, A, M; HX 9 is selected from the group consisting of N, I, L, Q, S, M and H; HXio is G; HXii is Q; HX 12 is F; HXi 3 is Q; HX 14 is G; HX 15 is S; HX 16 is selected from the group consisting of N and S; HX 17 is S; HX 18 is selected from the group consisting of N and S; HX 19 is F; 164 HX 2 0 is D; LXi is N; LX 2 is selected from the group consisting of N and I; LX 3 is I; LX 4 is S; LX 5 is K; LX 6 is S; LX, is D; and LX 8 is V. 63 The specific binding member according to claim 62 wherein: HXi is selected from the group consisting of N, Q and D; HX 3 is selected from the group consisting of L, V and HX 4 is selected from the group consisting of S, N, A and R; HX 5 is selected from the group consisting of A, D, T, G, N and Y; HX 6 is selected from the group consisting of N, A, P, S, D and I; HX7 is selected from the group consisting of N, T, D and G; HX 8 is selected from the group consisting of D,Q, Y and N; and HX 9 is selected from the group consisting of N, Q, S and I. 64 The specific binding member according to any one of claims 59 to 63, wherein said binding member neutralizes human IL-13. 65. The specific binding member according to claim 64, wherein said binding member neutralizes human IL-13, with a potency equal to or better than the potency of a IL-13 antigen-binding site formed by the BAK502G9 VH domain (SEQ ID NO: 15) and the 165 BAK502G9 VL domain (SEQ ID NO: 16), the potency of the specific binding member and the potency of the antigen-binding site being as determined under the same conditions. 66. The specific binding member according to any one of claims 59 to 65 that comprises an scFv antibody molecule. 67. The specific binding member according to any one of claims 59 to 65 that comprises an antibody constant region. 68. The specific binding member according to claim 67 that comprises a whole antibody. 69. The specific binding member according to claim 68, wherein the whole antibody is IgG4. 70. The isolated specific binding member according to any one of claims 59 to 69, wherein said binding member binds a human IL-13 variant in which arginine at position 130 is replaced by glutamine. 71. The isolated specific binding member according to any one of claims 59 to 69, wherein said binding member binds a non human primate IL-13. 72. The isolated specific binding member according to claim 71, wherein the non-human primate IL-13 is rhesus or cynomolgus. 73. An isolated antibody VH domain of the specific binding member according to any one of claims 59 to 72. 166 74. An isolated antibody VL domain of a specific binding member according to any one of claims 59 to 72. 75. A composition comprising the specific binding member, antibody VH domain or antibody VL domain of any one of claims 59 to 74 and at least one additional component. 76. The composition according to claim 75 comprising a pharmaceutically acceptable excipient, vehicle or carrier. 77. An isolated nucleic acid comprising a nucleotide sequence encoding the specific binding member or antibody VH or VL domain of a specific binding member according to any one of claims 59 to 74. 78. A host cell transformed with the isolated nucleic acid according to claim 77. 79. A method of producing a specific binding member or antibody VH or VL domain, the method comprising culturing the host cell according to claim 78 under conditions sufficient to produce a specific binding member or antibody VH or VL domain encoded by the isolated nucleic acid. 80. The method according to claim 79, wherein said method further comprises isolating and/or purifying the produced specific binding member or antibody VH or VL variable domain. 81. The method according to claim 79 or 80, wherein said method further comprises formulating the specific binding member or antibody VH or VL variable domain into a composition by admixture with at least one additional component. 167 82. The method according to any one of claims 79 to 81, wherein said method further comprises binding the specific binding member to human IL-13 to IL-13 or a fragment of IL-13. 83. A method comprising binding the specific binding member according to any one of claims 59 to 72 to human IL-13 or a fragment of human IL-13. 84. The method according to claim 82 or 83, wherein said method further comprises determining a level of binding of specific binding member to IL-13 or a fragment of IL-13. 85. The method according to any one of claims 79 to 84, wherein said method further comprises manufacturing a medicament comprising the specific binding member for treatment of a disease or disorder selected from the group consisting of asthma, atopic dermatitis, allergic rhinitis, fibrosis, inflammatory bowel disease and Hodgkin's lymphoma. 86. Use of a specific binding member according to any one of claims 59 to 72 in the manufacture of a medicament for treatment of a disease or disorder selected from the group consisting of asthma, atopic dermatitis, allergic rhinitis, fibrosis, inflammatory bowel disease and Hodgkin's lymphoma. 87. A method of treatment of a disease or disorder selected from the group consisting of asthma, atopic dermatitis, allergic rhinitis, fibrosis and Hodgkin's lymphoma, the method comprising administering the specific binding member according to any one of claims 59 to 72 to a patient with the disease or disorder or at risk of developing the disease or disorder. 168 88. The specific binding member according to any one of claims 1 to 16, 53, or 59 to 72, or the antibody VH domain of claim 17 or 73, or the antibody VL domain of claim 18 or 74, or the composition according to any one of claims 19, 20, 54, 75 or 76, or the isolated nucleic acid of claim 21 or 77, or the host cell of claim 22 or 78, or the method according to any one of claims 23 to 52, 55, 56, 58, 79 to 85, or 87, or the use according to claim 57 or 86, substantially as described herein with reference to any one or more of the Examples and/or Drawings. DATED this TWENTY FIFTH day of JANUARY, 2011 MedImmune Limited By patent attorneys for the applicant: FB RICE & CO 169
AU2011200308A 2003-07-15 2011-01-25 Human antibody molecules for IL-13 Active AU2011200308B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2011200308A AU2011200308B2 (en) 2003-07-15 2011-01-25 Human antibody molecules for IL-13

Applications Claiming Priority (11)

Application Number Priority Date Filing Date Title
US48751203P 2003-07-15 2003-07-15
US60/487,512 2003-07-15
US55821604P 2004-03-31 2004-03-31
US60/558,216 2004-03-31
GBGB0407315.1A GB0407315D0 (en) 2003-07-15 2004-03-31 Human antibody molecules
GB0407315.1 2004-03-31
US57379104P 2004-05-24 2004-05-24
US60/573,791 2004-05-24
PCT/GB2004/003059 WO2005007699A2 (en) 2003-07-15 2004-07-15 Human antibody molecules for il-13
AU2004256976A AU2004256976B2 (en) 2003-07-15 2004-07-15 Human antibody molecules for IL-13
AU2011200308A AU2011200308B2 (en) 2003-07-15 2011-01-25 Human antibody molecules for IL-13

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU2004256976A Division AU2004256976B2 (en) 2003-07-15 2004-07-15 Human antibody molecules for IL-13

Publications (2)

Publication Number Publication Date
AU2011200308A1 true AU2011200308A1 (en) 2011-02-17
AU2011200308B2 AU2011200308B2 (en) 2011-07-28

Family

ID=44535047

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2011200308A Active AU2011200308B2 (en) 2003-07-15 2011-01-25 Human antibody molecules for IL-13

Country Status (8)

Country Link
JP (3) JP5519567B2 (en)
KR (1) KR101073590B1 (en)
AU (1) AU2011200308B2 (en)
CY (1) CY1111467T1 (en)
HK (3) HK1097853A1 (en)
MX (1) MXPA06000258A (en)
NO (2) NO338853B1 (en)
RU (1) RU2009148270A (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2952241A1 (en) * 2014-06-18 2015-12-23 Medimmune, Llc Cell culture methods and media comprising n-acetylcysteine

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU5010793A (en) * 1992-08-21 1994-03-15 Schering Corporation Human interleukin-13
US20030235555A1 (en) * 2002-04-05 2003-12-25 David Shealey Asthma-related anti-IL-13 immunoglobulin derived proteins, compositions, methods and uses
JP2005512522A (en) * 2001-10-26 2005-05-12 セントカー・インコーポレーテツド IL-13 mutein proteins, antibodies, compositions, methods and uses
AU2002359568B2 (en) * 2001-12-03 2008-02-21 Alexion Pharmaceuticals, Inc. Hybrid antibodies
AU2003260748A1 (en) * 2002-08-30 2004-03-19 Glaxo Group Limited Il-14 vaccine for the treatment of asthma and atopic disorders
GB0407315D0 (en) * 2003-07-15 2004-05-05 Cambridge Antibody Tech Human antibody molecules
TWI307630B (en) * 2004-07-01 2009-03-21 Glaxo Group Ltd Immunoglobulins

Also Published As

Publication number Publication date
RU2009148270A (en) 2011-06-27
JP5519567B2 (en) 2014-06-11
MXPA06000258A (en) 2006-07-03
JP2014101372A (en) 2014-06-05
NO338853B1 (en) 2016-10-24
HK1097853A1 (en) 2007-07-06
KR101073590B1 (en) 2011-10-14
AU2011200308B2 (en) 2011-07-28
KR20060054312A (en) 2006-05-22
NO20160707A1 (en) 2016-04-27
HK1216427A1 (en) 2016-11-11
CY1111467T1 (en) 2015-08-05
JP2011147454A (en) 2011-08-04
JP5922686B2 (en) 2016-05-24
HK1155185A1 (en) 2012-05-11
JP2016047843A (en) 2016-04-07
NO20060730L (en) 2006-04-10
NO342727B1 (en) 2018-07-30

Similar Documents

Publication Publication Date Title
US9856317B2 (en) Human antibody molecules for IL-13
MXPA06000240A (en) Device for measuring time-resolved volumetric throughflow processes.
AU2011200308B2 (en) Human antibody molecules for IL-13

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)