Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
DESIGNED ANKYRIN REPEAT PROTEINS BINDING TO HEPATOCYTE GROWTH FACTOR
Document Type and Number:
WIPO Patent Application WO/2014/191574
Kind Code:
A1
Abstract:
New designed ankyrin repeat proteins with binding specificity for HGF are described, as well as nucleic acids encoding such HGF binding proteins, pharmaceutical compositions comprising such proteins and the use of such proteins in the treatment of diseases.

Inventors:
TRESCH GABY (CH)
BAKKER TALITHA (CH)
PHILLIPS DOUGLAS (CH)
MERZ FRIEDER W (CH)
BINZ KASPAR H (CH)
Application Number:
PCT/EP2014/061368
Publication Date:
December 04, 2014
Filing Date:
June 02, 2014
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
MOLECULAR PARTNERS AG (CH)
International Classes:
C07K14/475; A61K38/18
Domestic Patent References:
WO2012149439A22012-11-01
WO2006105511A12006-10-05
Other References:
TOSCHI L ET AL: "Single-agent and combination therapeutic strategies to inhibit hepatocyte growth factor/MET signaling in cancer", CLINICAL CANCER RESEARCH, THE AMERICAN ASSOCIATION FOR CANCER RESEARCH, US, vol. 14, no. 19, 1 October 2008 (2008-10-01), pages 5941 - 5946, XP002712514, ISSN: 1078-0432, DOI: 10.1158/1078-0432.CCR-08-0071
BINZ H ET AL: "Designing Repeat Proteins: Well-expressed, Soluble and Stable Proteins from Combinatorial Libraries of Consensus Ankyrin Repeat Proteins", JOURNAL OF MOLECULAR BIOLOGY, ACADEMIC PRESS, UNITED KINGDOM, vol. 332, no. 2, 12 September 2003 (2003-09-12), pages 489 - 503, XP027101280, ISSN: 0022-2836, [retrieved on 20030912], DOI: 10.1016/S0022-2836(03)00896-9
BINZ H KASPAR ET AL: "High-affinity binders selected from designed ankyrin repeat protein libraries", NATURE BIOTECHNOLOGY, NATURE PUBLISHING GROUP, NEW YORK, NY, US, vol. 22, no. 5, 1 May 2004 (2004-05-01), pages 575 - 582, XP002343919, ISSN: 1087-0156, DOI: 10.1038/NBT962
Attorney, Agent or Firm:
MICHALSKI HÜTTERMANN & PARTNER PATENTANWÄLTE MBB et al. (Düsseldorf, DE)
Download PDF:
Claims:
Claims

1 . A recombinant binding protein comprising at least one ankyrin repeat domain, wherein said ankyrin repeat domain binds HGF in PBS with a dissociation constant (KD) below 10_7M.

2. The binding protein of claim 1 , wherein said ankyrin repeat domain inhibits the binding of HGF to c-Met in PBS with an IC50 value below 10 7M.

3. The binding protein of claim 1 , wherein said ankyrin repeat domain inhibits the HGF stimulated phosphorylation of c-Met in A549 cells with an IC5o value below 10~7M.

4. The binding protein of claim 1 , wherein said ankyrin repeat domain inhibits the HGF stimulated proliferation of U87 cells with an IC5o value below 10~6M. 5. The binding protein of claim 1 , wherein said ankyrin repeat domain competes for binding to HGF with an ankyrin repeat domain selected from the group consisting of SEQ ID NOs: 33 to 61 .

6. The binding protein of claim 1 , wherein said ankyrin repeat domain comprises at least an amino acid sequence that has at least 90% amino acid sequence identity with one ankyrin repeat domain selected from the group consisting of SEQ ID NOs: 33 to 61 , wherein G at position 1 and/or S at position 2 of said ankyrin repeat domain are optionally missing; and L at the second last position and/or N at the last position of said ankyrin repeat domain are optionally exchanged by A.

7. The binding protein of claim 1 , wherein said ankyrin repeat domain is selected from the group consisting of SEQ ID NOs:33 to 61 ,

wherein G at position 1 and/or S at position 2 of said ankyrin repeat domain are optionally missing; and L at the second last position and/or N at the last position of said ankyrin repeat domain are optionally exchanged by A.

8. The binding protein of claim 1 , wherein said ankyrin repeat domain comprises at least an ankyrin repeat module having an amino acid sequence selected from the group consisting of SEQ ID NO:12 to 25 and sequences in which up to 9 amino acids in SEQ ID NO:12 to 25 are exchanged by any amino acid.

9. The binding protein of claim 1 , wherein said ankyrin repeat domain comprises at least an ankyrin repeat module having an amino acid sequence selected from the group consisting of SEQ ID NO: 12 to 25 and sequences in which up to 9 amino acids in SEQ ID NO: 12 to 25 are exchanged by any amino acid, and of which up to 4 of said 9 amino acid exchanges are located in positions 1 , 3, 4, 6, 14, and/or 15 of SEQ ID NOs: 12 to 25.

10. The binding protein of claim 1 , wherein said ankyrin repeat domain comprises at least an ankyrin repeat module having the amino acid sequence

KDRYGDTPLHLAADIGHLEIVEVLLKAGADVNA (SEQ ID NO:18)

and sequences in which up to 9 amino acids in SEQ ID NO:18 are exchanged by any amino acid and

wherein

K at position 1 is optionally exchanged by Q, H, I, V

R at position 3 is optionally exchanged by A, T or N

Y at position 4 is optionally exchanged by F or W;

D at position 6 is optionally exchanged by N;

D at position 14 is optionally exchanged by S;

I at position 15 is optionally exchanged by Y or A; and

E at position 19 is optionally exchanged by K.

1 1 . The binding protein of claim 1 , wherein said ankyrin repeat domain comprises at least an ankyrin repeat module having the amino acid sequence

EDYFGNTPLHLAASYGHLEIVEVLLKAGADVNA (SEQ ID NO:19)

and sequences in which up to 9 amino acids in SEQ ID NO:19 are exchanged by any amino acid and

wherein

E at position 1 is optionally exchanged by D, F, A, or L;

Y at position 3 is optionally exchanged by W, S, or V;

F at position 4 is optionally exchanged by Y;

N at position 6 is optionally exchanged by D;

Y at position 15 is optionally exchanged by M, L, S, or A.

12. The binding protein of claim 1 , wherein said ankyrin repeat domain comprises at least the ankyrin repeat module of claim 10 and the ankyrin repeat module of claim 1 1 .

13. The binding protein according to anyone of claims 1 to 12, comprising a peptide of any one of the sequences SEQ ID NO:1 to 25 and 33 to 62.

14. A nucleic acid encoding a binding protein according to anyone of claims 1 to 13.

15. A pharmaceutical composition comprising the binding protein of anyone of claims 1 to 13 or the nucleic acid of claim 14, and optionally a pharmaceutical acceptable carrier and/or diluent.

Description:
Designed ankyrin repeat proteins binding to hepatocyte growth factor Field of the invention The present invention relates to designed ankyrin repeat proteins with binding specificity for hepatocyte growth factor (HGF) as well as nucleic acids encoding such HGF binding proteins, pharmaceutical compositions comprising such proteins and the use of such proteins in the treatment of diseases. Background of the invention

The MET proto-oncogene encodes a receptor tyrosine kinase (MET or c-Met) also known as hepatocyte growth factor receptor (HGFR; human HGFR has the UniProtKB/Swiss- Prot number P08581 ), which is essential for embryonic development and wound healing. Hepatocyte growth factor (HGF, also known as scatter factor; human HGF has the

UniProtKB/Swiss-Prot number P14210) is the only known ligand of c-Met. The receptor is normally expressed by cells of epithelial origin, while expression of HGF is restricted to cells of mesenchymal origin. Upon HGF stimulation, c-Met induces several biological responses that collectively give rise to a program known as invasive growth. Abnormal c-Met activation in cancer correlates with poor prognosis, where aberrantly active c-Met triggers tumor growth, tumor cell survival, angiogenesis and metastasis. c-Met is deregulated in many types of human malignancies, including cancers of kidney, liver, stomach, breast, and brain and is a main driver for resistance to other targeted therapies (Bottaro, D.P., Rubin, J.S., Faletto, D.L., Chan, A.M., Kmiecik, T.E., Vande Woude, G.F. and Aaronson, S.A., Science 251(4995), 802-804, 1991 ; Comoglio, P.M, Giordano, S. and Trusolino, L, Nat. Rev. Drug Discov. 7(6), 504-516, 2008).

HGF is secreted as a single inactive single-chain precursor polypeptide (sc-HGF, sometimes also referred as pro-HGF) and is cleaved by serine proteases into a 69-kDa alpha-chain and 34-kDa beta-chain. Active HGF is a disulfide linked heterodimer consisting of the alpha- and beta-chain. HGF shares a high degree of homology with coagulation factors, as the alpha-chain contains plasminogen-like "kringle" structural motifs, and the beta-chain contains a domain homologous to serine proteases, but devoid of enzymatic activity (Naldini, L, Weidner, K.M., Vigna, E., Gaudino, G., Bardelli, A., Ponzetto, C, Narsimhan, R.P., Hartmann, G., Zarnegar, R., Michalopoulos, G.K., et al., EMBO J. 10, 2867-2878, 1991 ). Through binding to its receptor, HGF mediates a number of cellular responses, including scattering of various cell types, the formation of tubules and lumens, epithelial- mesenchymal transition, angiogenesis, liver regeneration, wound healing and

embryological development. The HGF/c-Met signaling pathway has also been shown to play a role in various diseases, among which many human solid tumors, in which it participates in tumor development, invasion and metastasis. In human cancers, both paracrine and autocrine activation of c-Met by HGF, the expression of these molecules in tumors and elevated concentrations of circulating HGF are associated with poor prognosis and increased risk of tumor metastasis (Comoglio et al., 2008, loc. cit.)

Because of the active role HGF in the development in cancer and other diseases, an agent blocking HGF activity could be beneficial in countering this effect. Various anti-HGF (aHGF) monoclonal antibodies (mAbs) are under clinical development at the moment, including AMG102 (i.e. rilotumumab), a humanized antihuman HGF lgG2 from Amgen, AV-299 from Schering/Aveo, and TAK-701 from Millennium.

There are, beside antibodies, novel binding proteins or binding domains that can be used to specifically bind a target molecule (e.g. Binz, H.K., Amstutz, P. and Pluckthun, A., Nat. Biotechnol. 23, 1257-1268, 2005) and thereby act as an antagonist. One such novel class of binding proteins or binding domains not possessing an Fc are based on designed repeat proteins or designed repeat domains (WO 2002/020565; Binz, H.K., Amstutz, P., Kohl, A., Stumpp, M.T., Briand, C, Forrer, P., Grutter, M.G., and Pluckthun, A., Nat. Biotechnol. 22, 575-582, 2004; Stumpp, M.T., Binz, H.K and Amstutz, P., Drug Discov. Today 13, 695-701 , 2008). WO 2002/020565 describes how large libraries of repeat proteins can be constructed and their general application. Nevertheless, WO 2002/020565 does neither disclose the selection of repeat domains with binding specificity for HGF nor concrete repeat modules or repeat sequence motifs of repeat domains that specifically bind to HGF. Furthermore, WO 2002/020565 does not suggest that repeat domains with binding specificity for HGF could be used to regulate the HGF mediated signaling pathways to successfully treat diseases. These designed repeat domains harness the modular nature of repeat proteins and may possess N-terminal and C-terminal capping modules to prevent the designed repeat domains from aggregation by shielding the hydrophobic core of the domain (Forrer, P., Stumpp, M.T., Binz, H.K. and Pluckthun, A., FEBS letters 539, 2-6, 2003). The technical problem underlying the present invention is identifying novel binding proteins, such as ankyrin repeat proteins or domains, with binding specificity to HGF to regulate HGF mediated signaling pathways for an improved treatment of certain cancers, vascular disorders, angiogenesis related diseases and other pathological conditions. The solution to this technical problem is achieved by providing the embodiments characterized in the claims.

Summary of the invention The present invention relates to a recombinant binding protein comprising at least one ankyrin repeat domain, wherein said ankyrin repeat domain binds HGF in PBS with a Kd below 10 7 M.

More particularly, the invention relates to a recombinant binding protein comprising at least one ankyrin repeat domain, wherein said ankyrin repeat domain competes for binding to HGF with an ankyrin repeat domain selected from the group consisting of SEQ ID NOs:33 to 61

The invention further relates to a recombinant binding protein comprising at least one ankyrin repeat domain with binding specificity for HGF, which comprises an ankyrin repeat module having an amino acid sequence selected from the group consisting of SEQ ID NO:12 to 25 and sequences in which up to 9 amino acids in SEQ ID NO:12 to 25 are exchanged by any amino acid. In particular, the invention relates to a recombinant binding protein comprising at least one ankyrin repeat domain that has at least 90% amino acid sequence identity with one ankyrin repeat domain selected from the group consisting of SEQ ID NOs:33 to 61 wherein G at position 1 and/or S at position 2 of said ankyrin repeat domain are optionally missing; and L at the second last position and/or N at the last position of said ankyrin repeat domain are optionally exchanged by A.

In particular the invention relates to a recombinant HGF binding protein comprising a peptide of any one of the sequences SEQ ID NO:12 to 25 and 33 to 62. The invention further relates to nucleic acid molecules encoding the binding proteins of the present invention, and to a pharmaceutical composition comprising one or more of the above mentioned binding proteins or nucleic acid molecules. The invention further relates to a method of treatment of a pathological condition using the binding proteins of the invention.

Brief Description of the Figures Figure 1 . Surface Plasmon Resonance analysis of a selected DARPin.

Surface Plasmon Resonance (SPR) analysis of DARPin binding to HGF (exemplified by DARPin #51 ). Various concentrations (1 .6, 3.1 , 6.3, 12.5 and 25 nM) of DARPin were applied to a flow cell with immobilized human HGF for 120 seconds, followed by washing with buffer flow. A global fit of the obtained traces resulted in a Kd value of 51 pM for the DARPin #51 interaction with human HGF. RU, Resonance Units; s, time in seconds. See below for the definition of DARPin #51 .

Figure 2. Competition Surface Plasmon Resonance analysis.

SPR analysis of DARPins binding to human HGF (exemplified by DARPin #51 , DARPin #57 and DARPin #43). 10OnM of DARPin #51 (i.e. D1 ) was applied to a flow cell with immobilized human HGF for 120 seconds, followed by injection of 10OnM of a second DARPin (either DARPin #57 or DARPin #33; i.e. D2 or D3, respectively) for 60 seconds. Thus, D1 and D3 compete for binding to HGF and D1 and D2 do not compete for binding to HGF.

RU, Resonance Units; s, time in seconds; D1 , DARPin #51 ; D2, DARPin #57; D3,

DARPin #33. See below for the definition of DARPin #51 , DARPin #57 and DARPin #43.

Figure 3. Inhibition of HGF stimulated c-Met phosphorylation

The inhibition of the HGF stimulated c-Met phosphorylation in A549 cells by various concentrations of DARPins with specificity for human HGF (exemplified by DARPin #42 and #48) and corresponding fitted inhibition curves are shown. The IC 5 o values were determined to be 0.32 and 0.1 nM for DARPin #42 and 48, respectively.

OD, optical density at 450 - 620 nm; C, DARPin concentration in nM; D1 , DARPin #48; D2, DARPin #42; MA, maximal activation; Ml, minimal activation. The X axis is shown in logarithmic scale. See below for the definition of DARPin #52 and DARPin #48. Figure 4. Inhibition of U87 cell proliferation

The inhibition of the proliferation of U87 glioblastoma cells by various concentrations of DARPins with specificity for HGF(exemplified by DARPin #51 and #43) and corresponding fitted inhibition curves are shown. The IC50 values were determined to be 72and 1 16 nM for DARPin #43 and 51 , respectively.

OD, optical density at 450 - 620 nm; C, DARPin concentration in nM; D1 , DARPin #51 ; D2, DARPin #43. The X axis is shown in logarithmic scale. See below for the definition of DARPin #51 and DARPin #43. Figure 5. c-Met competition assay.

The inhibition of the binding of HGF to c-Met by various concentrations of DARPins with specificity for human HGF and the corresponding fitted inhibition curves are shown for a distinct single experiment. The IC50 values were then calculated to be about 1 .36 and 0.62 nM for the DARPins #43 and #51 , respectively. OD, optical density at 450 - 620 nm; C, concentration of DARPins in nM, D1 , DARPin #51 ; D2, DARPin #43. The X axis is shown in logarithmic scale. See below for the definitions of DARPin #51 and 43.

Figure 6 Efficacy of an anti-HGF DARPin vs vehicle in a U87 tumor xenograft model . U87-MG tumor cells were subcutaneously injected into nude mice. After tumor growth animals were randomized into four groups and treated with different concentrations (intravenous injection three-times a week, in total animals were dosed seven times) of PEGylated DARPins. The PEGylated DARPins were generated as described in Example 6.

T, tumor volume in mm 3 ; d, time in days; V, vehicle (i.e. PBS); D1 , PEGylated DARPin #62 at a dose of 0.1 1 mg/kg per injection; D2, PEGylated DARPin #62 at a dose of

1 .1 mg/kg per injection; D3, PEGylated DARPin #62 at a dose of 1 1 mg/kg per injection.

Figure 7 Multiple sequence alignment of SEQ ID NOs: 12 to 28. SEQ ID NOs: 12 to 28 are aligned in a multiple alignment according to the residue position. The ankyin repeat residue position numbers are indicated at the top. Residue positions 2, 5, 7-13, and 16-33 correspond to positions which typically contain framework residues. Residue positions 1 , 3, 4, 6, 14, and 15 correspond to positions which potentially contain target interaction residues. Detailed description of the invention

The recombinant binding protein or domain according to the invention is specific for a mammalian HGF. Preferably, the recombinant binding domain according to the invention is specific for a HGF of mice, rat, dog, rabbit, monkey or human origin. More preferably, the recombinant binding domain according to the invention is specific for a HGF of human origin.

The term "protein" refers to a polypeptide, wherein at least part of the polypeptide has, or is able to acquire a defined three-dimensional arrangement by forming secondary, tertiary, or quaternary structures within and/or between its polypeptide chain(s). If a protein comprises two or more polypeptides, the individual polypeptide chains may be linked non- covalently or covalently, e.g. by a disulfide bond between two polypeptides. A part of a protein, which individually has, or is able to acquire, a defined three-dimensional arrangement by forming secondary or tertiary structures, is termed "protein domain". Such proteins or protein domains are well known to the practitioner skilled in the art.

The term "recombinant" as used in recombinant protein, recombinant protein domain, recombinant binding protein and the like, means that said polypeptides are produced by the use of recombinant DNA technologies well known by the practitioner skilled in the relevant art. For example, a recombinant DNA molecule (e.g. produced by gene synthesis) encoding a polypeptide can be cloned into a bacterial expression plasmid (e.g. pQE30, Qiagen), yeast expression plasmid or mammalian expression plasmid. When, for example, such a constructed recombinant bacterial expression plasmid is inserted into an appropriate bacteria (e.g. Escherichia coli), this bacteria can produce the polypeptide encoded by this recombinant DNA. The correspondingly produced polypeptide is called a recombinant polypeptide.

In the context of the present invention, the term "polypeptide" relates to a molecule consisting of one or more chains of multiple, i.e. two or more, amino acids linked via peptide bonds. Preferably, a polypeptide consists of more than eight amino acids linked via peptide bonds.

The term "polypeptide tag" refers to an amino acid sequence attached to a

polypeptide/protein, wherein said amino acid sequence is useful for the purification, detection, or targeting of said polypeptide/protein, or wherein said amino acid sequence improves the physicochemical behavior of the polypeptide/protein, or wherein said amino acid sequence possesses an effector function. The individual polypeptide tags, moieties and/or domains of a binding protein may be connected to each other directly or via polypeptide linkers. These polypeptide tags are all well known in the art and are fully available to the person skilled in the art. Examples of polypeptide tags are small polypeptide sequences, for example, His (e.g. the His-tag of SEQ ID NO:9), myc, FLAG, or Strep-tags or moieties such as enzymes (for example enzymes like alkaline

phosphatase), which allow the detection of said polypeptide/protein, or moieties which can be used for targeting (such as immunoglobulins or fragments thereof) and/or as effector molecules.

The term "polypeptide linker" refers to an amino acid sequence, which is able to link, for example, two protein domains, a polypeptide tag and a protein domain, a protein domain and a non-polypeptide moiety such as polyethylene glycol or two sequence tags. Such additional domains, tags, non-polypeptide moieties and linkers are known to the person skilled in the relevant art. A list of example is provided in the description of the patent application WO 2002/020565. Particular examples of such linkers are glycine-serine- linkers and proline-threonine-linkers of variable lengths; preferably, said linkers have a length between 2 and 24 amino acids; more preferably, said linkers have a length between 2 and 16 amino acids. An example of a glycine-serine-linker is provided in SEQ ID NO:10 and an example of a proline-threonine-linker is provided in SEQ ID NO:1 1 . Preferably, the proline-threonine-linker of SEQ ID NO:1 1 is preceded by GS and/or followed by GS. The term "polymer moiety" refers to either a proteinaceous polymer moiety or a non- proteinaceous polymer moiety. A "proteinaceous polymer moiety" preferably is a polypeptide that does not form a stable tertiary structure. Examples of proteinaceous polymer moieties are XTEN® (a registered trademark of Amunix; WO 2007/103515) polypeptides, or polypeptides comprising proline, alanine and serine residues as described in WO 2008/155134. Such proteinaceous polymer moieties can be covalently attached to, for example, a binding domain of the invention by the generation of genetic fusion polypeptides using standard DNA cloning technologies, followed by their standard expression and purification. A "non-proteinaceous polymer moiety" is a polymer moiety not built from polypeptides. Examples of non-proteinaceous polymer moieties are hydroxyethyl starch (HES), polyethylene glycol (PEG), polypropylene glycol, or polyoxyalkylene. The term "PEGylated" means that a PEG moiety is covalently attached to, for example, a polypeptide of the invention. A polymer moiety of the invention may vary widely in molecular weight. Preferably, said polymer moiety is connected by a polypeptide linker to a binding domain. In a specific embodiment, a PEG moiety or any other non-proteinaceous polymer can, e.g., be coupled to a cysteine thiol via a maleimide linker with the cysteine being coupled via a peptide linker to the N or C terminus of a binding domain as described herein.

The term "binding protein" refers to a protein comprising one or more binding domains, one or more bioactive compounds and one or more polymer moieties as further explained below. Preferably, said binding protein comprises up to four binding domains. More preferably, said binding protein comprises up to two binding domains. Most preferably, said binding protein comprises only one binding domain. Furthermore, any such binding protein may comprise additional protein domains that are not binding domains, multimerization moieties, polypeptide tags, polypeptide linkers and/or one or more Cys residues. Examples of multimerization moieties are immunoglobulin heavy chain constant regions which pair to provide functional immunoglobulin Fc domains, and leucine zippers or polypeptides comprising a free thiol which forms an intermolecular disulfide bond between two such polypeptides. A Cys residue may be used for conjugating other moieties to the polypeptide, for example, by using the maleimide chemistry well known to the person skilled in the art. Preferably, said binding protein is a recombinant binding protein. Also preferably, the binding domains of binding protein possess different target specificities. The term "bioactive compound" refers to a compound that is disease modifying when applied to a mammal having said disease. A bioactive compound may have antagonistic or agonistic properties and can be a proteinaceous bioactive compound or a non- proteinaceous bioactive compound. Such proteinaceous bioactive compounds can be covalently attached to, for example, a binding domain of the invention by the generation of genetic fusion polypeptides using standard DNA cloning technologies, followed by their standard expression and purification. Such non-proteinaceous bioactive compounds can be covalently attached to, for example, a binding domain of the invention by chemical means, e.g., by coupling to a cysteine thiol via a maleimide linker with a cysteine being coupled via a peptide linker to the N or C terminus of a binding domain as described herein. Examples of proteinaceous bioactive compounds are binding domains having a distinct target specificity (e.g. neutralizing a growth factor by binding to it), cytokines (e.g. interleukins), growth factors (e.g. human growth hormone), antibodies and fragments thereof, hormones (e.g. GLP-1 ), toxins (e.g. Pseudomonas aeruginosa exotoxin A) and any possible proteinaceous drug. Examples of non-proteinaceous bioactive compounds are, toxins (e.g. DM1 from ImmunoGen), small molecules targeting GPCRs, antibiotics and any possible non-proteinaceous drug.

The term "binding domain" means a protein domain exhibiting the same "fold" (three- dimensional arrangement) as a protein scaffold and having a predetermined property, as defined below. Such a binding domain may be obtained by rational, or most commonly, combinatorial protein engineering techniques, skills which are known in the art (Binz et al., 2005, loc. cit.). For example, a binding domain having a predetermined property can be obtained by a method comprising the steps of (a) providing a diverse collection of protein domains exhibiting the same fold as a protein scaffold as defined further below; and (b) screening said diverse collection and/or selecting from said diverse collection to obtain at least one protein domain having said predetermined property. The diverse collection of protein domains may be provided by several methods in accordance with the screening and/or selection system being used, and may comprise the use of methods well known to the person skilled in the art, such as phage display or ribosome display. Preferably, said binding domain is a recombinant binding domain.

The term "protein scaffold" means a protein with exposed surface areas in which amino acid insertions, substitutions or deletions are highly tolerable. Examples of protein scaffolds that can be used to generate binding domains of the present invention are antibodies or fragments thereof such as single-chain Fv or Fab fragments, protein A from Staphylococcus aureus, the bilin binding protein from Pieris brassicae or other lipocalins, ankyrin repeat proteins or other repeat proteins, and human fibronectin. Protein scaffolds are known to the person skilled in the art (Binz et al., 2005, loc. cit.; Binz et al., 2004, loc. cit.). The term "target" refers to an individual molecule such as a nucleic acid molecule, a polypeptide or protein, a carbohydrate, or any other naturally occurring molecule, including any part of such individual molecule, or complexes of two or more of such molecules. The target may be a whole cell or a tissue sample, or it may be any non- natural molecule or moiety. Preferably, the target is a naturally occurring or non-natural polypeptide or a polypeptide containing chemical modifications, for example modified by natural or non-natural phosphorylation, acetylation, or methylation. In the particular application of the present invention, the target is HGF.

The term "predetermined property" refers to a property such as binding to a target, blocking of a target, activation of a target-mediated reaction, enzymatic activity, and related further properties. Depending on the type of desired property, one of ordinary skill will be able to identify format and necessary steps for performing screening and/or selection of a binding domain with the desired property. Preferably, said predetermined property is binding to a target.

The definitions hereinafter for repeat proteins are based on those in patent application WO 2002/020565. Patent application WO 2002/020565 further contains a general description of repeat protein features, techniques and applications. The term "repeat proteins" refers to a protein comprising one or more repeat domains. Preferably, each of said repeat proteins comprises up to four repeat domains. More preferably, each of said repeat proteins comprises up to two repeat domains. Most preferably, each of the repeat proteins comprises only one repeat domain. Furthermore, said repeat protein may comprise additional non-repeat protein domains, polypeptide tags and/or polypeptide linkers.

The term "repeat domain" refers to a protein domain comprising two or more consecutive repeat units (modules) as structural units, wherein said structural units have the same fold, and stack tightly to create a superhelical structure having a joint hydrophobic core. Preferably, a repeat domain further comprises an N-terminal and/or a C-terminal capping unit (or module). Even more preferably, said N-terminal and/or C-terminal capping units (or modules) are capping repeats.

The term "designed repeat protein" and "designed repeat domain" refer to a repeat protein or repeat domain, respectively, obtained as the result of the inventive procedure explained in patent application WO 2002/020565. Designed repeat proteins and designed repeat domains are synthetic and not from nature. They are man-made proteins or domains, respectively, obtained by expression of correspondingly designed nucleic acids.

Preferably, the expression is done in eukaryotic or prokaryotic cells, such as bacterial cells, or by using a cell-free in vitro expression system. Accordingly, a designed ankyrin repeat protein (i.e. a DARPin) corresponds to a recombinant binding protein of the invention comprising at least one ankyrin repeat domain.

The term "structural unit" refers to a locally ordered part of a polypeptide, formed by three- dimensional interactions between two or more segments of secondary structure that are near one another along the polypeptide chain. Such a structural unit exhibits a structural motif. The term "structural motif" refers to a three-dimensional arrangement of secondary structure elements present in at least one structural unit. Structural motifs are well known to the person skilled in the art. Structural units alone are not able to acquire a defined three-dimensional arrangement; however, their consecutive arrangement, for example as repeat modules in a repeat domain, leads to a mutual stabilization of neighboring units resulting in a superhelical structure.

The term "repeat unit" refers to amino acid sequences comprising repeat sequence motifs of one or more naturally occurring repeat proteins, wherein said "repeat units" are found in multiple copies, and which exhibit a defined folding topology common to all said motifs determining the fold of the protein. Such repeat units correspond to the "repeating structural units (repeats)" of repeat proteins as described by Forrer et al., 2003, loc. cit. or the "consecutive homologous structural units (repeats)" of repeat proteins as described by Binz et al, 2004, loc. cit.. Such repeat units comprise framework residues and interaction residues. Examples of such repeat units are armadillo repeat units, leucine-rich repeat units, ankyrin repeat units, tetratricopeptide repeat units, HEAT repeat units, and leucine- rich variant repeat units. Naturally occurring proteins containing two or more such repeat units are referred to as "naturally occurring repeat proteins". The amino acid sequences of the individual repeat units of a repeat protein may have a significant number of mutations, substitutions, additions and/or deletions when compared to each other, while still substantially retaining the general pattern, or motif, of the repeat units.

Accordingly, the term "ankyrin repeat unit" shall mean a repeat unit, which is an ankyrin repeat as described, for example, by Forrer et al., 2003, loc. cit.. Ankyrin repeats are well known to the person skilled in the art. The term "ankyrin repeat domain" refers to a repeat domain comprising two or more consecutive ankyrin repeat units (modules) as structural units, and, preferably, an N-terminal and/or a C-terminal capping unit (or module). The term "framework residues" relates to amino acid residues of the repeat units, or the corresponding amino acid residues of the repeat modules, which contribute to the folding topology, i.e. which contribute to the fold of said repeat unit (or module) or which contribute to the interaction with a neighboring unit (or module). Such contribution might be the interaction with other residues in the repeat unit (or module), or the influence on the polypeptide backbone conformation as found in a-helices or β-sheets, or amino acid stretches forming linear polypeptides or loops.

The term "target interaction residues" refers to amino acid residues of the repeat units, or the corresponding amino acid residues of the repeat modules, which contribute to the interaction with target substances. Such contribution might be the direct interaction with the target substances, or the influence on other directly interacting residues, e.g. by stabilizing the conformation of the polypeptide of a repeat unit (or module) to allow or enhance the interaction of directly interacting residues with said target. Such framework and target interaction residues may be identified by analysis of the structural data obtained by physicochemical methods, such as X-ray crystallography, NMR and/or CD spectroscopy, or by comparison with known and related structural information well known to practitioners in structural biology and/or bioinformatics.

Preferably, the repeat units used for the deduction of a repeat sequence motif are homologous repeat units, wherein the repeat units comprise the same structural motif and wherein more than 70% of the framework residues of said repeat units are homologous to each other. Preferably, more than 80% of the framework residues of said repeat units are homologous. Most preferably, more than 90% of the framework residues of said repeat units are homologous. Computer programs to determine the percentage of homology between polypeptides, such as Fasta, Blast or Gap, are known to the person skilled in the art. Further preferably, the repeat units used for the deduction of a repeat sequence motif are homologous repeat units obtained from repeat domains selected on a defined target.

The term "repeat sequence motif" refers to an amino acid sequence, which is deduced from one or more repeat units or repeat modules. Preferably, said repeat units or repeat modules are from repeat domains having binding specificity for the same target. Such repeat sequence motifs comprise framework residue positions and target interaction residue positions. Said framework residue positions correspond to the positions of framework residues of the repeat units (or modules). Typically, positions 2, 5, 7-13, and 16-33 of an ankyrin repeat contain framework residues. The numbering of positions of ankyrin repeats is shown in Figure 7. Likewise, said target interaction residue positions correspond to the positions of target interaction residues of the repeat units (or modules). Typically, positions 1 , 3, 4, 6, 14, and 15 of an ankyrin repeat potentially contain target interaction residues. Repeat sequence motifs comprise fixed positions and randomized positions. The term "fixed position" refers to an amino acid position in a repeat sequence motif, wherein said position is set to a particular amino acid. Most often, such fixed positions correspond to the positions of framework residues and/or the positions of target interaction residues that are specific for a certain target. The term "randomized position" refers to an amino acid position in a repeat sequence motif, wherein two or more amino acids are allowed at said amino acid position, for example, wherein any of the usual twenty naturally occurring amino acids are allowed, or wherein most of the twenty naturally occurring amino acids are allowed, such as amino acids other than cysteine, or amino acids other than glycine, cysteine and proline. Most often, such randomized positions correspond to the positions of target interaction residues. However, some positions of framework residues may also be randomized. The term "folding topology" refers to the tertiary structure of said repeat units or repeat modules. The folding topology will be determined by stretches of amino acids forming at least parts of a-helices or β-sheets, or amino acid stretches forming linear polypeptides or loops, or any combination of a-helices, β-sheets and/or linear polypeptides/loops. For example, an ankyrin repeat unit/module consists of a β-turn, followed by two antiparallel a- helices and a loop that reaches the turn of the next repeat unit/module.

The term "consecutive" refers to an arrangement, wherein the repeat units or repeat modules are arranged in tandem. In designed repeat proteins, there are at least 2, usually about 2 to 6, in particular at least about 6, frequently 20 or more repeat units (or modules). In most cases, repeat units (or modules) of a repeat domain will exhibit a high degree of sequence identity (same amino acid residues at corresponding positions) or sequence similarity (amino acid residues being different, but having similar physicochemical properties), and some of the amino acid residues might be key residues being strongly conserved. However, a high degree of sequence variability by amino acid insertions and/or deletions, and/or substitutions between the different repeat units (or modules) of a repeat domain may be possible as long as the common folding topology of the repeat units (or modules) is maintained.

Methods for directly determining the folding topology of repeat proteins by physico- chemical means such as X-ray crystallography, NMR or CD spectroscopy, are well known to the practitioner skilled in the art. Methods for identifying and determining repeat units or repeat sequence motifs or for identifying families of related proteins comprising such repeat units or motifs, such as homology searches (BLAST etc.), are well established in the field of bioinformatics, and are well known to the practitioner in the art. The step of refining an initial repeat sequence motif may comprise an iterative process.

The term "repeat modules" refers to the repeated amino acid sequences of the designed repeat domains, which are originally derived from the repeat units of naturally occurring repeat proteins. Each repeat module comprised in a repeat domain is derived from one or more repeat units of the family or subfamily of naturally occurring repeat proteins, e.g. the family of armadillo repeat proteins or ankyrin repeat proteins. Further preferably, each repeat module comprised in a repeat domain comprises a repeat sequence motif deduced from homologous repeat units obtained from repeat domains selected on the same target (e.g. HGF), for example as described in Example 1 . Accordingly, the term "ankyrin repeat module" shall mean a repeat module, which is originally derived from the repeat units of naturally occurring ankyrin repeat proteins. Ankyrin repeat proteins are well known to the person skilled in the art.

"Repeat modules" may comprise positions with amino acid residues present in all copies of corresponding repeat modules ("fixed positions") and positions with differing or "randomized" amino acid residues ("randomized positions").

The term "capping module" refers to a polypeptide fused to the N- or C-terminal repeat module of a repeat domain, wherein said capping module forms tight tertiary interactions (i.e. tertiary structure interactions) with said repeat module thereby providing a cap that shields the hydrophobic core of said repeat module at the side not in contact with the consecutive repeat module from the solvent. Said N- and/or C-terminal capping module may be, or may be derived from, a capping unit or other structural unit found in a naturally occurring repeat protein adjacent to a repeat unit. The term "capping unit" refers to a naturally occurring folded polypeptide, wherein said polypeptide defines a particular structural unit which is N- or C-terminally fused to a repeat unit, wherein said polypeptide forms tight tertiary structure interactions with said repeat unit thereby providing a cap that shields the hydrophobic core of said repeat unit at one side from the solvent. Preferably, capping modules or capping units are capping repeats. The term "capping repeat" refers to a capping module or capping unit having a similar or the same fold as said adjacent repeat unit (or module) and/or sequence similarities to said adjacent repeat unit (or module). Capping modules and capping repeats are described in WO 2002/020565, WO 2012/069655 and by Interlandi et al., 2008 (loc. cit.). Examples of N-terminal ankyrin capping modules (i.e. N-terminal capping repeats) are SEQ ID NO:1 to 3 and examples of ankyrin C-terminal capping modules (i.e. C-terminal capping repeats) are SEQ ID NO:4 to 8.

For example, the N-terminal ankyrin capping module of SEQ ID NO:33 is encoded by the amino acids from position 1 to 32 and the C-terminal capping module of SEQ ID NO:33 is encoded by the amino acids form position 132 to 159.

A recombinant binding protein according to the invention comprises at least one ankyrin repeat domain, wherein said ankyrin repeat domain has binding specificity for mammalian HGF. The term "has binding specificity for a target", "specifically binding to a target" or "target specificity" and the like means that a binding protein or binding domain binds in PBS to a target with a lower dissociation constant than to an unrelated protein such as the E. coli maltose binding protein (MBP). Preferably, the dissociation constant in PBS for the target is at least 10, more preferably at least 10 2 , even more preferably at least 10 3 , or most preferably at least 10 4 times lower than the corresponding dissociation constant for MBP.

Recombinant binding proteins comprising an ankyrin repeat domain with binding specificity for HGF are shown in the Examples. In particular, the invention relates to a recombinant binding protein as defined herein comprising at least one ankyrin repeat domain, wherein said ankyrin repeat domain specifically binds HGF (Hepatocyte Growth Factor).

Such specific binding refers to the selective affinity of the ankyrin repeat domains according to the invention for HGF. All ankyrin repeat domains that bind to HGF, or an epitope thereof, are deemed to specifically bind to that entity. Thus all ankyrin repeat domains that bind to HGF have binding specificity to the latter.

Preferably, said ankyrin repeat domain binds HGF in PBS with a dissociation constant (Kd) below 10 ~7 M; more preferably, below 10 ~8 M, 10 ~9 M or 10 ~10 M, or most preferably below 10 "11 M. Methods to determine dissociation constants of protein-protein interactions, such as surface plasmon resonance (SPR) based technologies (e.g. SPR equilibrium or kinetic analysis) or isothermal titration calorimetry (ITC) are well known to the person skilled in the art. The measured Kd values of a particular protein-protein interaction can vary if measured under different conditions (e.g., salt concentration, pH). Thus, measurements of Kd values are preferably made with standardized solutions of protein and a standardized buffer, such as PBS. Example 2 shows how such IC50 values of such ankyrin repeat inhibiting the inhibiting the binding of HGF to c-Met can be determined and recombinant binding proteins comprising an ankyrin repeat domain binding HGF with a Kd in PBS below 10 ~7 M

Preferred is a recombinant binding protein comprising an ankyrin repeat domain with binding specificity for human HGF.

Further preferred is a recombinant binding protein comprising an ankyrin repeat domain comprising between 70 and 300 amino acids, in particular between 90 and 200 amino acids.

A binding domain of the invention is an ankyrin repeat domain or a designed ankyrin repeat domain, preferably as described in WO 2002/020565. Examples of designed ankyrin repeat domains with binding specificity for HGF are shown in the Examples. In a further embodiment, the invention relates to a recombinant binding protein comprising at least one ankyrin repeat domain with binding specificity for a mammalian HGF, wherein the ankyrin repeat domain inhibits the binding of HGF to c-Met in PBS with an IC50 value below 10 7 M. Preferably, said ankyrin repeat domain inhibits the binding of HGF to c-Met in PBS with an IC50 value below 10 8 M, more preferably below 10 9 M, 10 10 M, or most preferably below 10 ~11 M.

The half maximal inhibitory concentration (IC50) is a measure of the effectiveness of a compound, such as a binding domain of the invention, in inhibiting a biological, biochemical or biophysical function. Methods to determine IC50 values of inhibition of protein-protein interactions, such as competition ELISAs are well known to the person skilled in the art. The measured IC50 values of a particular inhibitor of a protein-protein interaction can vary if measured under different conditions (e.g., salt concentration, pH). Thus, measurements of IC 5 o values are preferably made with standardized solutions of protein and a standardized buffer, such as PBS. Example 5 shows how suchJCso values of such ankyrin repeat inhibiting the inhibiting the binding of HGF to c-Met can be determined and recombinant binding proteins comprising an ankyrin repeat domain inhibiting the binding of HGF to c-Met in PBS with an IC50 value below 10 7 M. In a further embodiment, the invention relates to a recombinant binding protein comprising at least one ankyrin repeat domain with binding specificity for HGF, which inhibits the HGF stimulated phosphorylation of c-Met in A549 cells (ATCC, cat number: CCL-185). A549 cells were starved for 24 hours and stimulated with HGF in the presence of a titration of an ankyrin repeat domain with specificity for HGF. Assessment of the ability of the compounds of the invention to inhibit HGF stimulated cMet phosphorylation in A549 cells was measured by standard means well known to the person skilled in the art. Example 4 shows how such_IC 5 o values of such ankyrin repeat domains or proteins inhibiting the phosphorylation of c-Met can be determined and shows recombinant binding proteins comprising an ankyrin repeat domain inhibiting the phosphorylation of c-Met in A549 cells with an IC50 value below 10 ~7 M

Preferably, said repeat domain inhibits the HGF stimulated phosphorylation of c-Met in A549 cells with an IC50 value below 10 ~8 M, more preferably below 10 ~9 M, 10 ~10 M, or most preferably 10 ~11 M. In a further embodiment, the invention relates to a recombinant binding protein comprising at least one ankyrin repeat domain with binding specificity for HGF, which inhibits the HGF stimulated proliferation of U87 cells (ATCC, cat number: HTB-14) with an IC50 value below 10 6 M. Preferably, said repeat domain inhibits the HGF stimulated proliferation of U87 cells with an IC50 value below 10 7 M, more preferably below 10 8 M, 10 9 M, 10 10 M, or most preferably 10 11 M.

U87 cells are responsive to HGF for growth and as such can be used to measure the functional inhibitory capability of the compounds of the invention. U87 cells are grown in culture medium and seeded in DMEM containing 1 % FBS (assay medium). Cells are cultured for 24h prior to addition of HGF and a titration of the anti-HGF DARPin.

Assessment of the ability of the compounds of the invention to inhibit HGF is determined by the proliferative capacity of the U87 cells as measured by standard measurements well known to the person skilled in the art. Example 4 shows how suchJCso values of such ankyrin repeat proteins inhibiting the proliferation of U87 cells can be determined and recombinant binding proteins comprising an ankyrin repeat domain inhibiting the HGF stimulated proliferation of U87 cells with an IC 5 o value below 10 ~6 M.

The invention relates to a recombinant binding protein comprising at least one ankyrin repeat domain with binding specificity for HGF, wherein said binding protein and/or ankyrin repeat domain has a midpoint denaturation temperature (Tm) above 40°C upon thermal unfolding in PBS and forms less than 5% (w/w) insoluble aggregates at concentrations up to 10 g/L when incubated at 37°C for 1 day in PBS.

The term "PBS" means a phosphate buffered water solution containing 137 mM NaCI, 10 mM phosphate and 2.7 mM KCI and having a pH of 7.4.

Preferably, the recombinant binding protein and/or binding domain has a midpoint denaturation temperature (Tm) above 45°C, more preferably above 50°C, more preferably above 55°C, and most preferably above 60°C upon thermal unfolding in PBS at pH 7.4. A binding protein or a binding domain of the invention possesses a defined secondary and tertiary structure under physiological conditions. Thermal unfolding of such a polypeptide results in a loss of its tertiary and secondary structure, which can be followed, for example, by circular dichroism (CD) measurements. The midpoint denaturation temperature of a binding protein or binding domain upon thermal unfolding corresponds to the temperature at the midpoint of the cooperative transition in physiological buffer upon heat denaturation of said protein or domain by slowly increasing the temperature from 10°C to about 100°C. The determination of a midpoint denaturation temperature upon thermal unfolding is well known to the person skilled in the art. This midpoint denaturation temperature of a binding protein or binding domain upon thermal unfolding is indicative of the thermal stability of said polypeptide.

Also preferred is a recombinant binding protein and/or ankyrin repeat domain forming less than 5% (w/w) insoluble aggregates at concentrations up to 20 g/L, preferably up 40 g/L, more preferably up to 60 g/L, even more preferably up to 80 g/L, and most preferably up to 100 g/L when incubated for over 5 days, preferably over 10 days, more preferably over 20 days, more preferably over 40 days, and most preferably over 100 days at 37°C in PBS. The formation of insoluble aggregates can be detected by the appearance of visual precipitations, gel filtration or dynamic light scattering, which strongly increases upon formation of insoluble aggregates. Insoluble aggregates can be removed from a protein sample by centrifugation at 10Ό00 x g for 10 minutes. Preferably, a recombinant binding protein and/or ankyrin repeat domain forms less than 2%, more preferably less than 1 %, 0.5%, 0.2%, 0.1 %, or most preferably less than 0.05% (w/w) insoluble aggregates under the mentioned incubation conditions at 37°C in PBS. Percentages of insoluble aggregates can be determined by separation of the insoluble aggregates from soluble protein, followed by determination of the protein amounts in the soluble and insoluble fraction by standard quantification methods.

Also preferred is a recombinant binding protein and/or ankyrin repeat domain that does not lose its native three-dimensional structure upon incubation in PBS containing 100 mM dithiothreitol (DTT) for 1 or 10 hours at 37°C. In one particular embodiment the invention relates to a recombinant binding protein comprising an ankyrin repeat domain, specifically binding to HGF and having the indicated or preferred midpoint denaturation temperature and non-aggregating properties as defined above. In a further embodiment, the invention relates to a recombinant binding protein comprising at least one ankyrin repeat domain with binding specificity for a mammalian HGF, wherein the ankyrin repeat domain competes for binding to a mammalian HGF with an ankyrin repeat domain selected from the group consisting of SEQ ID NOs: 33 to 61 ; preferably SEQ ID NOs: 33, 37, 41 , 42, 43, 48, 51 , 52, 57, 58, 59, 60 and 61 ; more preferably, 37, 42, 43, 51 , 52 and 60; even more preferably, 37, 43, 51 and 60, in particular SEQ ID NO:51 and 60.

Also preferably said ankyrin repeat domain competes for binding to a mammalian HGF with a binding protein selected from the group of DARPins #33 to 61 . Preferably, said repeat domain competes for binding to a mammalian HGF with a binding protein from the group of DARPins #24, 45 and 50. More preferably, said ankyrin repeat domain competes for binding to a mammalian HGF with binding protein DARPin #24 or 50.

The term "compete for binding" means the inability of two different binding domains of the invention to bind simultaneously to the same target, while both are able to bind the same target individually. Thus, such two binding domains compete for binding to said target. Preferably, said two competing binding domains bind to an overlapping or the same binding epitope on said target. Methods, such as competition Enzyme-Linked Immuno Sorbent Assay (ELISA) or competition SPR measurements (e.g. by using the Proteon instrument from BioRad), to determine if two binding domains compete for binding to a target, are well known to the practitioner in the art. An example of such an competition SPR measurement is shown in Example 2.

In a further embodiment, the invention relates to a recombinant binding protein comprising at least one ankyrin repeat domain with binding specificity for a mammalian HGF, wherein said ankyrin repeat domain comprises an amino acid sequence that has at least 70% amino acid sequence identity with one ankyrin repeat domain selected from the group consisting of SEQ ID NOs: 33 to 61 ,

wherein G at position 1 and/or S at position 2 of said ankyrin repeat domain are optionally missing; and

L at the second last position and/or N at the last position of said ankyrin repeat domain are optionally exchanged by A.

Preferably, such an ankyrin repeat domain in a recombinant binding protein of the invention comprises an amino acid sequence that has at least 70% amino acid sequence identity with one ankyrin repeat domain selected from the group consisting of SEQ ID

NOs: 33, 37, 41 , 42, 43, 48, 51 , 52, 57, 58, 59, 60 and 61 ; more preferably, 37, 42, 43, 51 , 52 and 60; even more preferably, 37, 43, 51 and 60, in particular SEQ ID NO:51 and 60.

Preferably, such an ankyrin repeat domain in a recombinant binding protein of the invention comprises an amino acid sequence with at least 70% amino acid sequence identity with one, two or three ankyrin repeat modules present between the N-terminal and C-terminal capping modules of an ankyrin repeat domain selected from the group consisting of SEQ ID NOs: 33 to 61 . Preferably, such an ankyrin repeat domain or such one, two or three repeat modules comprises an amino acid sequence with at least 70, 71 , 72, 73, 74, 75, 76, 77, 78, 79, 80, 81 , 82, 83, 84, 85, 86, 87, 88, 89, 90, 91 , 92, 93, 94, 95, 96, 97, 98, 99, or 100 % amino acid sequence identity. Preferably, up to 30, 29, 28, 27, 26, 25, 24, 23, 22, 21 , 20, 19, 18, 17, 16, 15, 14, 13, 12, 1 1 , 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1 amino acids in the repeat domains SEQ ID NO:33 to 61 are exchanged by another amino acid. Preferably, when amino acids are exchanged in the capping modules of SEQ ID NO: 1 to 8, the repeat modules of SEQ ID NO:12 to 25 or the repeat domains of SEQ ID NO:33 to 61 , these amino acids are selected from the group consisting of A, D, E, F, H, I, K, L, M, N, Q, R, S, T, V, W and Y; more preferably from the group consisting of A, D, E, H, I, K, L, Q, R, S, T, V, and Y. Also preferably, an amino acid is exchanged by a homologous amino acid; i.e. an amino acid is exchanged by an amino acid having a side chain with similar biophysical properties. For example, the negative charged amino acid D may be replaced by the negative charged amino acid E, or a hydrophobic amino acid such as L may be replaced by A, I or V. The techniques of exchanging an amino acid by another amino acid in a polypeptide are well known to the person skilled in the art.

In a further embodiment, the invention relates to a recombinant binding protein comprising at least one ankyrin repeat domain with binding specificity for a mammalian HGF, wherein said ankyrin repeat domain is selected from the group consisting of SEQ ID NOs: 33 to 61 ,

wherein G at position 1 and/or S at position 2 of said ankyrin repeat domain are optionally missing; and

L at the second last position and/or N at the last position of said ankyrin repeat domain are optionally exchanged by A. Preferably, such an ankyrin repeat domain is selected from the group consisting of SEQ ID NO: 33, 37, 41 , 42, 43, 48, 51 , 52, 57, 58, 59, 60 and 61 ; more preferably, 37, 42, 43, 51 , 52 and 60; even more preferably, 37, 43, 51 and 60, in particular SEQ ID NO:51 and 60.

In a further embodiment, the invention relates to a recombinant binding protein comprising at least one ankyrin repeat domain with binding specificity for a mammalian HGF, wherein said ankyrin repeat domain comprises an ankyrin repeat module having an amino acid sequence selected from the group consisting of SEQ ID NO:12 to 25 and sequences in which up to 9 amino acids in SEQ ID NO:12 to 25 are exchanged by any amino acid. Preferably, such a recombinant binding protein comprises at least an ankyrin repeat module having an amino acid sequence selected from the group consisting of SEQ ID NO: 12 to 25 and sequences in which up to 9 amino acids in SEQ ID NO: 12 to 25 are exchanged by any amino acid, and of which up to 6; preferably, 5; more preferably, 4; more preferably, 3; more preferably, 2; most preferably, 1 of said 9 amino acid exchanges are located in positions 1 , 3, 4, 6, 14, and/or 15 of SEQ ID NOs: 12 to 25.

Preferably, such an ankyrin repeat module of said ankyrin repeat domain is selected from the group consisting of SEQ ID NO: 12, 15, 19, 21 and 24; more preferably, 19 and 21 .

More preferably, such an ankyrin repeat module of said ankyrin repeat domain is selected from the group consisting of SEQ ID NO: 12, 13, 14, 15, 18, 19, 20, 21 , and 22; even more preferably, 12, 15, 18, 19, 20, 21 and 24; more preferably, 18, 19, 20, and 21 . Preferably, up to 8 amino acids in the repeat modules of SEQ ID NO:12 to 25 are exchanged by another amino acid, more preferably up to 7 amino acids, more preferably up to 6 amino acids, more preferably up to 5 amino acids, even more preferably up to 4 amino acids, more preferably up to 3 amino acids, more preferably up to 2 amino acids, and most preferably 1 amino acid.

In a further embodiment, the invention relates to a recombinant binding protein, wherein the ankyrin repeat domain with binding specificity to HGF comprises a repeat module with the ankyrin repeat sequence

KDRYGDTPLHLAADIGHLEIVEVLLKAGADVNA (SEQ ID NO:18) and sequences in which up to 9 amino acids in SEQ ID NO:18 are exchanged by any amino acid and

wherein

K at position 1 is optionally exchanged by Q, H, N, I, T, Y, V, F, D, R, L or S; preferably by

Q, H, N, I, T, Y, V or F; also preferably by Q, H, I, T, Y, V; most preferably by Q or H;

R at position 3 is optionally exchanged by A, T, K, H, N, Y, F, S or D; preferably by A, T, K, H or N; most preferably by A;

Y at position 4 is optionally exchanged by F, W, R, L, S or T; preferably by F or W;

D at position 6 is optionally exchanged by L, A, N, E, S T; preferably by L or A;

D at position 14 is optionally exchanged by S, N, M, T, F; preferably by S or N;

I at position 15 is optionally exchanged by Y, A, N, T, R, V, K, D, L, S; preferably by Y, A, N, T, R or V; most preferably by Y or A; and

E at position 19 is optionally exchanged by K. Preferred is a recombinant binding protein, wherein the ankyrin repeat domain with binding specificity to HGF comprises a repeat module with the ankyrin repeat sequence KDRYGDTPLHLAADIGHLEIVEVLLKAGADVNA (SEQ ID NO:18) and sequences in which up to 9 amino acids in SEQ ID NO:18 are exchanged by any amino acid and

wherein

K at position 1 is optionally exchanged by Q, H, I, V

R at position 3 is optionally exchanged by A, T or N; preferably by A;

Y at position 4 is optionally exchanged by F or W;

D at position 6 is optionally exchanged by N;

D at position 14 is optionally exchanged by S;

I at position 15 is optionally exchanged by Y or A; and

E at position 19 is optionally exchanged by K.

In a further embodiment, the invention relates to a recombinant binding protein, wherein the ankyrin repeat domain with binding specificity to HGF comprises a repeat module with the ankyrin repeat sequence

EDYFGNTPLHLAASYGHLEIVEVLLKAGADVNA (SEQ ID NO:19) and sequences in which up to 9 amino acids in SEQ ID NO:19 are exchanged by any amino acid and wherein

E at position 1 is optionally exchanged by D, H, N, F, A, L, Q, I; preferably by D, H, N; most preferably by D or H;

Y at position 3 is optionally exchanged by W, S, H, V, F, A or D; preferably by W, S, H, V, A or F; most preferably by W or S;

F at position 4 is optionally exchanged by Y, A or W; preferably by Y or A;

N at position 6 is optionally exchanged by D, H, I or M; preferably by D or H; most preferably D;

S at position 14 is optionally exchanged by N or H; and

Y at position 15 is optionally exchanged by M, W, L, T, V, S, A, I or N; preferably by M, W, L, T, A or V; also preferably by M, A or W; most preferably by A.

Preferred is a recombinant binding protein, wherein the ankyrin repeat domain with binding specificity to HGF comprises a repeat module with the ankyrin repeat sequence EDYFGNTPLHLAASYGHLEIVEVLLKAGADVNA (SEQ ID NO:19) and sequences in which up to 9 amino acids in SEQ ID NO:19 are exchanged by any amino acid and wherein E at position 1 is optionally exchanged by D, F, A, or L; most preferably D, or A;

Y at position 3 is optionally exchanged by W, S, or V;

F at position 4 is optionally exchanged by Y;

N at position 6 is optionally exchanged by D;

Y at position 15 is optionally exchanged by M, L, S, or A; preferably M, A, or L; most preferably A, or L.

Preferred is a recombinant binding protein, wherein said ankyrin repeat domain comprises said ankyrin repeat module of SEQ ID NO:18 and said ankyrin repeat module of SEQ ID NO:19. Preferably, said ankyrin repeat module of SEQ ID NO:19 directly follows said ankyrin repeat module of SEQ ID NO:18 in said ankyrin repeat domain. For example, in the ankyrin repeat domain of DARPin #51 said ankyrin repeat module of SEQ ID NO:19 directly follows said ankyrin repeat module of SEQ ID NO:18.

In yet another embodiment, the invention relates to a recombinant binding protein, wherein the ankyrin repeat domain with binding specificity to HGF comprises a repeat module with the ankyrin repeat sequence

KDDYGNTPLHLAANTGHLEIVEVLLKAGADVNA (SEQ ID NO:20) and sequences in which up to 9 amino acids in SEQ ID NO:20 are exchanged by any amino acid and wherein

K at position 1 is optionally exchanged by M, I, D, L, Q, V, R, T or H; preferably by M, I or D; most preferably by I or D;

D at position 3 is optionally exchanged by H, Y, T, W or F; preferably by H or Y;

Y at position 4 is optionally exchanged by S, N, A, G, F or R; preferably by S, N, A or G; most preferably by S;

N at position 6 is optionally exchanged by S, T, L or Y; preferably by S;

N at position 14 is optionally exchanged by L, F, M or I; preferably by L or I;

T at position 15 is optionally exchanged by S, W, V, E, F or A; preferably by S;

H at position 17 is optionally exchanged by R; and

E at position 19 is optionally exchanged by K.

Preferred is a recombinant binding protein, wherein the ankyrin repeat domain with binding specificity to HGF comprises a repeat module with the ankyrin repeat sequence KDDYGNTPLHLAANTGHLEIVEVLLKAGADVNA (SEQ ID NO:20) and sequences in which up to 9 amino acids in SEQ ID NO:20 are exchanged by any amino acid and wherein

K at position 1 is optionally exchanged by M, D, L, Q, or V;

D at position 3 is optionally exchanged by Y, or T;

Y at position 4 is optionally exchanged A, or N;

T at position 15 is optionally exchanged by S;

H at position 17 is optionally exchanged by R; and

E at position 19 is optionally exchanged by K.

Also preferred is a recombinant binding protein, wherein said ankyrin repeat domain comprises said ankyrin repeat module of SEQ ID NO:19 and said ankyrin repeat module of SEQ ID NO:20. Preferably, said ankyrin repeat module of SEQ ID NO:20 directly follows said ankyrin repeat module of SEQ ID NO:19 in said ankyrin repeat domain. For example, in the ankyrin repeat domain of DARPin #51 said ankyrin repeat module of SEQ ID NO:20 directly follows said ankyrin repeat module of SEQ ID NO:19.

Also preferred is a recombinant binding protein, wherein said ankyrin repeat domain comprises said ankyrin repeat module of SEQ ID NO:18, said ankyrin repeat module of SEQ ID NO:19 and said ankyrin repeat module of SEQ ID NO:20. Preferably, said ankyrin repeat module of SEQ ID NO:19 directly follows said ankyrin repeat module of SEQ ID NO:18 in said ankyrin repeat domain and said ankyrin repeat module of SEQ ID NO:20 directly follows said ankyrin repeat module of SEQ ID NO:19 in said ankyrin repeat domain. . For example, in the ankyrin repeat domain of DARPin #51 said ankyrin repeat module of SEQ ID NO:20 directly follows said ankyrin repeat module of SEQ ID NO:19, which directly follows said ankyrin repeat module of SEQ ID NO:18..

In yet another embodiment, the invention relates to a recombinant binding protein, wherein the ankyrin repeat domain with binding specificity to HGF comprises a repeat module with the ankyrin repeat sequence

HDYSGFTPLHLAAYYGHLEIVEVLLKHGADVNA (SEQ ID NO:12) and sequences in which up to 9 amino acids in SEQ ID NO:12 are exchanged by any amino acid and wherein

H at position 1 is optionally exchanged by T, S, N, Q, I, K, F, Y or V; preferably by T, S, N, Q or I; most preferably by T or S;

Y at position 3 is optionally exchanged by R, N, D, M, W, E, A, Q or L; preferably by R, N or D; most preferably by R or N;

S at position 4 is optionally exchanged by N, T or W; preferably by N or T; F at position 6 is optionally exchanged by I;

Y at position 14 is optionally exchanged by F;

Y at position 15 is optionally exchanged by W or H; and

I at position 20 is optionally exchanged by V or L.

In yet another embodiment, the invention relates to a recombinant binding protein, wherein the ankyrin repeat domain with binding specificity to HGF comprises a C-terminal capping module with the sequence

FDDWGHTPLHLAARYGHLEIVEVLLKYGADVNA (SEQ ID NO:13) and sequences in which up to 9 amino acids in SEQ ID NO:13 are exchanged by any amino acid and wherein

F at position 1 is optionally exchanged by T, H, L, D, Y, K or E; preferably by T, H, L or D; most preferably by T or H;

D at position 3 is optionally exchanged by R, K, A, N, Y, S, L, T or F; preferably by R, K, A or N; most preferably by A, R or K;

W at position 4 is optionally exchanged by F, Y or R; preferably by F;

H at position 6 is optionally exchanged by L, I, M, N or K; preferably by L or I;

R at position 14 is optionally exchanged by H, Y, S, F, A, N or I; preferably by H, Y or S; most preferably by H; and

Y at position 15 is optionally exchanged by F, L, T, K or R; preferably by F, L or T; most preferably by F.

Preferred is a recombinant binding protein, wherein said ankyrin repeat domain comprises said ankyrin repeat module of SEQ ID NO:12 and said ankyrin repeat module of SEQ ID NO:13. Preferably, said ankyrin repeat module of SEQ ID NO:13 directly follows said ankyrin repeat module of SEQ ID NO:12 in said ankyrin repeat domain. For example, in the ankyrin repeat domain of DARPin #33 said ankyrin repeat module of SEQ ID NO:13 directly follows said ankyrin repeat module of SEQ ID NO:12. In yet another embodiment, the invention relates to a recombinant binding protein, wherein the ankyrin repeat domain with binding specificity to HGF comprises a C-terminal capping module with the sequence

KYEDGLTPLHLAAFYGHLEIVEVLLRHGADVNA (SEQ ID NO:15) and sequences in which up to 9 amino acids in SEQ ID NO:15 are exchanged by any amino acid and wherein

A at position 13 is optionally exchanged by V; and R at position 26 is optionally exchanged by K.

In yet another embodiment, the invention relates to a recombinant binding protein, wherein the ankyrin repeat domain with binding specificity to HGF comprises a C-terminal capping module with the sequence

TDAWGHTPLHLAAYLGHLEIVEVLLKYGADVNA (SEQ ID NO:16) and sequences in which up to 9 amino acids in SEQ ID NO:16 are exchanged by any amino acid and wherein

P at position 8 is optionally exchanged by T;

A at position 12 is optionally exchanged by T;

Y at position 14 is optionally exchanged by S, H or A; preferably by S or H

L at position 15 is optionally exchanged by Y, N or S; preferably by Y or N; and

A at position 33 is optionally exchanged by T. Preferred is a recombinant binding protein, wherein said ankyrin repeat domain comprises said ankyrin repeat module of SEQ ID NO:15 and said ankyrin repeat module of SEQ ID NO:16. Preferably, said ankyrin repeat module of SEQ ID NO:16 directly follows said ankyrin repeat module of SEQ ID NO:15 in said ankyrin repeat domain. For example, in the ankyrin repeat domain of DARPin #41 said ankyrin repeat module of SEQ ID NO:16 directly follows said ankyrin repeat module of SEQ ID NO:15.

In yet another embodiment, the invention relates to a recombinant binding protein, wherein the ankyrin repeat domain with binding specificity to HGF comprises a repeat module with the ankyrin repeat sequence

HDTWGLTPLHLAAFHGHQEIVEVLLKHGADVNA (SEQ ID NO:21 ) and sequences in which up to 9 amino acids in SEQ ID NO:21 are exchanged by any amino acid.

In yet another embodiment, the invention relates to a recombinant binding protein, wherein the ankyrin repeat domain with binding specificity to HGF comprises a repeat module with the ankyrin repeat sequence

QDFYGKTPLHLAALRGHLEIVEVLLKYGADVNA (SEQ ID NO:22) and sequences in which up to 9 amino acids in SEQ ID NO:22 are exchanged by any amino.

Preferred is a recombinant binding protein, wherein said ankyrin repeat domain comprises said ankyrin repeat module of SEQ ID NO:21 and said ankyrin repeat module of SEQ ID NO:22. Preferably, said ankyrin repeat module of SEQ ID NO:22 directly follows said ankyrin repeat module of SEQ ID NO:21 in said ankyrin repeat domain. For example, in the ankyrin repeat domain of DARPin #60 said ankyrin repeat module of SEQ ID NO:22 directly follows said ankyrin repeat module of SEQ ID NO:21 . In yet another embodiment, the invention relates to a recombinant binding protein, wherein the ankyrin repeat domain with binding specificity to HGF comprises a repeat module with the ankyrin repeat sequence

HDYLGLTPLHLAASDGHLEIVEVLLKHGADVNA (SEQ ID NO:23) and sequences in which up to 9 amino acids in SEQ ID NO:23 are exchanged by any amino acid and wherein

H at position 1 is optionally exchanged by N, L, K, R, F, Q or D; preferably by N, L or K; most preferably by N;

Y at position 3 is optionally exchanged by F, Q, T, R, N, S or D; preferably by F, Q or T; most preferably by F or Q;

L at position 4 is optionally exchanged by V, T, Q, Y, D or F; preferably by V or T;

L at position 6 is optionally exchanged by D;

S at position 14 is optionally exchanged by A, N or F; preferably by A or N; and

D at position 15 is optionally exchanged by I, T, S, R, A, Y or M; preferably by I, T, S or R; most preferably I or T.

In yet another embodiment, the invention relates to a recombinant binding protein, wherein the ankyrin repeat domain with binding specificity to HGF comprises a repeat module with the ankyrin repeat sequence

YDYNGLTPLHLAANNGHLEIVEVLLKYGADVNA (SEQ ID NO:24) and sequences in which up to 9 amino acids in SEQ ID NO:24 are exchanged by any amino acid and wherein

Y at position 1 is optionally exchanged by S, I, T, Q, E, M, K, D or V; preferably by S, I or T; most preferably by S or I;

Y at position 3 is optionally exchanged by A, F, V, M, W, T, R or Q; preferably by A, F, V or M; most preferably by A or V;

N at position 4 is optionally exchanged by Y, F, T or W; preferably by Y;

L at position 6 is optionally exchanged by F, H, Y, N or W; preferably by F, H, Y or N; most preferably by F or H;

H at position 10 is optionally exchanged by Y;

A at position 12 is optionally exchanged by T or V;

N at position 14 is optionally exchanged by S; N at position 15 is optionally exchanged by V, M or T; preferably V; and I at position 20 is optionally exchanged by V or L.

In yet another embodiment, the invention relates to a recombinant binding protein, wherein the ankyrin repeat domain with binding specificity to HGF comprises a repeat module with the ankyrin repeat sequence

FD VAG YTP LH LAAYFG H LE I VE VLLKYG AD VN A (SEQ ID NO:25) and sequences in which up to 9 amino acids in SEQ ID NO:25 are exchanged by any amino acid and wherein

F at position 1 is optionally exchanged by I, M, T, D, Y, Q, E, H, A or S; preferably by I, M, T, D, Y or Q; also preferably I, T, D or Y; most preferably by I or Y;

Y at position 3 is optionally exchanged by I, H, S, A, D, or W; preferably I, H or S; most preferably by I or S;

A at position 4 is optionally exchanged by F, Y, V, M, N, L, T, H or I; preferably by F, Y, V or M; most preferably by F or Y;

Y at position 6 is optionally exchanged by F, H, T, W, M, N, Q or S; preferably by F, H, T or W; most preferably by F or T;

Y at position 14 is optionally exchanged by H, M, L, N, I, R, W or T; preferably by H, L, N, I, R or T; most preferably by H or L; and

F at position 15 is optionally exchanged by Y, H, M, T, V, L, N or I; preferably Y, H, T or V; most preferably T or V.

Preferred is a recombinant binding protein, wherein said ankyrin repeat domain comprises said ankyrin repeat module of SEQ ID NO:23 and said ankyrin repeat module of SEQ ID NO:24. Preferably, said ankyrin repeat module of SEQ ID NO:24 directly follows said ankyrin repeat module of SEQ ID NO:23 in said ankyrin repeat domain. For example, in the ankyrin repeat domain of DARPin #57 said ankyrin repeat module of SEQ ID NO:24 directly follows said ankyrin repeat module of SEQ ID NO:23. Also preferred is a recombinant binding protein, wherein said ankyrin repeat domain comprises said ankyrin repeat module of SEQ ID NO:24 and said ankyrin repeat module of SEQ ID NO:25. Preferably, said ankyrin repeat module of SEQ ID NO:25 directly follows said ankyrin repeat module of SEQ ID NO:24 in said ankyrin repeat domain. For example, in the ankyrin repeat domain of DARPin #57 said ankyrin repeat module of SEQ ID NO:25 directly follows said ankyrin repeat module of SEQ ID NO:24. Also preferred is a recombinant binding protein, wherein said ankyrin repeat domain comprises said ankyrin repeat module of SEQ ID NO:23, said ankyrin repeat module of SEQ ID NO:24 and said ankyrin repeat module of SEQ ID NO:25. Preferably, said ankyrin repeat module of SEQ ID NO:24 directly follows said ankyrin repeat module of SEQ ID NO:23 in said ankyrin repeat domain and said ankyrin repeat module of SEQ ID NO:25 directly follows said ankyrin repeat module of SEQ ID NO:24 in said ankyrin repeat domain. For example, in the ankyrin repeat domain of DARPin #57 said ankyrin repeat module of SEQ ID NO:25 directly follows said ankyrin repeat module of SEQ ID NO:24, which directly follows said ankyrin repeat module of SEQ ID NO:23.

Further preferred is a N-terminal or C-terminal ankyrin capping module comprising an N- terminal or C-terminal ankyrin capping repeat, respectively, wherein one or more of the amino acids residues in said capping repeat are replaced by an amino acid residue found at the corresponding position on alignment of a corresponding ankyrin capping unit or ankyrin repeat unit.

The replacement of amino acids can be by any of the 20 most often naturally occurring amino acids, preferably by amino acids selected from the group consisting of A, D, E, F, H, I, K, L, M, N, Q, R, S, T, V, W and Y; and more preferably from the group consisting of A, D, E, H, I, K, L, Q, R, S, T, V, and Y. Also preferably, the replacement of amino acids is by a homologous amino acid; i.e. an amino acid is replaced by an amino acid having a side chain with similar biophysical properties. For example, the negative charged amino acid D may be replaced by the negative charged amino acid E, or a hydrophobic amino acid such as L may be replaced by A, I or V. The replacement of an amino acid by a homologous amino acid is well known to the person skilled in the art.

Also preferred is a C-terminal ankyrin capping module comprising the amino acid A at position 27 and 28 of any of the above C-terminal capping modules based on SEQ ID NO:4 to 8.

Also preferred is a C-terminal capping module comprising the amino acids from position 1 to 26 or from position 1 to 27 of any of the above C-terminal capping modules based on SEQ ID NO:4 to 8. Amino acids G at position 1 and/or S at position 2 of SEQ ID NO:1 to 3 can be removed from N-terminal ankyrin capping modules without any apparent influence on the properties. These two amino acids serve as linkers to connect the ankyrin repeat domain to further amino acids and proteins. The invention also comprises such ankyrin repeat domains comprising N-terminal ankyrin capping modules wherein G at position 1 and/or S at position 2 are removed. It is understood that the amino acid positions (e.g. "position 33") in an ankyrin repeat domain as defined herein are adapted accordingly, resulting in a number shift, e.g. "position 33" will become "position 32", if one amino acid is missing, or "position 33" will become "position 31 ", if two amino acid are missing.

An ankyrin capping module of an ankyrin repeat domain of the invention can be exchanged by an ankyrin capping module by combining techniques, such as alignment of amino acid sequences, mutagenesis and gene synthesis, known to the person skilled in the art. For example, the C-terminal capping repeat of SEQ ID NO:33 can be replaced by the C-terminal capping repeat of SEQ ID NO:8 by (i) determination of the C-terminal capping repeat of SEQ ID NO:33 (i.e. sequence position 132 to 159) by sequence alignment with SEQ ID NO:8, (ii) replacing the sequence of the determined C-terminal capping repeat of SEQ ID NO:33 with the sequence of SEQ ID NO:8, (iii) generation of a gene encoding the repeat domain encoding the exchanged C-terminal capping module, (iv) expressing of the modified repeat domain in the cytoplasm of E. coli and (v) purification of the modified repeat domain by standard means. As a further example, the N-terminal capping repeat of SEQ ID NO:33 can be replaced by the N-terminal capping repeat of SEQ ID NO:2 by (i) determination of the N-terminal capping repeat of SEQ ID NO:33 (i.e. sequence position 1 to 32) by sequence alignment with SEQ ID NO:2, (ii) replacing the sequence of the determined N-terminal capping repeat of SEQ ID NO:33 with the sequence of SEQ ID NO:2, (iii) generation of a gene encoding the repeat domain encoding the exchanged N-terminal capping module, (iv) expressing of the modified repeat domain in the cytoplasm of E. coli and (v) purification of the modified repeat domain by standard means.

Furthermore, an ankyrin repeat domain of the invention can be constructed genetically by assembling a N-terminal ankyrin capping module (e.g. the N-terminal capping repeat of SEQ ID NO:2) followed by one or more repeat modules (e.g. the three ankyrin repeat modules comprising the amino acid residues from position 33 to 131 of SEQ ID NO:33) and a C-terminal capping module (e.g. the C-terminal capping repeat of SEQ ID NO:8) by means of gene synthesis. The genetically assembled repeat domain gene can then be expressed in E. coli as described herein. Further preferred is a recombinant binding protein, repeat domain, repeat module, N- terminal capping module or C-terminal capping module having an amino acid sequence devoid of amino acids C, M or N. Further preferred is a recombinant binding protein, repeat domain, repeat module, N- terminal capping module or C-terminal capping module having an amino acid sequence devoid of amino acid D, E or N followed by G.

Further preferred is a recombinant binding protein or repeat domain comprising any such N-terminal or C-terminal capping module.

In a further preferred embodiment of a recombinant binding protein comprising an ankyrin repeat domain according to the present invention, one or more of the amino acid residues of the N-terminal capping module of said repeat domain is exchanged by an amino acid residue found at the corresponding position on alignment of an N-terminal capping unit. Preferably, up to 30% of the amino acid residues are exchanged, more preferably, up to 20%, and even more preferably, up to 10% of the amino acid residues are exchanged. Most preferably, such an N-terminal capping unit is a naturally occurring N-terminal capping unit.

In a further preferred embodiment of a recombinant binding protein comprising an ankyrin repeat domain according to the present invention, one or more of the amino acid residues of the C-terminal capping module of said repeat domain is exchanged by an amino acid residue found at the corresponding position on alignment of a C-terminal capping unit. Preferably, up to 30% of the amino acid residues are exchanged, more preferably, up to 20%, and even more preferably, up to 10% of the amino acid residues are exchanged. Most preferably, such a C-terminal capping unit is a naturally occurring C-terminal capping unit. In still another particular embodiment, up to 30, 29, 28, 27, 26, 25, 24, 23, 22, 21 , 20, 19, 18, 17, 16, 15, 14, 13, 13, 12, 1 1 , 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1 % of the amino acid residues are exchanged with amino acids which are not found in the corresponding positions of repeat units, N-terminal capping units or C-terminal capping units. The term "consensus sequence" refers to an amino acid sequence, wherein said consensus sequence is obtained by structural and/or sequence aligning of multiple repeat units. Using two or more structural and/or sequence aligned repeat units, and allowing for gaps in the alignment, it is possible to determine the most frequent amino acid residue at each position. The consensus sequence is that sequence which comprises the amino acids which are most frequently represented at each position. In the event that two or more amino acids are represented above-average at a single position, the consensus sequence may include a subset of those amino acids. Said two or more repeat units may be taken from the repeat units comprised in a single repeat protein, or from two or more different repeat proteins. Consensus sequences and methods to determine them are well known to the person skilled in the art.

A "consensus amino acid residue" is the amino acid found at a certain position in a consensus sequence. If two or more, e.g. three, four or five, amino acid residues are found with a similar probability in said two or more repeat units, the consensus amino acid may be one of the most frequently found amino acids or a combination of said two or more amino acid residues.

Further preferred are non-naturally occurring capping modules, repeat modules, binding proteins or binding domains.

The term "non-naturally occurring" means synthetic or not from nature, more specifically, the term means made from the hand of man. The term "non-naturally occurring binding protein" or "non-naturally occurring binding domain" means that said binding protein or said binding domain is synthetic (i.e. produced by chemical synthesis from amino acids) or recombinant and not from nature. "Non-naturally occurring binding protein" or "non- naturally occurring binding domain" is a man-made protein or domain, respectively, obtained by expression of correspondingly designed nucleic acids. Preferably, the expression is done in eukaryotic or bacterial cells, or by using a cell-free in vitro expression system. Further, the term means that the sequence of said binding protein or said binding domain is not present as a non-artificial sequence entry in a sequence database, for example in GenBank, EMBL-Bank or Swiss-Prot. These databases and other similar sequence databases are well known to the person skilled in the art. In one particular embodiment the invention relates to a recombinant binding protein comprising an ankyrin repeat domain specifically binding to HGF and further comprising an ankyrin repeat domain specifically binding to vascular endothelial growth factors A (VEGF-A). Examples of ankyrin repeat domains with specificity for HGF are given herein and examples of ankyrin repeat domains with specificity to VEGF-A are described in WO 2010/060748 or WO 201 1 /135067. Also preferably, such a recombinant binding protein further comprises one ore more, preferably one or two, ankyrin repeat domains with specificity for human serum albumin. Examples of ankyrin repeat domains with specificity to human serum albumin are described in WO 2012/069654. Any such repeat domains with specificities for serum albumin, VEGF-A or HGF can be linked by polypeptide linkers (e.g. SEQ ID NO:10 or 1 1 ) by genetic means by methods known to the person skilled in the art.

Another preferred embodiment is a recombinant binding protein comprising an ankyrin repeat domain with binding specificity for HGF comprising one, two, three or more internal repeat modules that will participate in binding to HGF. Preferably, such an ankyrin repeat domain comprises an N-terminal capping module, two to four internal repeat modules, and a C-terminal capping module. Preferably, said capping modules are capping repeats. Also preferably, said capping modules will participate in binding to HGF.

Further preferred is a recombinant binding protein comprising two or more of said ankyrin repeat domains with binding specificity for HGF. Preferably, said binding protein comprises 2 or 3 of said repeat domains. Said two or more repeat domains have the same or different amino acid sequence.

In a further preferred embodiment of a recombinant binding protein comprising an ankyrin repeat domain according to the present invention, one or more of the amino acid residues of the repeat modules of said ankyrin repeat domain are exchanged by an amino acid residue found at the corresponding position on alignment of a repeat unit. Preferably, up to 30% of the amino acid residues are exchanged, more preferably, up to 20%, and even more preferably, up to 10% of the amino acid residues are exchanged. Most preferably, such a repeat unit is a naturally occurring repeat unit.

In still another particular embodiment, up to 30% of the amino acid residues, more preferably, up to 20%, and even more preferably, up to 10% of the amino acid residues are exchanged with amino acids which are not found in the corresponding positions of repeat units. In further embodiments, any of the recombinant HGF binding proteins or domains described herein may be covalently bound to one or more additional moieties, including, for example, a moiety that binds to a different target to create a dual-specificity binding agent, a bioactive compound, a labeling moiety (e.g. a fluorescent label such as fluorescein, or a radioactive tracer), a moiety that facilitates protein purification (e.g. a small peptide tag, such as a His- or strep-tag), a moiety that provides effector functions for improved therapeutic efficacy (e.g. the Fc part of an antibody to provide antibody- dependent cell-mediated cytotoxicity, a toxic protein moiety such as Pseudomonas aeruginosa exotoxin A (ETA) or a small molecular toxic agent such as maytansinoids or DNA alkylating agents) or a moiety that provides improved pharmacokinetics. Improved pharmacokinetics may be assessed according to the perceived therapeutic need. Often it is desirable to increase bioavailability and/or increase the time between doses, possibly by increasing the time that a protein remains available in the serum after dosing. In some instances, it is desirable to improve the continuity of the serum concentration of the protein over time (e.g., decrease the difference in serum concentration of the protein between the concentration shortly after administration and the concentration shortly before the next administration). Moieties that tend to slow clearance of a protein from the blood include hydroxyethyl starch (HES), polyethylene glycol (PEG), sugars (e.g. sialic acid), well-tolerated protein moieties (e.g. Fc fragments or serum albumin), and binding domains or peptides with specificity and affinity for abundant serum proteins, such as antibody Fc fragments or serum albumin. Examples of such binding domains with affinity for serum albumin are provided in WO 2012/069654. The recombinant binding protein of the invention may be attached to a moiety that reduces the clearance rate of polypeptides in a mammal (e.g. in mouse, rat, or human) by greater than three-fold relative to the unmodified polypeptides.

In a further embodiment, the invention relates to nucleic acid molecules encoding the particular recombinant binding proteins, the particular ankyrin repeat domains, the particular ankyrin repeat modules and the particular capping modules. Further, a vector comprising said nucleic acid molecule is considered.

Further, a pharmaceutical composition comprising one or more of the above mentioned recombinant binding proteins, in particular binding proteins comprising repeat domains, or nucleic acid molecules encoding the particular binding proteins, and optionally a pharmaceutical acceptable carrier and/or diluent is considered. Pharmaceutical acceptable carriers and/or diluents are known to the person skilled in the art and are explained in more detail below. Even further, a diagnostic composition comprising one or more of the above mentioned recombinant binding proteins, in particular binding proteins comprising repeat domains, is considered. A pharmaceutical composition comprises recombinant binding proteins as described above and a pharmaceutically acceptable carrier, excipient or stabilizer, for example as described in Remington's Pharmaceutical Sciences 16 th edition, Osol, A. Ed. [1980]. Suitable carriers, excipients or stabilizers known to the skilled man are saline, Ringer's solution, dextrose solution, Hank's solution, fixed oils, ethyl oleate, 5% dextrose in saline, substances that enhance isotonicity and chemical stability, buffers and preservatives. Other suitable carriers include any carrier that does not itself induce the production of antibodies harmful to the individual receiving the composition such as proteins, polysaccharides, polylactic acids, polyglycolic acids, polymeric amino acids and amino acid copolymers. A pharmaceutical composition may also be a combination formulation, comprising an additional active agent, such as an anti-cancer agent or an anti-angiogenic agent.

The formulations to be used for in vivo administration must be aseptic or sterile. This is readily accomplished by filtration through sterile filtration membranes.

The pharmaceutical composition may be administered by any suitable method within the knowledge of the person skilled in the art.

Further, any of the above mentioned pharmaceutical composition is considered for the treatment of a disorder.

The invention further provides methods of treatment. The method comprises

administering, to a patient in need thereof, a therapeutically effective amount of a recombinant binding protein of the invention.

Further, a method of treating a pathological condition in a mammal including man, comprising administering to a patient in need thereof an effective amount of the above mentioned pharmaceutical composition is considered. Examples of such pathological conditions are atherosclerosis, restenosis, pulmonary hypertension, inflammatory joint diseases, ocular and retinal diseases and fibrotic diseases, including pulmonary fibrosis, liver cirrhosis and other disorders of the liver, scleroderma, glomerulosclerosis and cardiac fibrosis. In addition, anti-HGF therapy is useful for oncology pathological conditions, such as solid and hematological tumors. For example, gliomas, sarcomas, osteogenic sarcomas, multiple myeloma, leukemias, lymphomas and epithelial cancers, including bone metastases.

The recombinant binding protein or ankyrin repeat domain according to the invention may be obtained and/or further evolved by several methods such as display on the surface of bacteriophages (WO 1990/002809, WO 2007/006665) or bacterial cells (WO 1993/ 010214), ribosomal display (WO 1998/048008), display on plasmids (WO 1993/008278) or by using covalent RNA-repeat protein hybrid constructs (WO 2000/032823), or intracellular expression and selection / screening such as by protein complementation assay (WO 1998/341 120). Such methods are known to the person skilled in the art. A library of ankyrin repeat proteins used for the selection/screening of a recombinant binding protein or ankyrin repeat domain according to the invention may be obtained according to protocols known to the person skilled in the art (WO 2002/020565, Binz, H.K., et al., J. Mol. Biol., 332, 489-503, 2003, and Binz et al., 2004, loc. cit). The use of such libraries for the selection of ankyrin repeat domains with specificity for HGF is exemplified in Example 1 . Furthermore, ankyrin repeat domains of the present invention may be modularly assembled from ankyrin repeat modules according to the current invention and appropriate capping modules or capping repeats (Forrer, P., et al., FEBS letters 539, 2-6, 2003) using standard recombinant DNA technologies (e.g.

WO 2002/020565, Binz et al., 2003, loc. cit. and Binz et al., 2004, loc. cit).

The invention is not restricted to the particular embodiments described in the Examples. Other sources may be used and processed following the general outline described below.

Examples

All of the starting materials and reagents disclosed below are known to those skilled in the art, and are available commercially or can be prepared using well-known techniques.

Materials

Chemicals were purchased from Sigma-Aldrich (USA). Oligonucleotides were from Microsynth (Switzerland). Unless stated otherwise, DNA polymerases, restriction enzymes and buffers were from New England Biolabs (USA) or Fermentas (Lithuania). The cloning and protein production strain was E. co// ' XL1 -blue (Stratagene, USA) or BL21 (Novagen, USA). Recombinant human HGF was purchased Peprotech (USA, product number 100- 39), and recombinant mouse HGF was purchased from RnD Systems (USA; product number 2207-HG/CF). Biotinylated HGF was obtained chemically via coupling of the biotin moiety to primary amines of the protein using standard biotinylation reagents and methods (Pierce, USA).

Cell lines were purchased from LGC/ATCC (France/USA; Cat. No: A549 - CCL-185, U87MG -HTB-14). Cell culture media were from Invitrogen / Lubio (Switzerland). Fetal calf serum was from Promocell (Germany; C-37350) Assay reagent for detection of cell proliferation, Cell Proliferation ELISA, BrdU (colorimetric) (Cat. No. 1 164722900) was from Roche, Switzerland. Assay reagents for detection of P-c-Met were from RnDSystems (Human Phospho-HGF R/c-MET DuoSet IC; Cat No. DYC2480-5; Sample Diluent Concentrate 2; DXC002). Human HGF for cellular assays was from Peprotech (Cat No. 100-39) and reconstituted in cell culture medium containing 10% FBS.

Molecular Biology

Unless stated otherwise, methods are performed according to described protocols (Sambrook J., Fritsch E.F. and Maniatis T., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory 1989, New York).

Designed ankyrin repeat protein libraries

Methods to generate designed ankyrin repeat protein libraries are described (WO

2002/020565; Binz et al. 2003, loc. cit.; Binz et al. 2004, loc. cit.). By such methods designed ankyrin repeat protein libraries having randomized ankyrin repeat modules and/or randomized capping modules can be constructed. For example, such libraries could accordingly be assembled based on a fixed N-terminal capping module (e.g. the N- terminal capping module of SEQ ID NO: 2) or a randomized N-terminal capping module according to SEQ ID NO: 29, one or more randomized repeat modules according to the sequence motif of SEQ ID NO: 26, 27 or 28, and a fixed C-terminal capping module (e.g. the C-terminal capping module of SEQ ID NO: 8) or a randomized C-terminal capping module according to SEQ ID NO: 30. Preferably, such libraries are assembled to not have the amino acids C, G, M, N (in front of a G residue) or P at randomized positions of repeat or capping modules. In addition, randomized repeat modules according to the sequence motif of SEQ ID NO: 26, 27 or 28 could be further randomized at position 10 and/or position 17; the randomized N-terminal capping module according to the sequence motif of SEQ ID NO: 29 could be further randomized at position 7 and/or position 9; and the randomized C-terminal capping modules according to the sequence motif of SEQ ID NO: 30 could be further randomized at positions 10, 1 1 and/or 17. Furthermore, such randomized modules in such libraries may comprise additional polypeptide loop insertions with randomized amino acid positions. Examples of such polypeptide loop insertions are complement determining region (CDR) loop libraries of antibodies or de novo generated peptide libraries. For example, such a loop insertion could be designed using the structure of the N-terminal ankyrin repeat domain of human ribonuclease L (Tanaka, N., Nakanishi, M, Kusakabe, Y, Goto, Y., Kitade, Y, Nakamura, K.T., EMBO J. 23(30), 3929-3938, 2004) as guidance. In analogy to this ankyrin repeat domain where ten amino acids are inserted in the beta-turn present close to the boarder of two ankyrin repeats, ankyrin repeat proteins libraries may contain randomized loops (with fixed and randomized positions) of variable length (e.g. 1 to 20 amino acids) inserted in one or more beta-turns of an ankyrin repeat domain.

Any such N-terminal capping module of an ankyrin repeat protein library preferably possesses the RELLKA or RILKAA motif instead of the RILLAA motif (e.g. present from position 21 to 26 in SEQ ID NO:29) and any such C-terminal capping module of an ankyrin repeat protein library preferably possesses the KAA or KLA motif instead of the KLN motif (e.g. the last three amino acids in SEQ ID NO:30).

The design of such an ankyrin repeat protein library may be guided by known structures of an ankyrin repeat domain interacting with a target. Examples of such structures, identified by their Protein Data Bank (PDB) unique accession or identification codes (PDB-IDs), are 1 WDY, 3V31 , 3V30, 3V2X, 3V20, 3UXG, 3TWQ-3TWX, 1 N1 1 , 1 S70 and 2ZGD.

Examples of designed ankyrin repeat protein libraries, such as the N2C and N3C designed ankyrin repeat protein libraries, are described (WO 2002/020565; Binz et al. 2003, loc. cit.; Binz et al. 2004, loc. cit.). The digit in N2C and N3C describes the number of randomized repeat modules present between the N-terminal and C-terminal capping modules.

The nomenclature used to define the positions inside the repeat units and modules is based on Binz et al. 2004, loc. cit. with the modification that borders of the ankyrin repeat modules and ankyrin repeat units are shifted by one amino acid position. For example, position 1 of an ankyrin repeat module of Binz et al. 2004 (loc. cit.) corresponds to position 2 of a ankyrin repeat module of the current disclosure and consequently position 33 of a ankyrin repeat module of Binz et al. 2004, loc. cit. corresponds to position 1 of a following ankyrin repeat module of the current disclosure.

All the DNA sequences were confirmed by sequencing, and the calculated molecular weight of all described proteins was confirmed by mass spectrometry.

Example 1 : Selection of binding proteins comprising an ankyrin repeat domain with binding specificity for HGF

Using ribosome display (Hanes, J. and Pluckthun, A., PNAS 94, 4937-42, 1997) many designed ankyrin repeat proteins (DARPins) with binding specificity for HGF were selected from DARPin libraries as described by Binz et al. 2004 (loc. cit.). The binding of the selected clones toward specific (HGF) and unspecific (MBP, E. coli maltose binding protein) targets was assessed by crude extract ELISA, indicating that hundreds of HGF binding proteins were successfully selected. For example, the ankyrin repeat domains of SEQ ID NO: 33 to 61 constitute amino acid sequences of selected binding proteins comprising an ankyrin repeat domain with binding specificity for HGF. Individual ankyrin repeat modules from such ankyrin repeat domains with binding specificity to HGF are provided in SEQ ID NO: 12 to 25.

Selection of HGF specific ankyrin repeat proteins by ribosome display

The selection of HGF specific ankyrin repeat proteins was performed by ribosome display (Hanes and Pluckthun, loc. cit.) using human and/or mouse HGF as target proteins, libraries of designed ankyrin repeat proteins as described above and established protocols (Zahnd, C, Amstutz, P. and Pluckthun, A., Nat. Methods 4, 69-79, 2007). The number of reverse transcription (RT)-PCR cycles after each selection round was constantly reduced from 45 to 25, adjusting to the yield due to enrichment of binders. The first four rounds of selection employed standard ribosome display selection, using decreasing target concentration and increasing washing stringency to increase selection pressure from round 1 to round 4 (Binz et al. 2004, loc. cit.). To enrich high affinity anti- HGF DARPins, the output from the fourth round of standard ribosome display selection (above) was subjected to one or two off-rate selection rounds with increased selection stringency (Zahnd, 2007, loc. cit.). A final standard selection round was performed after each off-rate selection round to amplify and recover the off-rate selected binding proteins. To increase the incidence of selected binders cross-reactive to human and mouse HGF, an additional selection round was performed on mouse HGF after having done an off-rate selection round on human HGF. Selected clones bind specifically to HGF as shown by crude extract receptor competition ELISA

Individual selected DARPins specifically binding HGF were identified by a receptor competition enzyme-linked immunosorbent assay (ELISA) using crude Escherichia coli extracts of DARPin expression cells using standard protocols. DARPins selected by ribosome display were cloned into the pQE30 (Qiagen) expression vector, transformed into E. coli XL1 -Blue (Stratagene) and then grown overnight at 37°C in a 96-deep-well plate (each clone in a single well) containing 1 .2 ml growth medium (TB containing 1 % glucose and 50 μg/ml ampicillin). 0.9 ml of fresh LB medium containing 50 μg/ml ampicillin was inoculated with 50 μΙ of the overnight culture in a fresh 96-deep-well plate. After incubation for 60-90 minutes at 37°C, expression was induced with IPTG (0.5 mM final concentration) and continued for 3 to 4 hours. Cells were harvested, resuspended in 50 μΙ B-PERII (Pierce) and incubated for 15 min at room temperature with shaking. Then, 950 μΙ PBS was added and cell debris were removed by centrifugation. The extract of each lysed clone was applied as a 1 :50 dilution in PBSTC (PBS supplemented with 0.1 % Tween 20® and 0.25% (w/v) Casein, pH 7.4) together with 1 nM biotinylated human or mouse HGF, to a well of a Protein G coated MaxiSorp plate containing either human or mouse c-Met-Fc fusion and incubated for 10 minutes at RT. After extensive washing with PBS-T (PBS supplemented with 0.1 % Tween 20®, pH 7.4) the plate was developed using standard ELISA procedures and streptavidin-HRP conjugate (1 1 089 153 001 , Roche). Binding was then detected by POD substrate (Roche). The color development was measured at 405 nm. Screening of several hundred clones by such a crude cell extract ELISA revealed more than a hundred different DARPins with specificity for HGF. Examples of amino acid sequences of selected ankyrin repeat domains that specifically bind to human HGF are provided in SEQ ID NO:33 to 61 .

These ankyrin repeat domains with binding specificity for HGF and negative control DARPins with no binding specificity for HGF (i.e. DARPin #28 and #29) were cloned into a pQE (QIAgen, Germany) based expression vector providing an N-terminal His-tag to facilitate simple protein purification as described below. For example, expression vectors encoding the following DARPins were constructed:

DARPin #33 (SEQ ID NO:33 with a His-tag (SEQ ID NO:9) fused to its N terminus); DARPin #34 (SEQ ID NO 34 wi th a H s-tag (SEQ ID NO 9) fused to its N terminus)

DARPin #35 (SEQ ID NO 35 wi th a H s-tag (SEQ ID NO 9) fused to its N terminus)

DARPin #36 (SEQ ID NO 36 wi th a H s-tag (SEQ ID NO 9) fused to its N terminus)

DARPin #37 (SEQ ID NO 37 wi th a H s-tag (SEQ ID NO 9) fused to its N terminus)

DARPin #38 (SEQ ID NO 38 wi th a H s-tag (SEQ ID NO 9) fused to its N terminus)

DARPin #39 (SEQ ID NO 39 wi th a H s-tag (SEQ ID NO 9) fused to its N terminus)

DARPin #40 (SEQ ID NO 40 wi th a H s-tag (SEQ ID NO 9) fused to its N terminus)

DARPin #41 (SEQ ID NO 41 wi th a H s-tag (SEQ ID NO 9) fused to its N terminus)

DARPin #42 (SEQ ID NO 42 wi th a H s-tag (SEQ ID NO 9) fused to its N terminus)

DARPin #43 (SEQ ID NO 43 wi th a H s-tag (SEQ ID NO 9) fused to its N terminus)

DARPin #44 (SEQ ID NO 44 wi th a H s-tag (SEQ ID NO 9) fused to its N terminus)

DARPin #45 (SEQ ID NO 45 wi th a H s-tag (SEQ ID NO 9) fused to its N terminus)

DARPin #46 (SEQ ID NO 46 wi th a H s-tag (SEQ ID NO 9) fused to its N terminus)

DARPin #47 (SEQ ID NO 47 wi th a H s-tag (SEQ ID NO 9) fused to its N terminus)

DARPin #48 (SEQ ID NO 48 wi th a H s-tag (SEQ ID NO 9) fused to its N terminus)

DARPin #49 (SEQ ID NO 49 wi th a H s-tag (SEQ ID NO 9) fused to its N terminus)

DARPin #50 (SEQ ID NO 50 wi th a H s-tag (SEQ ID NO 9) fused to its N terminus)

DARPin #51 (SEQ ID NO 51 wi th a H s-tag (SEQ ID NO 9) fused to its N terminus)

DARPin #52 (SEQ ID NO 52 wi th a H s-tag (SEQ ID NO 9) fused to its N terminus)

DARPin #53 (SEQ ID NO 53 wi th a H s-tag (SEQ ID NO 9) fused to its N terminus)

DARPin #54 (SEQ ID NO .54 wi th a H s-tag (SEQ ID NO .9) fused to its N terminus)

DARPin #55 (SEQ ID NO :55 wi th a H s-tag (SEQ ID NO :9) fused to its N terminus)

DARPin #56 (SEQ ID NO :56 wi th a H s-tag (SEQ ID NO :9) fused to its N terminus)

DARPin #57 (SEQ ID NO :57 wi th a H s-tag (SEQ ID NO :9) fused to its N terminus)

DARPin #58 (SEQ ID NO :58 wi th a H s-tag (SEQ ID NO :9) fused to its N terminus)

DARPin #59 (SEQ ID NO :59 wi th a H s-tag (SEQ ID NO :9) fused to its N terminus)

DARPin #60 (SEQ ID NO :60 wi th a H is-tag (SEQ ID NO :9) fused to its N terminus)

DARPin #61 (SEQ ID NO:61 wi th a H is-tag (SEQ ID NO:9) fused to its N terminus)

DARPin #62 (SEQ ID NO:62 wi th a H i-tag (SEQ ID NO:9) fused to its N terminus;

High level and soluble expression of DARPins

For further analysis, the selected clones showing specific HGF binding in the crude cell extract ELISA as described above were expressed in E. coli BL21 or XL1 -Blue cells and purified using their His-tag using standard protocols. 25 ml of stationary overnight cultures (TB, 1 % glucose, 100 mg/l of ampicillin; 37°C) were used to inoculate 500ml cultures (same medium). At an absorbance of 1 .0 at 600 nm, the cultures were induced with 0.5 mM IPTG and incubated at 37°C for 4-5 h. The cultures were centrifuged and the resulting pellets were resuspended in 40 ml of TBS500 (50 mM Tris-HCI, 500 mM NaCI, pH 8) and sonicated. The lysate was recentrifuged, and glycerol (10% (v/v) final concentration) and imidazole (20 mM final concentration) were added to the resulting supernatant. Proteins were purified over a Ni-nitrilotriacetic acid column (2.5 ml column volume) according to the manufacturer's instructions (QIAgen, Germany). Alternatively, DARPins or selected repeat domains devoid of a 6xHis-tag were purified by anion exchange chromatography followed by size exclusion chromatography according to standard resins and protocols known to the person skilled in the art. Up to 200 mg of highly soluble DARPins with binding specificity to HGF can be purified from one liter of E. coli culture with a purity > 95% as estimated from SDS-15% PAGE. Such purified DARPins are used for further

characterizations.

Example 2: Characterization of the DARPins with binding for specificity for HGF by Surface Plasmon Resonance analysis

Human HGF molecules were directly immobilized in a flow cell and the interaction with various selected DARPins was analyzed. Kd determination by Surface Plasmon Resonance (SPR) analysis

SPR was measured using a ProteOn instrument (BioRad) and measurement was performed according standard procedures known to the person skilled in the art. The running buffer was PBS, pH 7.4, containing 0.005% Tween 20®. Human HGF was covalently immobilized on a GLC chip (BioRad) to a level of about 5000 resonance units (RU). The interaction of DARPin and HGF was then measured by injecting 200 μΙ running buffer (PBS containing 0.005% Tween®) containing serial dilutions of DARPins of concentration of 25, 12.5, 6.26, 3.13 and 1 .67 nM (on-rate measurement), followed by a running buffer flow for 25 minutes at a constant flow rate of 100 μΙ/min (off-rate

measurement). The signals (i.e. resonance unit (RU) values) of the interspots and a reference injection (i.e. injection of running buffer only) were subtracted from the RU traces obtained after injection of DARPin (double-referencing). From the SPR traces obtained from the on-rate and off-rate measurements the on- and off-rate of the corresponding DARPin HGF interaction can be determined. As an example, Figure 1 shows the obtained SPR traces for DARPin #51 . Dissociation constants (Kd) were calculated from the estimated on- and off-rates using standard procedures known to the person skilled in the art. Kd values of selected DARPins were determined to be in the range of 10 pM to 20 nM. As an example, Table 1 summarizes the Kd values for some of the selected DARPins.

Table 1 : Kd values of DARPin human HGF interactions

DARPin # Kd [pM]

DARPin #33 148

DARPin #37 1 1

DARPin #38 1600

DARPin #42 84

DARPin #43 25

DARPin #48 16

DARPin #51 51

DARPin #56 736

DARPin #57 16600

DARPin #58 578

DARPin #59 528

DARPin #60 26

DARPin #61 160

Competition SPR analysis

To check if a binding protein of the invention competes for binding to HGF with an ankyrin repeat domain, competition SPR analysis was carried out by binding a first DARPin to saturation on human HGF and performing a subsequent injection of a second DARPin. When no increase in signal was seen upon injection of the second DARPin, the first and second DARPins were defined to compete for binding to HGF. When an increase in signal was seen, the first and second DARPins were defined to not compete for binding to HGF. An example analysis for each two DARPins competing for binding to HGF and two DARPins non-competing to HGF is given in Figure 2.

Example 3: Inhibition of HGF induced c-Met phosphorylation by DARPins with binding specificity for HGF A549 cells were seeded in complete medium (DMEM; 10% FBS) in 96 well plates at

50.000 cells per well. 24h later, medium was replaced by serum-free medium. Cells were incubated for another 24h and stimulated by 1 nM human HGF in the presence and absence of DARPin. HGF and DARPin were preincubated for at least 30 min at room temperature prior to addition to cells. Cells were stimulated for 10 minutes under cell culture conditions. Stimulation was terminated by removing the cell supernatant (by flicking) and addition of cell lysis buffer (RIPA; Sample Diluent Concentrate 2;

RnDSystems). Phosphorylated c-Met (P-cMet) levels in cell lysates were determined using the Human Phospho-HGF R/c-MET DuoSet IC (RnD Systems) according to the manufacturers' protocol. Data were analyzed using GraphPad Prism software (GraphPad Software Inc., CA, USA). The percentage inhibition of phosphorylation was calculated by setting the OD obtained in the non-stimulated control as 100% inhibition and the control without inhibitor as 0% inhibition. For the inhibition assay, the data were fitted to a log (inhibitor) versus response/variable slope.

Example results are summarized in Table 3. IC 5 o values were calculated from the titration curves obtained as described above using standard procedures known to the person skilled in the art. % inhibition was calculated as described above. Example titration curves are given for DARPin #51 and DARPin #43 in Figure 3.

Table 3: DARPin inhibition potency of HGF stimulated c-Met phosphorylation

DARPin # ICso [nM] % inhibition DARPin #33 0.20 100

DARPin #37 0.07 95

DARPin #38 2.59 88

DARPin #41 0.13 99

DARPin #42 0.32 98

DARPin #43 0.16 101

DARPin #48 0.10 95

DARPin #51 0.01 81

DARPin #56 0.53 72

DARPin #57 1 .72 100

DARPin #58 1 .63 89

DARPin #59 0.85 100

DARPin #60 0.05 94

DARPin #61 0.39 82

Example 4: Inhibition of U87 cell proliferation by DARPins with binding specificity for HGF U87 cells were culture in DMEM containing 10% FBS (Promocell- cytokine -low) at 37°C at 5% C02. Cells were split in a 1 :3 ratio twice a week. 2500 cells per 96 well were seeded in 100 ul DMEM containing 1 % FBS and incubated overnight. The next day

DARPins were added (1 Oul of 10x final concentration) and incubated for another 5 days. At day 4 10ul BrdU labeling solution was added (10 ul 1 :1000 diluted) per well. On day 5 the assay was analyzed according to the manufacturer's protocol. DARPins were titrated in a range of 1 Ό00 nM down to 2 nM. The experiment was analyzed using the GraphPad Prism software. IC50s were calculated from a non-linear fit of the obtained titration curves.

Example results are summarized in Table 4. IC 5 o values were calculated from the titration curves obtained as described above using standard procedures known to the person skilled in the art. Example titration curves are given for DARPin #51 and DARPin #43 in Figure 4.

Table 4: Inhibition of U87 proliferation by DARPins binding HGF

DARPin # IC-50 [nM]

33 137

37 400

41 1 13

42 82

43 71 51 1 16

57 730

59 3'280

60 78

Example 5: Characterization of DARPins with binding specificity for HGFby receptor competition assay

The potency of anti-HGF DARPins to inhibit the binding of human HGF to its receptor c-Met was determined in a receptor competition ELISA. c-Met (Fc chimera; RnD systems, USA, 358-MT-100/CF) was pre-coated on a microplate. DARPins and HGF were premixed in PBSTC and incubated at room temperature for 0.5 hours. These

preincubation mixtures were then transferred into the with c-Met precoated wells and any HGF that was not blocked by the DARPins was bound by the immobilized receptor. After washing away any unbound substances, an polyclonal antibody specific for HGF (RnD systems, USA, AF-294-NA) was added to the wells. After washing away any unbound substances, an horse radish peroxidase conjugated antibody (Novus Biologicals, USA, NB7357) binding to the polyclonal antibody specific for HGF was added to the wells. Following a wash to remove any unbound antibody-enzyme reagent, a substrate solution (Roche, Switzerland, 1 1484281001 ) was added to the wells and color was developing in proportion to the amount of HGF bound. The color development was stopped and the intensity of the color was measured at 405 nm. In this assay, the tested DARPins showed high HGF inhibition potency. Example results are summarized in Table 5. Example titration curves are given for a set of DARPins in Figure 5. IC 5 o values were calculated from such titration curves obtained as described above using standard procedures known to the person skilled in the art.

Table 5: Inhibition of the HGF interaction with its receptor c-Met by DARPins

DARPin # IC-50 [nM]

37 1.33

43 1.36

48 0.91

51 0.62

57 1.71

60 0.95

Example 6: Inhibition of U87 tumor growth by DARPins with binding specificity for HGF 10 7 U87-MG cells were subcutaneously injected into the right flank of Swiss nude mice. 25 days later, when tumors reached an average size of 126 ± 51 mm3 mice were randomly distributed to the treatment groups with 8 mice per group and treatment was started. The mice received seven i.v injections of 0.1 ml of vehicle solution / PBS or various DARPins in PBS (at doses of 0.1 1 mg/kg, 1 .1 mg/kg and 1 1 mg/kg) with a 3-day interval between each dosing (Q3Dx7). Body weight and tumor volume were monitored twice weekly. The experiment was terminated 21 days after treatment start. As an example, Figure 6 shows the tumor volume plotted against the time of treatments given as days after treatment start with PEGylated DARPin #62. The DARPins used for this tumor growth inhibition experiment comprise a N-terminal His-tag (SEQ ID NO:9), an ankyrin repeat domain with binding specificity for HGF according the invention, a GS-linker (SEQ ID NO:10) and a C- terminal Cys residue. The protein moiety of the DARPins was purified by standard means using their His-tag and the coupled to a 40 kDa PEG using standard maleimide chemistry. Other well-known methods (other than PEGylation, see above) to prolong the terminal half-life of the ankyrin repeat domains of the invention could be used to test these domains on their in vivo efficacy in such a tumor model.