专利摘要:
MODIFIED BINDING PROTEINS THATINHIBIT VEIGF-A RECEIVER INTERACTION. The present invention relates to to proteins that specifically bind to VEIGF-A, specifically torecombinant binding proteins containing a cluster ofpolyethylene glycol and a binding domain, which inhibits the binding ofVEGF-Axxx to VEGFR-2. Examples of these recombinant binding proteins include proteins that comprise an ankyrin repeat domain.with the desired binding specificity and a cluster ofpolyethylene glycol. Binding proteins are useful in the treatment ofcancer and other pathological conditions, eg eye diseasessuch as age-related macular degeneration.
公开号:BR112012027368A2
申请号:R112012027368-0
申请日:2011-04-29
公开日:2021-06-29
发明作者:Hans Kaspar Binz;Patrik Forrer;Michael Tobias Stumpp
申请人:Molecular Partners Ag;
IPC主号:
专利说明:

Descriptive Report of the Patent of Invention for "MODIFIED BINDING PROTEINS THAT INHIBIT VEGF-A RECEIVER INTERACTION". Field of the invention The present invention relates to recombinant proteins which specifically bind VEGF-A, as well as pharmaceutical compositions comprising such proteins and the use of these proteins in the treatment of tumors and eye diseases.
Background of the Invention Angiogenesis, the growth of new blood vessels from the pre-existing vasculature, is a fundamental process in several pathological conditions, including the growth of tumors and eye diseases, especially eye diseases causing neovascularization such as macular degeneration related to age (AMD) or diabetic macular edema (DME) (Carmeliet, P., Nature 438, 932-936, 2005). Vascular endothelial growth factors (VEGFs) stimulate angiogenesis and lymphangiogenesis by activating VEGF receptor tyrosine kinases (VEG-FR) in endothelial cells (Ferrara, N., Gerber, H.
P. and LeCouter, J., Nature Med. 9, 669-676, 2003). 20 The mammalian VEGF family consists of five glycoproteins that are called VEGF-A, VEGF-B, VEGF-C, VEGF-D (also known as FIGF) and placental growth factor (PIGF, co- also known as PGF). VEGF-A has been shown to be an effective target for antiangiogenic therapy (Ellis, L.
M. and Hicklin, D.
J., Nature Rev.
Cancer 8, 579-591, 2008). VEGF-A ligands bind to and activate three structurally similar type III receptor tyrosine kinases, designated VEGFR-1 (also known as FLT1), VEGFR-2 (also known as KDR), and VEGFR-3 (also known as FLT1) FLT4). VEGF ligands have different binding specificities for each of these 30 receptor tyrosine kinases, which contribute to their diversity of function.
In response to ligand binding, VEGFR tyrosine kinases activate a network of distinct downward signaling pathways. O
VEGFR-1 and VEGFR-2 are found primarily in the vascular endothelium, whereas VEGFR-3 is mainly present in the lymphatic endothelium.
All of these receptors have an extracellular domain, a unique transmembrane region and a tyrosine kinase consensus sequence interrupted by a domain for kinase insertion.
More recently, neuropilin (NRP-1), originally identified as a receptor for the semaphorin/collapsin family of mediators of neuronal orientation, has been shown to act as a receptor for specific isoforms of VEGF-A.
There are several known isoforms of VEGF-A that are generated by alternative splicing (alternative RNA splicing) from eight exons inserted in the VEGF-A gene.
All isoforms contain exons 1-5 and the terminal exon, exon 8. Exons 6 and 7, which encode heparin-binding domains, may be included or excluded.
This gives rise to a family of proteins named according to their amino acid number: VEGF-A165, VEGF-A121, VEGF-A189, and so on.
Exon 8, however, contains two 3' junction sites in the nucleotide sequences, which can be used by the cell to generate two families of isoforms of identical length but differing in the amino acid sequences of the C- terminal (Varey, AHR et al., British J.
Cancer 98, 1366-20 1379, 2008). VEGF-Axxx ("xxx" indicates the number of amino acids of the mature protein), the pro-angiogenic family of isoforms, is generated using the most proximal sequence in exon 8 (resulting in the inclusion of exon 8a). The most recently described antiangiogenic isoforms, VEGF-Axxxb, are generated by using a distal junction site, 66 bp, further on in gene 25 of the proximal junction site.
This results in the processing and separation of exon 8a and the production of mRNA sequences that encode the VEGF-Axxxb family.
VEGF-A165 is the dominant pro-angiogenic isoform and is commonly overexpressed in a variety of human solid tumors.
The first isoform identified among those encoded by exon 8b was VEGF-A165b, which was shown to have antiangiogenic effects (Varey et al., loc. cit; Konopatskaya, O. et al., Molecular Vision 12, 626- 632, 2006). This endogenous inhibitory form of VEGF-A decreases the proliferation and migration of endothelial cells, induced by VEGF-A.
Although it can bind to VEGFR-2, binding of VEGF-A165b does not result in phosphorylation or activation of receptors in the downward signaling pathways.
There are several strategies to inhibit VEGF-A signaling, 5 including ligand or receptor neutralization by antibody and blocking VEGF-A receptor activation and signaling with tyrosine kinase inhibitors.
VEGF-A-targeted therapy has been shown to be effective as a single agent in AMD, DME, renal cell carcinoma, and hepatocellular carcinoma, whereas it is only beneficial when combined with chemotherapy for patients with metastatic colorectal cancer, breast cancer. non-small cell lung and metastatic breast cancer (Narayanan, R. et al., Nat Rev.
Drug Discovery 5, 815-816, 2005; Ellis and Hicklin, loc. cit). In addition to antibodies, other binding domains can be used to neutralize a ligand or a receptor (Skerra, A., J.
Mol.
Recording 15 13, 167-187, 2000; Binz, H.
K., Amstutz, P. and Plückthun, A., Nat.
Biotechnology. 23, 1257-1268, 2005). One of such new binding domain classes is based on the creation of repeat domains (WO 02/20565; 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). WO 02/20565 describes how large libraries of repeating proteins can be constructed and their general application.
However, the document WO 02/20565 does not describe the selection of repeat domains with binding specificity for VEGF-Axxx, nor does it embody sequence motifs with repeat domains that specifically bind to the VEGF-Axxx.
Targeting VEGF-A with currently available therapeutic agents is not effective in all patients or for all diseases (eg, cancers with EGFR expression). It has even become increasingly evident that the therapeutic benefit associated with VEGF-A-targeted therapy is complex and likely to include multiple mechanisms (Ellis and Hicklin, loc. cit). For example, marketed anti-VEGF drugs such as bevacizumab (Avastin®) or ranibizumab (Lucen).
tis®) (see WO 96/030046, WO 98/045331 and WO 98/045332) or drugs in clinical development such as VEGF-Trap® (WO 00/075319) do not distinguish between pro and anti-angiogenic forms of VEGF-A, so they inhibit both.
As a result, these drugs inhibit angiogenesis, but they also deprive healthy tissues of an essential factor for survival, namely, VEGF-Axxxb, resulting in cytotoxicity and dose-limiting side effects, which in turn instead, they restrict effectiveness.
Common side effects of current anti-VEGF-A therapies are gastrointestinal perforations, bleeding, hypertension, thromboembolic events, and proteinuria (Kamba, T. and McDonald, D.M., Br. J.
Cancer 96, 1788-95, 2007). Another anti-VEGF drug marketed for the treatment of AMD is pegaptanib (WO 98/018480; Macugen®, a registered trademark of Pfizer). Pegaptanib is a PEGylated anti-VEGF aptamer, a single-stranded nucleic acid that specifically binds to the target protein.
For the treatment of neovascular AMD, there is ample evidence that vision results with Lucentis® are superior to those obtained with Macugen®, and there is no definitive evidence to suggest a difference between the drugs in terms of security.
As a result, Macugen® is not a commonly used therapy for this disease. 20 Overall, there is a need to improve anti-angiogenic agents to treat cancer and other pathological conditions.
The technical problem underlying the present invention is to identify new antiangiogenic agents, such as repeat domains with binding specificity for VEGF-Axxx, aiming to improve the treatment of cancer and other pathological conditions, for example, eye diseases such as AMD or DME.
This technical problem is solved by the embodiments characterized in the claims.
Summary of the Invention The present invention relates to a recombinant binding protein, comprising an ankyrin repeat domain and a polyethylene glycol grouping, having a molecular weight of at least 5 kDa, wherein said ankyrin domain binds. to VEGF-Axxx with Kd below
10-9 M and inhibit the binding of VEGF-Axxx to VEGFR-2. In a preferred embodiment, the polyethylene glycol group is coupled to a single Cys residue of the binding domain.
The invention further relates to a pharmaceutical composition comprising one or more of the aforementioned binding proteins or nucleic acid molecules.
The invention further relates to a method of treating cancer and other pathological conditions, for example ocular diseases such as AMD or DME, using the binding proteins of the invention. 10 Brief description of the figures Figure 1. Canine VEGF-A164 specific binding of proteins designed with ankyrin repeats The interaction of selected clones with canine VEGF-A164 (VEGF) and a negative control protein (MBP, mal-binding protein) 15 cough of Escherichia coli) is shown by ELISA analysis of crude extract.
Biotinylated canine VEGF-A164 and MBP were immobilized on NeutrAvidin.
Numbers refer to single DARPin clones selected in ribosomal imaging system against canine VEGF-A164 or the corresponding human VEGF-A165. 20 A = Absorbance.
White bars indicate binding to canine VEGF-A164, black bars show reference non-specific binding to MBP.
Figure 2. Inhibition of spheroid budding by a selected DARPin The extension of shoots in a spheroid budding inhibition assay is shown in the presence of various concentrations of (a) DAR-Pin #30 (amino acids 1 to 126 of SEQ ID NO:4), a DARPin with specificity for VEGF-Axxx, or (b) DARPin NC, a negative control DARPin with no specificity for VEGF-Axxx. 30 Figure 3. Specific recognition of VEGF-A isoforms.
Surface plasmon resonance (SPR) analysis of binding proteins in VEGF-A isoforms
(a) and (b): SPR analysis of Avastin®. Avastin® at 250 nM was applied to a flow cell with canine VEGF-A164 (a) or canine VEGF-A164b (b) immobilized for 100 seconds, followed by washing with flow buffer. 5(c) and (d): SPR analysis of DARPin #27 (amino acids 1 to 159 of SEQ ID NO:1). DARPin no 27 to 250 nM was applied to a flow cell with canine VEGF-A164 (c) or canine VEGF-A164b (d) immobilized for 100 sec, followed by washing with flow buffer.
RU = Resonance Units. 10 Figure 4. Efficient inhibition of human VEGF-A165 in the rabbit eye Rabbit vascular extravasation model to show the efficacy of a DARPin in inhibiting human VEGF-A165 in the eye compared to Lucentis®. On Day 1, PBS, DARPin #30 or Lucentis® is given 15 by intravitreal injection into one eye of each rabbit (treated eye). On Day 4 or Day 30, the two eyes of each rabbit were exposed, by intravitreal injection, to 500 ng of human VEGF-A165.
All eyes were evaluated 48 hours after VEGF-A165 injection by the fluorescein content measured in the vitreous humor and retina of all eyes one hour after intravenous injection of sodium fluorescein.
R = fluorescein ratio measured in treated/untreated eye.
Standard deviations are shown by an error bar. 4-PBS = ratio 4 days after PBS injection (control); 4-D = ratio 4 days after DARPin injection at 30; 30-D = ratio within 30 days after injection of DAR-25 Pin at 30; 4-L = ratio in 4 days after Lucentis® injection; 30-L = ratio within 30 days after Lucentis® injection. Detailed description of the invention Mammalian VEGF-A exists in two families of isoforms resulting from alternative junction: (i) the "VEGF-30 Axxx" pro-angiogenic isoforms generated by proximal junction of exon 8 and (ii) the anti-angiogenic isoforms. genes "VEGF-Axxxb" generated by distal junction of exon 8. Preferably, the binding domain according to the invention is specific for VEGF-
Pro-angiogenic Axxx of canine, rabbit, monkey or human origin.
More preferably, the binding domain according to the invention is specific for pro-angiogenic VEGF-Axxx of human origin.
The most preferred binding domain according to the invention is that specific for human VEGF-A165.
The term "protein" refers to a polypeptide, in which at least part of the polypeptide has or is capable of acquiring a three-dimensional arrangement defined by secondary, tertiary or quaternary structures formed within and/or between its chain(s)( s) polypeptide(s). If a protein comprises two or more polypeptides, the polypeptide chains may be individually linked non-covalently or covalently, for example, by a disulfide bridge between two polypeptides.
A part of a protein that individually has or is able to acquire a three-dimensional arrangement defined by secondary or tertiary structures formed is called a “protein domain”. Such protein domains are well known to the skilled person skilled in the art.
The term "recombinant", as used in recombinant protein, recombinant protein domain and the like, means that said polypeptides are produced by using recombinant DNA technologies, 20 well known to the skilled person skilled in the pertinent subject.
For example, a recombinant DNA molecule (eg, produced by gene synthesis) encoding a polypeptide can be cloned into a bacterial expression plasmid (eg, pQE30, Qiagen). When such a constructed recombinant expression plasmid is inserted into a bacterium (eg, E. coli), this bacterium is able to produce the polypeptide encoded by this recombinant DNA.
The correspondingly produced polypeptide is called recombinant polypeptide.
The term "polypeptide tag" refers to an amino acid sequence joined to a polypeptide/protein, wherein said amino acid sequence is useful for the purification, detection or targeting of said polypeptide/protein, or that said amino acid sequence enhances the physicochemical behavior of the polypeptide/protein, or wherein said amino acid sequence has an effector function.
The tags, groups and/or individual domains of a binding protein can be connected to each other by means of polypeptide linkers.
These polypeptide tags are well known in the state of the art and are fully available to the person skilled in the art.
Examples of polypeptide tags are small polypeptide sequences, for example, His, myc, FLAG or Strep-tags or groups such as enzymes (for example, alkaline phosphatase type enzymes), which enable the detection of said polypeptide/ protein, or groups that can be used for targeting (such as immunoglobulins or their fragments) and/or as effector molecules.
The term "polypeptide linker" refers to an amino acid sequence, which is capable of linking, for example, two protein domains, a polypeptide tag and a protein domain, and a non-polypeptide grouping such as polyethylene glycol or two tags of sequence.
Such domains, tags, non-polypeptide groups and additional linkers are known to the person skilled in the pertinent field.
A list of examples is provided in the descriptive report of patent application WO 02/20565. Specific examples of such linkers are glycine-serine linkers and proline-threonine linkers of 20 variable lengths; preferably, said linkers are between 2 and 24 amino acids in length; more preferably, said linkers are between 2 and 16 amino acids in length.
In the context of the present invention, the term "polypeptide" refers to a molecule consisting of one or more chains of multiples, i.e. two or more amino acids joined by means of peptide bonds.
Preferably, a polypeptide consists of more than eight amino acids joined by peptide bonds.
The term "polymer group" refers to a proteinaceous polymer group or a non-proteinaceous polymer group. A "proteinaceous polymer group" is preferably a polypeptide which does not form a stable tertiary structure while forming no more than 10% (preferably not more than 5%; further preferred not more than 2% ; even more preferably, not more than 1%; and most preferably, no detectable amount, as determined by size exclusion chromatography (SEC)) of oligomers or aggregates when stored at a concentration of approximately 0.1 5 mM in PBS at room temperature (RT) for one month.
In SEC, such groups of proteinaceous polymers run at an apparent molecular weight that is higher than their effective molecular weight when using globular proteins as molecular weight standards for SEC.
Preferably, the apparent molecular weight of said groups of proteinaceous polymers as determined by SEC is 1.5x, 2x or 2.5x higher than their effective molecular weight calculated from their amino acid sequence.
Also preferably, the apparent molecular weights of said groups of non-proteinaceous polymers, determined by SEC, are 2x, 4x or 8x higher than their effective molecular weight calculated from their molecular composition.
Preferably, greater than 50%, 70% or even 90% of the amino acids in said proteinaceous polymer cluster do not form stable secondary structures at a concentration of approximately 0.1 mM in PBS at RT as determined by Circular Dichroism (CD) measurements . More preferably, said proteinaceous polymer shows a typical near UV CD spectrum of a random coil conformation. Such CD analyzes are well known to the person skilled in the art.
Also preferred are groups of proteinaceous polymers comprising more than 50, 100, 200, 300, 400, 500, 600, 700 or 800 amino acids.
Examples of proteinaceous polymer clusters include 25 XTEN® polypeptides (a registered trademark of Amunix; WO 07/103515), or polypeptides comprising proline, alanine and serine residues, as described in WO 08/155134. Such clusters of proteinaceous polymers can be covalently joined, for example, to a binding domain of the invention by the generation of genetic fusion polypeptides, employing conventional DNA cloning technologies, followed by their standard expression and purification.
Examples of binding proteins comprising a repeat domain that binds to VEGF-Axxx and such groups.
proteinaceous polymer products are shown in SEQ ID NO:1 and SEQ ID NO:4. Amino acid positions 1 to 159 of SEQ ID NO:1 correspond to the repeat domain and amino acid position 161 to 1025 of SEQ ID NO:1 correspond to the proteinaceous polymer group.
The 5 amino acid positions 1 to 126 of SEQ ID NO:4 correspond to the repeat domain and amino acid positions 131 to 640 of SEQ ID NO:4 correspond to the proteinaceous polymer group.
The molecular weight of a polymer cluster of the invention can vary widely (i.e., from approximately 1 kDa to approximately 150 kDa). Preferably, the polymer group has a molecular weight of at least 2, 5, 10, 20, 30, 50, 70 or 100 kDa.
Preferably, said polymer group is connected by a polypeptide linker to a binding domain.
Examples of such polypeptide linkers are amino acids 1 to 8 of SEQ ID NO:8 and SEQ ID 15 NO:9. Examples of non-proteinaceous polymer groups are hydroxyethyl starch (HES), polyethylene glycol (PEG), polypropylene glycol or polyoxyalkylene.
The term "PEGylated" means that a PEG moiety is covalently joined to, for example, a polypeptide of the invention.
Examples of repeat proteins containing a polypeptide linker between the repeat domain and a Cys residue at the C-terminus useful for binding to a non-proteinaceous polymer cluster are SEQ ID NO:2, 3, 5, 6 and 7. In a specific embodiment, a PEG group or any other non-proteinaceous polymer can, for example, be coupled to a cysteine thiol via a maleimide linker, the cysteine being coupled via a peptide linker to the N or C -terminal of a binding domain as described in this report (for example, SEQ ID NO:3). The term "binding protein" refers to a protein comprising one or more binding domains and one or more groups of polymers as explained in more detail 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 a single binding domain.
Furthermore, any such binding protein may comprise additional protein domains other than binding domains, multimerization groups, polypeptide tags, polypeptide linkers and/or a single Cys residue.
Examples of multimerization clusters are immunoglobulin heavy chain constant regions that pair to provide functional immunoglobulin Fc domains, and leucine zippers or polypeptides comprising a free thiol that forms an intermolecular disulfide bridge between two such polypeptides.
The single Cys residue can be used to conjugate other groups to the polypeptide, using the chemical reaction involving maleimide well known to the person skilled in the art.
Preferably, said binding protein comprises up to four groups of polymers.
More preferably, said binding protein comprises up to two groups of polymers.
Most preferably, said binding protein comprises only a single polymer group.
Furthermore, preferably, said binding protein has an apparent molecular weight of at least 70, 100, 200, 300, 500 or 800 kDa when analyzed at a concentration of 0.1 mM in PBS at RT by SEC using globular proteins as molecular weight standards.
The term "binding domain" means a protein domain exhibiting the same "fold" (three-dimensional array) as the protein framework and having a predetermined property, as defined below.
Such binding domain can be obtained by rational techniques, or more commonly, combinatorial protein engineering, competences that are known in the state of the art (Skerra, 2000, loc. cit; Binz et al., 2005, loc. cit) . For example, a binding domain having a predetermined property can be obtained by a method including the steps of (a) obtaining a diverse collection of protein domains exhibiting the same fold as protein framework, as defined in more detail. below; and (b) tracking the
from the diverse collection and/or selecting from said diverse collection so as to obtain at least one protein domain endowed with said predetermined property.
The diverse collection of protein domains can be obtained by different methods according to the tracking and/or selection system used, and can comprise the use of methods well known by the technician versed in the field, such as phage visualization or ribosomal visualization.
The term "protein scaffold" means a protein with exposed surface areas in which insertions, substitutions or deletions of 10 amino acids 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 billin binding domain from Pieris brassicae or others lipocalins, ankyrin repeat proteins or proteins with other repeats and human fibronectin.
Protein frameworks are known to the person skilled in the field (Binz et al., 2005, loc. cit.; Binz et al., 2004, loc. cit.). The term "predetermined property" refers to a property such as binding to a target, blocking a target, activating a target-mediated reaction, enzymatic activity, and other related properties.
Depending on the type of property desired, any technician in the field will be able to identify the format and steps necessary to carry out the tracking and/or selection of a link domain with the desired property.
Preferably, said predetermined property is binding to a target.
Preferably, the binding protein of the invention is not an antibody or fragment thereof, such as Fab or scFv fragments.
Antibodies and their fragments are well known to the person skilled in the art.
Additionally, preferably, the binding domain of the invention does not comprise an immunoglobulin fold as present in antibodies and/or in the fibronectin type III domain.
An immunoglobulin fold is a fold common to all proteins that consists of 2 layers.
of the intercalated with approximately 7 antiparallel tapes, arranged in two sheets. Immunoglobulin folds are well known to the person skilled in the art.
For example, such binding domains comprising an immunoglobulin fold are described in WO 07/080392 or WO 508/097497. Still preferably, the binding domain of the invention does not comprise an immunoglobulin-like domain as found in VEGFR-1 or VEGFR-2. Such binding domains are described in document WO 00/075319. 10 A preferred binding domain is one exhibiting anti-angiogenic effects.
The anti-angiogenic effect of a binding domain can be determined by assays well known to the person skilled in the art, such as the HUVEC spheroid budding assay described in Example 2. Further preferred is a binding domain comprising between 15 70 and 300 amino acids, especially between 100 and 200 amino acids.
Still preferred is a binding domain devoid of a free Cys residue.
A Cys-free residue is not involved in the formation of a disulfide bridge.
Even more preferred is a binding domain free of any Cys residue. A preferred binding domain of the invention is a repeat domain or a designed repeat domain, preferably as described in WO 02/20565. An especially preferred residue is a designed ankyrin repeat domain (Binz, H.
K. et al., 2004, loc. cit), preferably 25 as described in WO 02/20565. Examples of designed ankyrin repeat domains are shown in the Examples.
The following definitions for repeating proteins are based on WO 02/20565. Patent Application WO 02/20565 further contains a general description of characteristics, techniques and applications of repeating proteins.
The term "repeat proteins" refers to a protein comprising one or more repeat domains.
Preferably, each of said repeating proteins comprises up to four repeat domains.
More preferably, each of said repeating proteins comprises up to two repeat domains.
Most preferred is that each of the repeating proteins comprise only a single repeat domain.
Furthermore, said repeating protein may comprise non-repeating protein domains, polypeptide tags and/or polypeptide linkers.
The term "repeat domain" refers to a protein domain comprising two or more consecutive repeating units (modules) as structural units, wherein said structural units have the same fold and stack tightly to create, by example, a super-helical structure containing a common hydrophobic core.
The term "designed repeat protein" and "designed repeat domain" refer to a repeating protein or a repeating domain, respectively, obtained as the result of the inventive procedure explained in patent application WO 02/20565 . Designed repeat proteins and designed repeat domains are synthetic and not derived from nature.
These proteins or domains are constructed by man, respectively, and obtained by expression from correspondingly designed nucleic acids.
Preferably, expression is carried out in eukaryotic or prokaryotic cells, such as bacterial cells, or using a cell-free in vitro expression system.
The term "structural unit" refers to a locally ordered portion of a polypeptide, formed by three-dimensional interactions between two or more segments of secondary structure that are close together along the polypeptide chain.
Such a structural unit exhibits a structural motif.
The term "structural motif" refers to a three-dimensional arrangement of elements of the secondary structure, present in at least one structural unit.
Structural motifs are well known to the person skilled in the art.
Structural units alone are not capable of acquiring a definite three-dimensional arrangement, however, their consecutive arrangement, for example, in mo-
repeating duos of a repeating domain, leads to a mutual stabilization of neighboring units resulting in a superhelical structure.
The term "repeat unit" refers to amino acid sequences comprising repeating motifs of sequences from one or more naturally occurring repeating proteins, wherein said "repeat units" are found in multiple copies and in which they exhibit a defined folding topology common to all said motifs, determining the protein folding.
Examples of such repeat units are armadillo repeat units, leucine-rich repeat units, ankyrin repeat units, tetratricopeptide repeat units, HEAT repeat units, and rich variant repeat units. leucine.
Natural proteins containing two or more such repeating units are termed "natural repeating proteins". The amino acid sequences of each repeating unit of a repeating protein can 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 repeating units.
Preferably, the repeat units used for the deduction of a sequence repeat motif are homologous repeat units, obtained from selected repeat domains in a target, for example, as described in Example 1 and having the same specificity for the target.
The term "sequence repeat motif" refers to an amino acid sequence, which is deduced from one or more repeat units.
Preferably, said repeat units come from repeat domains with binding specificity for the same target.
The term "folding topology" refers to the tertiary structure of said repeating units.
The folding topology will be determined by stretches of amino acids forming at least parts of helices or sheets, or stretches of amino acids forming linear polypeptides or loops, or any combination of helices, sheets and/or polypeptides.
neares/loops.
The term "consecutive" refers to an arrangement, in which repeating units or repeating modules are arranged in tandem.
In proteins designed with repeats, there are at least 2, usually around 5, around 2 to 6, especially at least around 6, often 20 or more repeat units.
In most cases, repeating units will exhibit a high degree of sequence identity (the same amino acid residues at corresponding positions) or sequence similarity (amino acid residues being different but having similar physical-chemical properties), and some of the amino acid residues could be essential residues strongly conserved in the different repeating units that are found in natural proteins.
However, a high degree of sequence variability will be possible by amino acid insertions and/or deletions and/or substitutions between the different repeating units that are found in natural proteins as long as the common folding topology is maintained.
Methods to directly determine the repeatable protein folding topology by physicochemical means such as, for example, X-ray crystallography, NMR or CD spectroscopy are well known to the skilled person skilled in the art.
Methods to identify and determine repeating units or motifs with sequence repeat or to identify families of related proteins comprising such repeating units or motifs, such as homology searches (BLAST etc.), are well established in the field of bioinformatics and they are well known by the technician with experience in the subject.
The step of refining an initial motif with sequence repetition can comprise an iterative process.
The term "repeat module" refers to the repeated amino acid sequences of the designed repeat domains, which are originally derived from repeating natural protein repeat units.
Each repeat module comprised in a repeat domain is derived from one or more repeating units of the family or subfamily of naturally-occurring proteins, e.g.
milia of armadillo repeat proteins or ankyrin repeat proteins. "Repeat modules" may comprise positions with amino acid residues present in all copies of corresponding repeat modules ("fixed positions") and positions with different or "random" amino acid residues ("random positions"). The term "capping module" refers to a polypeptide fused to the repeat module at the N or C-terminus of a repeat domain, wherein said capping module forms tight tertiary interactions with said repeat module , thus providing a crown that shields the hydrophobic core of said repeating module on the side that is not in contact with the consecutive repeating module against solvent. Said capping module at the N and/or C-terminus can be derived, a capping unit or other domain found in a natural protein 15 with repeat adjacent to a repeat unit. The term "cropping unit" refers to a naturally-folded polypeptide, wherein said polypeptide defines a specific structural unit that is fused at the N or C-terminus to a repeating unit, wherein said polypeptide forms tight tertiary interactions with said repeating unit, thus providing a crown which shields the hydrophobic core of said repeating unit on one side against solvent. Such capping units have sequence similarities with said motif with sequence repetition. Crowning modules and crowning repetitions are described in WO 02/020565. For example, the capping module of 25 SEQ ID NO:2 at the N-terminus is encoded by amino acids from position 1 to
32. Additionally, it is preferred that such a capping module at the N-terminus has a glycine or aspartate residue at position 5. The term "target" refers to an individual molecule, such as a nucleic acid molecule, polypeptide or protein, carbohydrate or any other natural molecule, including any part of this individual molecule, or complexes formed by two or more such molecules. The target can be an entire molecule or a tissue sample, or it can be any molecule.
cell or unnatural group.
Preferably, the target is a natural or non-natural polypeptide or a polypeptide that contains chemical modifications, for example, modified by phosphorylation, acetylation or methylation, natural or not.
In the specific application of the present invention, the target is VEGF-Axxx or 5 VEGFR-2. The term "consensus sequence" refers to an amino acid sequence, wherein said consensus sequence is obtained by structural and/or multiple repeat unit sequence alignment.
The use of two or more repeating units, structurally and/or sequentially aligned and allowing for gaps in the alignment, makes it possible to determine the most frequent amino acid residue at each position.
A consensus sequence is that sequence which comprises the amino acids that are most frequently represented at each position.
If two or more amino acids have above-average representation at a single position, the consensus sequence may include a subset of these amino acids.
Said two or more repeating units can be taken from repeating units comprised in a single repeating protein or from two or more different repeating proteins. 20 Consensus sequences and methods for determining them are well known to the skilled person.
A "consensus amino acid residue" is the amino acid found at a certain position in a consensus sequence.
If two or more, for example three, four or five amino acid residues are found with similar probability in said two or more repeating 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.
Additionally preferred are binding proteins or unnatural binding domains.
The term "unnatural" means synthetic or non-natural, more specifically it means handmade.
The term
"non-natural binding protein" or "non-natural 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.
The "unnatural binding protein" or the "unnatural binding domain" is a man-made protein or domain, respectively, obtained by the expression of correspondingly designed nucleic acids.
Preferably, expression is carried out in eukaryotic cells or bacterial cells, or using a cell-free in vitro expression system.
Additionally, the term means that the sequence of said binding protein or said binding domain is not present as a non-artificial sequence inserted in a sequence database, for example, in GenBank, EMBL-Bank or in Swiss -Prot.
These databases and other similar string databases are well known to the person skilled in the art. 15 A binding domain can inhibit the binding of VEGF-Axxx to VEGFR-2, either by binding to VEGF-Axxx or by binding to VEGFR-2, in such a way that the apparent dissociation constant (Kd) between VEGF-Axxx and VEGFR-2 is increased by more than 102 times, preferably by more than 103 times, more preferably by more than 104 times, more preferably by 20 by more than 105 times and most preferably by more than 106 times .
Preferably, the Kd for the binding domain interaction to VEGF-Axxx or VEGFR-2 is below 10-7 M, preferably below 10-8 M, more preferably below 10-9 M, more preferably below 10-10M and most preferably below 10-11M.
Methods to determine dissociation constants of protein-protein interactions, such as surface plasmon resonance (SPR) based technologies, are well known to the person skilled in the art.
A preferred binding domain binds to VEGF-Axxx.
Even more preferred is a binding domain that binds to human VEGF-A165. The term "PBS" means phosphate-buffered aqueous solution, containing 137 mM NaCl, 10 mM phosphate and 2.7 mM KCl and having a pH of 7.4. A binding protein and/or binding domain that does not lose its native three-dimensional structure when incubated in PBS containing 100 mM dithiothreitol (DTT) for 1 or 10 hours at 37 °C is preferred.
In a specific embodiment, the invention relates to a binding protein comprising a binding domain which inhibits the binding of VEGF-Axxx to VEGFR-2 and having the mean value of the denaturation temperature and the non-aggregating properties , indicated or preferred as defined above, wherein said binding protein inhibits the budding of HUVEC spheroids with IC50 value below 100 nM.
The term "HUVEC" means human umbilical vein endothelial cells which can be isolated from the normal human umbilical vein and which respond to stimulation by VEGF-A.
Assays for measuring the budding of HUVEC spheroids, such as that described in Example 2, are well known to the person skilled in the art.
IC50 value is the concentration of a substance, such as a binding protein or binding domain, that is required for 50% in vitro inhibition of a given experimental parameter, such as the budding of HUVEC spheroids.
IC50 values can be readily determined by the person skilled in the art (Korff T. and Augustin H.G., J.
Cell Biol. 143(5), 1341-52,1998). Preferred is a binding protein and/or binding domain which inhibits the budding of HUVEC spheroids with IC50 value below 10 nM, preferably below 1 nM, more preferably below 0.1 nM, and most preferably , below 0.05 nM.
Additionally, a binding protein and/or monomeric binding domain that inhibits the budding of HUVEC spheroids with an IC50 value less than the corresponding IC50 value of ranibizumab (Lucentis®, a registered trademark of Genentech) is preferred. bevacizumab (Avastin®, a registered trademark of Genentech), aflibercept (VEGF Trap®, a registered trademark of Regeneran) or pegaptanib (Macugen®, a registered trademark of Pfizer). The Kd for the interaction of a preferred binding domain to VEGF-B, VEGF-C, VEGF-D, PIGF or PDGF is above 1 nM, preferably.
above 10 nM, more preferably above 102 nM, even more preferably above 103 nM, and most preferably above 104 nM.
Preferably, the VEGF-Axxx is dog VEGF-A 164 or simian VEGF-A 165 or human VEGF-A165, and the VEGF-Axxxb is dog VEGF-A 164b 5 or simian or VEGF-A165b human VEGF-A165b.
Another preferred embodiment is a recombinant binding protein, comprising a binding domain, wherein said binding domain inhibits the binding of VEGF-Axxx to VEGFR-2 and wherein said binding domain is a repeat domain or a domain 10 repeat drawn.
Such a repeat domain may comprise one, two, three or more internal repeat modules that will participate in binding to VEGF-Axxx.
Preferably, such a binding domain comprises an N-terminal capping module, two to four internal repeating modules and a C-terminal capping module.
Preferably, said binding domain is an ankyrin repeat domain or a designed ankyrin repeat domain.
A preferred recombinant binding protein comprises a binding domain as described herein, conjugated to a polyethylene glycol (PEG) group, preferably wherein said 20 PEG group is coupled to a single Cys residue of said binding domain.
Preferably, said Cys residue is genetically introduced at the C-terminal end of said binding domain.
The PEG group can then be coupled by chemical means, for example the chemical reaction involving maleimide, well known to the person skilled in the art.
Examples of such binding proteins comprising a PEG group conjugated to a single Cys residue are provided in the Examples.
A preferred embodiment of the invention comprises a recombinant binding protein, containing a binding domain as described herein, wherein said binding domain is conjugated at its C-terminus via a peptide bond to SEQ ID NO:8 , which, in turn, is conjugated at the cysteine thiol at the C-terminus to a PEG coupled to maleimide, such as -[3-(3-maleimido-1-oxopropyl)amino]propyl- -
methoxy-polyoxyethylene (NOF, Sunbright ME-200MA (20kD) or Sunbright ME-400MA (40kD)). In one embodiment, the -[3-(3-maleimido-1-oxopropyl)amino]propyl-methoxy-polyoxyethylene has a molecular weight of at least about 2, 5, 10, 20, 30, 40, 50, 70 or 100 kD.
In certain embodiments, -[3-(3-maleimido-1-oxopropyl)amino]propyl-methoxy-polyoxyethylene has a molecular weight of at least about 20 or at least about 40 kD.
Another preferred embodiment is a recombinant binding protein, as defined above, comprising at least one repeat domain with binding specificity for VEGF-Axxx, wherein said repeat domain competes for binding to VEGF-Axxx with one of the - repeat domain selected from the group consisting of repeat domains of SEQ ID NO:1 to 7. Preferably, said repeat domain competes for binding to VEGF-Axxx with repeat domain of 15 SEQ ID NO :1 or 3. More preferably, said repeat domain competes for binding to VEGF-Axxx with the repeat domain of SEQ ID NO:3. 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 capable of binding to the same target individually.
In this way, these two binding domains are able to bind to said target.
Methods, such as competition ELISA or SPR competition measures (eg, using BioRad's Proteon instrument), to determine whether two binding domains compete for binding to a target are well known to the skilled artisan. - sumptuous.
A recombinant binding protein that competes for binding to VEGF-Axxx with a selected repeat protein can be identified by methods well known to one of ordinary skill in the art, such as the Competitive Enzyme-Linked Immunosorbent Assay (ELISA). Another preferred embodiment is a recombinant binding protein, comprising a repeat domain with binding specificity for VEGF-Axxx, selected from the group consisting of the repeat domains of SEQ ID NO:1 to 7. repeat domain is selected from the repeat domains of SEQ ID NO:2 or 3. More preferably, said repeat domain is the repeat domain of SEQ ID NO:3. One or more groups of polyethylene glycol can be attached at different positions on the binding protein, and such attachment can be achieved by reaction with amines, thiols or other suitable reactive groups.
The joining of polyethylene glycol groups (PEGylation) can be site-directed, in which a suitable reactive group is introduced into the protein to create a site where PEGylation preferably occurs, or is originally present in the binding protein.
The thiol group can be present on a cysteine residue; and the amine group can be, for example, a primary amine found at the N-terminus of the polypeptide or an amine group present on the side chain of an amino acid, such as lysine or arginine.
In a preferred embodiment, the binding protein is modified so as to contain a cysteine residue at a desired position, allowing site-directed PE-Guilation in cysteine, for example, by reaction with a polyethylene glycol derivative bearing a maleimide function.
Polyethylene glycol pool 20 can vary widely in terms of molecular weight (ie, around 1 kDa to around 100 kDa) and can be branched or linear.
Preferably, the polyethylene glycol has a molecular weight of around 1 to around 50 kDa, preferably around 10 to around 40 kDa, even more preferably around 15 to around 30 kDa and most preferable 25 , around 20 kDa.
In a further embodiment, the invention relates to nucleic acid molecules that encode certain recombinant binding proteins.
Furthermore, a vector comprising said nucleic acid molecule is considered. Additionally, a pharmaceutical composition comprising one or more of the binding proteins cited above, especially recombinant binding proteins containing repeat domains, or nucleic acid molecules encoding the certain recombinant binding proteins, and optionally a carrier and/or acceptable pharmaceutical diluent is considered.
Pharmaceutically acceptable carriers and/or diluents are known to the person skilled in the art and are explained in more detail below.
Still further, a diagnostic composition comprising one or more of the above-cited recombinant binding proteins, especially binding proteins containing repeat domains, is envisaged.
The binding protein of the invention suppresses or prevents VEGF-induced pathological angiogenesis, vascular leakage (edema), pulmonary hypertension, tumor formation and/or inflammatory disorders.
"Suppression" is to be understood as meaning that the recombinant protein prevents the cited pathologies to some extent, for example, by 10% or 20%, more preferably by 50%, especially by 70%, 80% or 90% 15 or even at 95%. The term "edema" means a condition that is caused by vascular leakage.
Vasodilation and increased permeability during inflammation may be predominant pathogenic mechanisms.
For example, edema contributes to the expansion of infarction after stroke and can cause life-threatening intracranial hypertension in cancer patients.
Additionally, the extravasation of plasma proteins favors the metastatic spread of hidden tumors, and the airway congestion that occurs during fatal asthma attacks.
Increased vascular leakage, which occurs during inflammation, can lead to breathing difficulties, ascites, sclerosis of the peritoneum (in patients undergoing dialysis), adhesion formation (abdominal surgery) and metastatic spread.
The term "angiogenesis" means a fundamental process by which new blood vessels are formed.
The primary angiogenic period 30 in humans occurs during the first three months of embryonic development, but angiogenesis also occurs as a normal physiological process during periods of tissue growth, such as an increase in muscle or fat tissue during the menstrual cycle and the pregnancy.
The term "pathological angiogenesis" refers to the formation and growth of blood vessels during the maintenance and progression of various disease states.
Specific examples of pathological angiogenesis 5 are found in blood vessels (atherosclerosis, hemangioma, hemangioendothelioma), bones and joints (rheumatoid arthritis, synovitis, bone and cartilage destruction, osteomyelitis, panicle growth, osteophyte formation, neoplasms and metastasis), skin (warts, pyogenic granulomas, capillary growth, Kaposi's sarcoma, scarring keloids, allergic edema, neoplasms), liver, kidney, lung, ear and other epithelia (inflammatory and infectious processes including hepatitis, glomerulonephritis, pneumonia; and asthma, nasal polyps, otitis, transplant disorders, liver regeneration disorders, neoplasms and metastasis), uterus, ovary and placenta (dysfunctional uterine bleeding from intrauterine contraceptive devices, cyst formation follicular, ovarian hyperstimulation syndrome, endometriosis, neoplasms), brain, nerves and eye (retinopathy of prematurity, ret. diabetic inopathy, choroidal and other intraocular disorders, leukomalacia, neoplasms and metastasis), heart and skeletal muscle due to work overload, adipose tissue (obesity), endocrine organs (thyroiditis, increased thyroid volume, trans - lathe resulting from pancreas transplantation), hematopoiesis (Kaposi's syndrome in AIDS), hematological malignancies (leukemias) and lymphatic vessels (tumor metastasis, lymphoproliferative disorders). The term "ischemic retinal disease" means that the retinal blood and oxygen supply is diminished, the peripheral parts of the retina lose their source of nutrition, and stop functioning properly.
A specific example of an ischemic retinal disease is retinopathy.
Common diseases that lead to retinopathy include diabetic retinopathy, central retinal vein occlusion, carotid artery stenosis, and sickle cell retinopathy.
Diabetic retinopathy is an important cause of vision loss in diabetic patients.
In the ischemic retina, new blood vessels grow (neovascularization). Is-
These vessels often grow on the surface of the retina, in the optic nerve, or in the frontal region of the eye in the iris.
The new vessels cannot replenish the flow of necessary nutrients and instead can cause many problems such as hemorrhage in the vitreous humor, retinal detachment and uncontrolled glaucoma.
These problems occur because the new vessels are fragile and prone to bleeding.
If affected in its early stages, proliferative diabetic retinopathy can sometimes be interrupted with panretinal photocoagulation.
However, in some cases, vitrectomy surgery is the only option. 10 In addition to these retinopathies, vascular diseases of the eye also include ocular neovascularization diseases, such as macular degeneration and diabetic macular edema (DME). Macular degeneration results from neovascular growth of the choroid vessel under the macula.
There are two types of macular degeneration: dry and wet.
Although wet macular degeneration comprises only 15% of all cases of macular degeneration, virtually all cases of wet macular degeneration lead to blindness.
Additionally, wet macular degeneration almost always results from dry macular degeneration.
Once one eye is affected by wet macular degeneration, the condition almost always affects the other eye.
Wet macular degeneration is called the age-related wet macular degeneration of wet AMD, as it is mainly found in elderly people.
Diabetic retinopathy (DR) and DME are the main causes of blindness in the working age population of most developed countries. 25 The growing number of people with diabetes around the world suggests that DR and DME will continue to contribute to vision loss and associated functional impairment for years to come.
Several biochemical mechanisms, including activation of protein kinase C- , increased production of vascular endothelial growth factor, oxidative stress, and accumulation of intracellular sorbitol and advanced glycosylation end products, may contribute to the vascular changes that characterize DR/DME.
Inhibition of these pathways is promising for intervention in DR and DME.
The term "pulmonary hypertension" means a disorder in which the blood pressure in the pulmonary arteries is abnormally high.
In the absence of other heart or lung diseases, pulmonary hypertension is called primary.
Diffuse narrowing of the pulmonary arterioles occurs as a result of pathological arteriogenesis, followed by pulmonary hypertension in response to increased resistance to blood flow.
The incidence is 8 in 100,000 people.
However, pulmonary hypertension can also occur as a complication of Chronic Obstructive Pulmonary Diseases (COPD), such as emphysema, chronic bronchitis, or diffuse interstitial fibrosis, and in patients with asthmatiform COPD.
The incidence of COPD is approximately 5 in 10,000 people.
Furthermore, the binding proteins of the invention can be used to treat inflammation and more specifically inflammatory disorders. 15 The term "inflammation" in this descriptive report means the local reaction to injury to living tissue, especially the local reaction of small blood vessels, their contents and their associated structures.
The passage of blood constituents through vessel walls into tissues is the hallmark of inflammation, and the collection of tissue thus formed is called exudates or edema.
Any harmful process that damages living tissue, eg bacterial infection, excessive heat, cold, mechanical injury such as crushing, acids, alkalis, irradiation or virus infection, can cause inflammation regardless of the organ or tissue involved.
It should be clarified that diseases classified as 'inflammatory diseases' and tissue reactions ranging from burns to pneumonia, leprosy, tuberculosis and rheumatoid arthritis are all 'inflammations'. The binding proteins according to the invention can be used to treat tumor formation.
The term "tumor" means a mass of abnormal tissue that arises without evident cause from preexisting body cells, has no purposeful function, and is characterized by a tendency to grow autonomously and without restriction.
Tumors are quite different from inflammatory and other edemas in that the appearance and other features of the tumor cells are abnormal.
Abnormal cells, that is, cells of the type with which tumors are usually formed, differ from normal cells in that they have undergone one or more of the following changes: (1) hypertrophy, or an increase in individual cell size; (2) 5 hyperplasia or increase in the number of cells within a given area; (3) anaplasia, or regression of the physical characteristics of a cell to a more primitive or undifferentiated type.
Tumors can be benign, for example lipomas, angiomas, osteomas, chondromas and adenomas.
Examples of malignant tumors are carcinomas (such as breast tumors, carcinomas in the respiratory and gastrointestinal tract, endocrine glands and genitourinary system), sarcomas (in connective tissues, including fibrous tissue, adipose tissue (fat), muscle, blood vessels, bone and cartilage), carcinosarcoma (in epithelial and connective tissue), leukemias and lymphomas, tumors of nerve tissue (including the brain) and melanoma (a cancer of the pigmented cells of the skin). The use of the binding proteins of the present invention against tumors can also be in combination with other antitumor therapy known in the art such as irradiation, photodynamic therapy, chemotherapy or surgery.
A pharmaceutical composition comprises binding proteins 20 as described above and a pharmaceutically acceptable carrier, excipient or stabilizer (Remington's Pharmaceutical Sciences, 16th edition, Osol, A.
Ed. [1980]). Suitable vehicles, excipients and stabilizers known to the technician in the field are saline solution, Ringer's solution, dextrose solution, Hank's solution, fixed oils, ethyl oleate, dextrose 5% 25 in saline solution, substances that intensify the isotonicity and chemical stability, buffers and preservatives.
Other suitable vehicles which do not themselves 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 can also be a combined formulation, comprising an additional active agent, such as an anti-cancer agent or an anti-angiogenic agent (for example, human VEGF-Axxxb;
reference, human VEGF-A165b). A preferred pharmaceutical composition for the treatment of eye diseases comprises binding proteins as described above and a detergent such as a non-ionic detergent, including but not limited to 5 polysorbate 20 (eg around 0.04%), a buffer such as histidine, phosphate or lactic acid, and a sugar such as sucrose or trehalose.
Preferably such a composition comprises binding proteins as described above and PBS.
Said or any other pharmaceutical compositions described herein may be administered locally, either topically to a part of the eye or injected into the eye, for example, in the subconjunctival, peri or retrobulbar space, or directly into the eye.
Alternatively, said or these other pharmaceutical compositions may be administered systemically by parental administration.
Preferably, said or this other pharmaceutical composition is applied to the eye by intravitreal injection. Further preferably, said pharmaceutical composition is applied to the eye in the form of an eye drop.
The eyedrop can be applied to the cornea (the clear part in the center of the eye), thus enabling the molecules to permeate into the eye.
For the treatment of a disease affecting the posterior region of the eye, it may be desirable for binding proteins to penetrate the sclera when injected under the conjunctiva or around the globe.
The binding protein can be administered after a preliminary eye surface modulation step to improve penetration of the molecules.
Preferably, the epithelial layer, such as the corneal epithelium, is modulated by a penetration enhancer to allow the molecules to penetrate sufficiently and rapidly as, for example, described above.
The use of binding proteins of the present invention against eye diseases can also be in combination with any other therapy known in the art such as photodynamic therapy.
The formulations to be used for in vivo administration 30 must be aseptic or sterile.
This is quickly accomplished by filtration through sterile filtration membranes.
Sustained-release preparations can be produced.
In one embodiment of the invention, an intraocular implant can be used to deliver the binding protein of the invention.
Suitable examples of sustained release preparations include semipermeable matrices of solid hydrophobic polymers containing a polypeptide of the invention, these matrices being in the form of molded items, for example, films or microcapsules.
Examples of sustained-release matrices include polyesters, hydrogels (eg, poly(2-hydroxyethyl-methacrylate) or poly(vinyl alcohol)), polylactides, copolymers of L-glutamic acid and gamma-ethyl-L-glutamate , non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as LUPRON DEPOT® (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), and poly acid -D-(-)-3-hydroxybutyric.
The pharmaceutical composition can be administered by any suitable method within the knowledge of the skilled person.
The preferred route of administration is parenteral.
In parental administration, the medicine of this invention will be formulated in injectable unit dose form such as a solution, suspension or emulsion, associated with the pharmaceutically acceptable excipients as defined above.
The dose and mode of administration will depend on the individual to be treated and the specific disease.
In general, the pharmaceutical composition is administered so that the binding protein of the present invention is provided at a dose between 1 ng/kg and 20 mg/kg, more preferably between 10 ng/kg and 5 mg/kg, most preferably between 0.1 and 2 mg/kg.
Preferably, it is provided as a bolus dose.
Continuous infusion can also be used and includes continuous subcutaneous delivery via an osmotic minipump.
If so, the pharmaceutical composition can be infused at a dose between 5 and 20 ng/kg/minute, more preferably between 7 and 15 ng/kg/minute.
In particular, the pharmaceutical composition is administered by injections into the eye such that the binding protein of the invention is provided at a dose between 0.1 mg and 10 mg per injection, more preferably between 0.3 and 6 mg per injection, most preferably between 1 mg and 4 mg per injection.
Additionally, the pharmaceutical composition is administered by eye drops to the eye, so that a single drop of a solution containing a concentration of the binding protein of the invention between 10 and 120 mg/ml, more preferably between 20 and 100 mg/ml , most preferably between 40 and 80 mg/ml is applied to the eye. In another embodiment of the invention, a binding protein that inhibits the activity of VEGF-Axxx, as described above, can be used in combination with a binding protein or small molecule inhibiting the activity of PIGF, at the same levels inhibition of PIGF than those described above for VEGF-Axxx.
This embodiment is based on the fact that PIGF is shown to be angiogenic in sites where VEGF-Axxx levels are increased.
In addition, a binding protein that inhibits the activity of VEGF-Axxx, as described above, can be used in combination with a binding protein or small molecule inhibitor of platelet-derived growth factor (PDGF) activity. of VEGF-C or 15 of other members of the VEGF protein family, tumor necrosis factor alpha (TNFalpha), delta ligand type 4 (DII4), interleukin 6 (IL-6), neuropilin or angiopoietin 2 (Ang2). The invention further provides methods of treatment.
In one aspect, a method of treating a retinopathy is provided, the method comprising administering to a patient in need thereof a therapeutically effective amount of a binding protein of the invention, specifically a binding protein that inhibits the interaction between Human VEGF-Axxx and human VEGFR-2, but not the interaction between human VEGF-Axxxb and human VEG-FR-2, and the binding protein inhibits VEGFR-2 mediated angiogenesis. The invention further concerns methods of using a binding protein as described to inhibit a biological activity of VEGF-A in a cell or to inhibit a biological activity mediated by VEGFR-2. The cell can be located in vivo or ex vivo and can be, for example, a living organism cell, cultured cell or cell in tissue sample.
The method may comprise contacting said cell with any of the binding proteins described herein that inhibit VEGF-A/VEGFR-2 interaction, in an amount and for a time sufficient to inhibit such biological activity.
The invention provides a method of treating a subject with a condition that responds to inhibition of VEGF-Axxx or VEGFR-2. This method 5 comprises administering to said subject an effective amount of a binding protein described herein.
The condition can be one characterized by inappropriate angiogenesis.
The condition can be a hyperproliferative condition.
Examples of conditions (or disorders) suitable for treatment include autoimmune disorders, inflammatory disorders, retinopathies 10 (especially proliferative retinopathies) and cancers, in particular one of the diseases described above.
Any of the binding proteins described herein can be used to prepare a drug for the treatment of such a disorder, especially a disorder selected from the group consisting of: autoimmune disorder, inflammatory disorder, retinopathy, and cancer.
Preferred conditions (or disorders) suitable for treatment are metastatic renal cell carcinoma in first-line treatment, relapse of glioblastoma multiforme, colon cancer in adjuvant treatment, HER2-negative breast cancer in adjuvant treatment, breast cancer. HER2-positive breast cancer in adjuvant treatment, non-small cell lung cancer in adjuvant treatment, diffuse large B-cell lymphoma, advanced gastric cancer in first-line treatment, HER2-negative metastatic breast cancer in first-line treatment, cancer. HER2-positive metastatic breast cancer in first-line treatment, 25 metastatic ovarian cancer in first-line treatment, gastrointestinal stromal tumors, high-risk carcinoid, hormone-refractory prostate cancer, newly diagnosed glioblastoma multiforme, metastatic head and neck cancer , relapse of platinum-sensitive ovarian cancer, metastatic breast cancer being treated for second line, extensive small cell lung cancer, non-squamous non-small cell lung cancer with previously treated CNS metastases and multiple myeloma relapse, prostate cancer, non-small cell lung cancer.
quena cell (NSCLC), colorectal cancer and pancreatic cancer, advanced ovarian cancer (AOC), patients with COC and symptomatic malignant ascites and non-Hodgkin's lymphoma.
The recombinant binding protein according to the invention can be obtained and/or further developed by various methods such as visualization on the surface of bacteriophages (WO 90/02809, WO 07/006665) or bacterial cells (WO 93/10214) , ribosomal visualization (WO 98/48008), visualization in plasmids (WO 93/08278) or using hybrids constructed with proteins containing 10 covalent RNA repeat (WO 00/32823), or by intracellular expression and selection/screening such as by assay of protein complementation (WO 98/341120). These methods are known to the person skilled in the art.
An ankyrin repeat protein library, used for the selection/screening of a recombinant binding protein according to the invention, can be obtained according to protocols known to the person skilled in the art (WO 02/020565 , Binz, HK et al., JMB, 332, 489-503, 2003, and Binz et al., 2004, loc. cit). The use of this library for the section on DARPins specific for VEGF-Axxx is provided in Example 1. In analogy, the ankyrin repeat sequence motifs as shown above can be used to build libraries of repeating proteins. ankyrin that can be used for the selection or screening of specific DARPins for VEGF-Axxx.
Furthermore, repeat domains of the present invention can be assembled by modules from repeating modules according to the present invention and 25 suitable capping modules (Forrer, P., et al., FEBS letters 539, 2- 6, 2003), using standard recombinant DNA technologies (e.g. WO 02/020565, Binz et al., 2003, loc. cit. and Binz et al., 2004, loc. cit). The invention is not restricted to the specific embodiments described in the Examples.
Other fonts can be used and processed following the general description given below.
Examples All of the starting materials and reagents disclosed below are known to those skilled in the art and are commercially available or can be prepared using well known techniques.
Materials 5 The chemical substances were purchased from Fluka (Switzerland). The oligonucleotides were from Microsynth (Switzerland). Unless otherwise stated, DNA polymerases, restriction enzymes and buffers were from New England Biolabs (USA) or Fermentas (Lithuania). The strain for cloning and protein production was E. coli XL-1-blue (Stratagene, EU-10A). VEGF variants were from R&D Systems (Minneapolis, USA) or were produced in Chinese Hamster Ovary Cells or in Pichia pastoris and purified according to standard protocols (Rennel, E.
S. et al., European J.
Cancer 44, 1883-94, 2008; Pichia expression system from Invitrogen). 15 Biotinylated VEGF variants were chemically obtained by coupling the biotin group to primary amines of purified VEGF variants, using standard biotinylation reagents and methods (Pierce, USA). Molecular Biology 20 Unless otherwise stated, methods were 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). Protein libraries designed with ankyrin repeat The N2C and N3C protein libraries designed with ankyrin repeat are described (WO 02/20565; Binz et al. 2003, loc. cit; Binz et al. 2004, loc. cit. cit. .). The number in N2C and N3C describes the amount of repeating random modules present between the capping modules at the N-terminus and at the C-terminus.
The nomenclature used to define the positions within the repeat units is based on Binz et al. 2004, location cit., with the modification that the boundaries of repeat modules and repeat units are shifted by an amino acid position.
For example, position 1 of a repetition module by Binz et al. 2004 (loc. cit.) corresponds to position 2 of a repeat module of the present invention and, consequently, position 33 of a repeat module of Binz et al. 2004, location cit.
It corresponds to position 1 of a next repeat module of the present invention.
All 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 a repeat domain with binding specificity for VEGF-Axxx Using visualization on ribosomes (Hanes, J. and Pluckthun, A., PNAS 94, 4937-42, 1997), many designed proteins with ankyrin repeat (DARPins) exhibiting binding specificity for VEGF-Axxx were selected from the DARPin N2C or N3C libraries described by Binz et al. 2004 (loc. cit.). The binding of selected clones to specific (VEGF-Axxx) and non-specific (MBP, E. coli maltose binding protein) targets was assessed by crude extract ELISA, indicating that VEGF-Axxx binding proteins were selected successfully (figure 1). The repeat domains of SEQ ID NO:1 to 7 constitute selected binding protein amino acid sequences comprising a repeat domain with binding specificity for VEGF-Axxx.
Analysis of specific linker sequences revealed specific motifs with ankyrin repeat sequence, inherent to certain selected families of linkers. 25 Selection of VEGF-Axxx-specific ankyrin-repeat proteins by visualization on ribosomes The selection of VEGF-Axxx-specific ankyrin-repeat proteins was performed by visualization on ribosomes (Hanes and Pluckthun, loc. cit.) using Canine VEGF-A164 or human VEGF-A165 30 as target proteins, the library of proteins designed with ankyrin repeat as described ( WO 02/020565, Binz et al., 2003, loc. cit. and Binz et al., 2004 , loc. cit) and established protocols (Zahnd, C, Amstutz, P. and
Pluckthun, A., Nat.
Methods 4, 69-79, 2007). Rounds of ribosome imaging selection were performed on canine and human variants of VEGF (including biotinylated variants, immobilized with neutrovidin or streptavidin) with both DARPin N2C and N3C DARPin 5 libraries using established protocols (Binz et al . 2004, loc. cit.). The number of reverse transcribed (RT)-PCR cycles after each round of selection was constantly reduced from 40 to 30, adjusting for the yield resulting from ligand enrichment.
Four initial rounds of selection on canine VEGF yielded clusters of DARPins with affinity at 10 nanomolar, as revealed by ELISA and SPR measurements of isolated clones.
In order to discover DARPins with more improved affinities, additional selections by dissociation were performed on human or canine biotinylated VEGF, immobilized with neutrovidin or streptavidin, removing clusters after the second and third initial round of selection by visualization on ribosomes , followed by a round of selection by dissociation in human VEGF.
Selected clones specifically bind to VEGF-Axxx as shown by crude extract ELISA DARP Select individual ins that specifically bound to VEGF-Axxx were identified by an enzyme-linked immunosorbent assay (ELISA) using crude extracts of Escherichia coli from cells expressing DARPin using standard protocols.
Selected clones were cloned into pQE30 expression vector (Qiagen), transformed into E. coli XL1-Blue (Stratagene) and then cultured overnight at 37 °C in a 96-deep well plate (each clone in an isolated cavity) containing 1 ml of growth medium (2YT including 1% glucose and 100 µg/ml ampicillin). 1 ml of fresh 2YT, containing 50 µg/ml ampicillin, was inoculated with 100 µl of overnight culture in a new 96-deep 30-well plate.
After incubation for 2 hours at 37 °C, expression was induced with IPTG (final concentration of 1 mM) and continued for 3 hours.
The cells were harvested, resuspended in 100 µL of B-PERII (Pierce) and incubated.
for 15 minutes at room temperature with stirring.
Then 900 µl of PBS-TB (PBS supplemented with 0.2% BSA, 0.1% Tween 20, pH 7.4) was added, and cell debris was removed by centrifugation. 5,100 µL of each lysate clone was applied to a well of a MaxiSorp plate coated with NeutrAvidin containing either a variant of VEGF-Axxx or the unrelated MBP, immobilized via its biotin cluster, and incubated for 1 hour at RT.
After intense washing with PBS-T (PBS supplemented with 0.1% Tween 20, pH 7.4) the plate was developed using standard ELISA procedures, using the anti-RGS(His)4 monoclonal antibody (34650, Qiagen ) as the primary antibody, and a polyclonal goat anti-mouse antibody conjugated to alkaline phosphatase (A3562, Sigma) as the secondary reagent.
Binding was then detected using disodium 4-nitrophenyl phosphate (4NPP, Fluka) as substrate for alkaline phosphatase.
Color development was measured at 405 nm.
The results of an example crude extract ELISA assay used to identify the binding of DARPins to VEGF-Axxx are shown in Figure 1. Screening several hundred clones by this crude cell extract ELISA assay revealed over hundreds of different ones DARPins with specificity for VEGF-Axxx.
These binding proteins were chosen for further analysis.
Examples of amino acid sequences of selected ankyrin repeat domains that specifically bind VEGF-Axxx are provided in SEQ ID NO:1 to 7. Deduction of sequence repeat motifs from selected repeat domains with specificity for VEGF-Axxx The amino acid sequences of selected repeat domains with binding specificity for VEGF-Axxx have been analyzed in more detail by tools to analyze sequences known to the skilled person skilled in the art (WO 02/020565; Forreret al. ., 30 2003, loc. cit.; Forrer, P., Binz, HK, Stumpp, MT and Plückthun, A., Chem-BioChem, 5(2), 183-189, 2004). However, unlike WO 02/020565, in which natural repetition motifs were used to de-
To produce sequence repeat motifs, here sequence repeat motifs were deduced from selected repeat domain repeat units with binding specificity for VEGF-Axxx.
In this way, families of selected repetition domains, comprising a common motif with sequence repetition, were determined.
High-Level and Soluble Expression of DARPins To be further analyzed, selected clones exhibiting specific binding to VEGF-Axxx in the crude cell extract ELISA assay as described above were expressed in E. coli XL1 cells -e purified, using their His tag and standard protocols. 25 ml of overnight stationary cultures (LB, 1% glucose, 100 mg/L ampicillin; 37 °C) were used to inoculate 1 liter cultures (same medium). At A(600) = 0.7, cultures were induced with 0.5 mM IPTG and incubated at 37 °C for 4 hours.
The cultures were centrifuged and the resulting granules were resuspended in 40 ml of TBS500 (50 mM Tris-HCI, 500 mM NaCl, pH 8) and subjected to sonication.
The lysate was again centrifuged, and glycerol (10% final concentration (v/v)) and imidazole (20 mM final concentration) were added to the resulting supernatant.
Proteins were purified on Ni-nitrilotriacetic acid column (column volume 2.5 20 ml) according to the manufacturer's instructions (QIAgen, Germany). From one liter of E. coli culture, up to 200 mg of highly soluble DARPins with binding specificity for VEGF-Axxx could be purified with > 95% purity, as estimated from SDS-15% PAGE.
These purified DARPins are used for further characterizations. 25 Example 2: Determination of IC50 values of selected DARPins with binding specificity for VEGF-Axxx in spheroid growth assay The addition of VEGF-Axxx to HUVEC spheroids embedded in collagen matrices leads to the budding of spheroids.
The addition of a VEGF-Axxx inhibitor will block the formation of shoots, which can be quantified statistically by the numbers and lengths of shoots.
The addition of different concentration of inhibitor and a constant amount of VEGF allows the determination of IC50. Inhibition of spheroid budding by DARPins specific for VEGF-Axxx Spheroid growth assays were performed according to standard protocols (Korff et al., loc. cit). DARPins with specificity for VEGF-Axxx were selected and purified to >96% purity as described in Example 1. Human umbilical vein cells were grown to confluence in monolayer culture.
After being treated with trypsin, the cell suspension was positioned so that a suspended drop 10 formed spheroids, that is, approximately 500 HUVEcs organized in aggregate.
The spheroids were embedded in a collagen matrix and stimulated with VEGF-A165 to initiate shoot growth.
Budding inhibitors were added to observe their effects on budding inhibition.
The number of shoots per spheroids and 15 the lengths of the shoots were quantified with graphic software.
The results of the two spheroid budding assays are shown in figure 2a (DARPin #30 with binding specificity for VEGF-Axxx) and in figure 2b (DARPin NC, negative control DARPin without binding specificity for VEGF-Axxx; for example , DARPin E3_5 20 (Binz et al., 2005, loc. cit.)). The DARPins with the best performances in this assay showed IC50 values in the range of 10 to 50 pM, while Avastin®, Lucentis® and Macugen® showed IC50 values in parallel experiments in the range of 150 and 500 pM.
Example 3: Determination of target specificity of DARPin 27 out of 25 compared to Avastin® by Surface Plasmonic Resonance analysis canine VEGF-A164 or canine VEGF-A164b were immobilized on a flow cell, and the interaction of DARPin on the 27 (the repeat domain of SEQ ID NO:1, corresponding to amino acids 1 to 159) and of Avastin® with the immobilized targets was analyzed.
Surface plasmon resonance (SPR) analysis The SPR was measured using a ProteOn instrument (BioRad). The running buffer was 20 mM HEPES, pH 7.4, 150 mM NaCl and 0.005% Tween 20. Approximately 1200 RU of canine VEGF-A164 or canine VEGF-A164b was immobilized on a GLC chip (BioRad). Interactions were measured at a flow of 60 µL/minute with buffer flow for 5 minutes, injection for 100 seconds of Avastin® or DARPin No. 27 at a concentration of 250 nM, and dissociation measurement of a few minutes with buffer flow.
The signal from an uncoated reference cell was subtracted from the measured values.
The results are shown in figure 3a (interaction of Avastin 10 with canine VEGF-A164), figure 3b (interaction of Avastin with canine VEGF-A164b), figure 3c (interaction of DARPin 27 with canine VEGF-A164) and figure 3d ( interaction of DARPin 27 with canine VEGF-A164b). While Avastin clearly interacts with the two immobilized isoforms of VEGF, DARPin no.27 shows interaction only with VEGF-A164 and not 15 with VEGF-A164b.
Example 4: In vivo Efficacy of DARPin #30 in Inhibiting VEGF-A165 in a Pegylated Rabbit Vascular Leakage Model DARPin #30 (the repeat domain of SEQ ID NO:4 corresponding to amino acids 1 to 126) or Lucentis® is applied by intravitreal injection into the eye of a rabbit to test its effectiveness in inhibiting vascular extravasation induced by a subsequent intravitreal injection of human VEGF-A165.
Measurement of vascular leakage in rabbits On Day 1, PBS, PEGylated DARPin at 30 (125 µg) or the equimolar amount of Lucentis® (162 µg) is applied by intravitreal injection into one eye of each rabbit (treated eye). On Day 4 or Day 30, the treated eye of each rabbit was exposed, by intravitreal injection, to 500 ng of human VEGF-A165.
Both eyes of all animals were evaluated 48 hours after VEGF-A165 injection for fluorescein content, measured in all 30 eyes 1 hour after an intravenous injection of sodium fluorescein (50 mg/kg animal body weight, 10% (w/v) in saline 0.9% (w/v)). The ratio of fluorescence values in treated and untreated eyes was calculated for every animal.
A ratio of one corresponds to the absence of fluorescent extravasation in the treated eye, a ratio greater than one indicates more fluorescent extravasation in the treated eye than in the untreated control eye. 5 Preparation of PEGylated DARPin Protein PEGylation using a single Cys residue and the chemical reaction involving maleimide is well known to the person skilled in the art and can be performed according to established protocols (eg Pierce's) . DARPin #30, comprising an additional linker 10 at the C-terminus (GGGSGGGSC, SEQ ID NO:8), was purified to near homogeneity by standard chromatographic methods.
The protein is completely reduced using DTT and purified by gel filtration to remove the DTT and by changing the buffer to PBS.
PEG-maleimide (methoxy-poly(ethylene glycol)-oxopropylamino-propyl maleimide; NOF, 15 ref.
Sunbright ME-200MA) dissolved in PBS is mixed with DARPin in PBS in approximately 15% molar excess of PEG-maleimide for 2-4 hours at room temperature.
PEGylated DARPin is then separated from non-reactive DARPin and non-reactive PEG groups by standard anion exchange chromatography. 20 The results are shown in figure 4. Both PEGylated DARPin 30 and Lucentis® were able to protect the rabbit eye from VEGF-A165 induced vascular extravasation after 4 days of being applied by intravitreal injections.
Nevertheless, only DARPin in the PEGylated 30, not Lucentis®, was able to protect the 25 Rabbit eye from vascular leakage induced by VEGF-A165 up to 30 days after the intravitreal injection.
In other experiments, the terminal intravitreal half-life of the different binding proteins of the invention was measured after intravitreal injections in the eyes of rabbits.
DARPin #30, comprising an additional linker at the C-terminus (GGGSGGGSC, SEQ ID NO:8), was conjugated to a 20 kDa and a 40 kDa non-proteinaceous PEG group using the respective maleimide PEGs of NOF (see Example 5). The terminal half-life was determined to be 3.5 days (+/- 0.3 days), 6.1 days (+/- 1.0 days) and 5.4 days (+/- 0.8 days ) for DARPin #30, DARPin #30 conjugated to the 20 kDa PEG pool and DARPin #30 conjugated to the 40 kDa PEG pool.
Surprisingly, increasing the molecular weight of the non-proteinaceous PEG pool from 20 kDa to 40 kDa did not result in an increased terminal half-life.
The same trend was observed in corresponding experiments, in which binding proteins comprising the repeat domain of SEQ ID NO:1 (amino acids 1 to 159) or SEQ ID NO:3 (amino acids 1 to 126), in instead of the 10 repeat domain of SEQ ID NO:4, were used.
Example 5: Recombinant binding proteins Examples of recombinant binding proteins, comprising a repeat domain that binds to VEGF-Axxx and a proteinaceous polymer cluster, are those of SEQ ID NO:1 and 4. The repeat domain The tion of SEQ ID NO:1 corresponds to amino acids 1 to 159 and the proteinaceous polymer group of SEQ ID NO:1 corresponds to amino acids 160 to 1,024. The repeat domain of SEQ ID NO:4 corresponds to amino acids 1 to 126 and the proteinaceous polymer cluster of SEQ ID NO:4 corresponds to amino acids 127 to 536. The binding proteins of SEQ ID NO:1 and 4 were expressed in the cytoplasm of Escherichia coli using standard techniques known to the person skilled in the art (see, for example, the pQE expression system from Qiagen (Germany)). The Met residue further encoded by the expression vector was efficiently cleaved in the cytoplasm of E. coli and separated from the expressed polypeptide, as the initial Met residue is followed by a short Gly (ie, the amino acid at position 1 of SEQ ID NO:1 and 4). Cells were lysed (eg, using a French press) and binding proteins were purified to near homogeneity from the crude cell extract using standard chromatographic techniques known to the person skilled in the art.
Examples of recombinant binding proteins, comprising a repeat domain that binds to VEGF-Axxx and a non-proteinaceous polymer cluster, were produced using the repeat proteins of SEQ ID NO:2, 3, 5, 6 and 7 These repeating proteins comprise a repeat domain at the N-terminus, followed by a polypeptide linker and a Cys at the C-terminus.
The respective 5-repeat domains correspond to amino acids 1 to 159 for SEQ ID NO:2 and 7, and amino acids 1 to 126 for SEQ ID NO:3 to 6. Proteins with a repeat of SEQ ID NO:2, 3, 5, 6 and 7 were expressed in the cytoplasm of Escherichia coli with standard techniques known to the person skilled in the art (see, for example, The Expressionist by Qiagen (Germany)). The Met residue additionally encoded by the expression vector was efficiently cleaved in the cytoplasm of E. coli and separated from the expressed polypeptide, as the initial Met residue is followed by a small Gly (ie, the amino acid at position 1 of the SEQ ID NO:2, 3, 5, 6 and 7). Cells were lysed (for example, using a French press) and binding proteins were purified to near homogeneity from the crude cell extract using standard chromatographic techniques known to one of ordinary skill in the art.
Repeat purified proteins, comprising a single Cys residue, were then conjugated to a non-proteinaceous polymer cluster by standard chemical reaction involving maleimide, as described in Example 4. In this way, a binding protein of the invention - tion, comprising the protein with SEQ ID NO:2 repeat and a 40 kDa non-proteinaceous PEG group (eg a 40 kDa maleimide-PEG ( -[3-(3-maleimido-1-oxopropyl) )amino]propyl-methoxy-polyoxyethylene) from NOF, product ref.
Sunbright ME-400MA), the protein with 25 repeat SEQ ID NO:3 and a 20 kDa non-proteinaceous PEG cluster (e.g., a 20 kDa maleimide-PEG ( -[3-(3-maleimido-1- oxopropyl)amino]propyl-methoxy-polyoxytethylene) from NOF, product ref.
Bright.
ME-200MA), the protein with SEQ ID NO:5 repeat and a 12 kDa non-proteinaceous PEG cluster (for example, a 12 kDa 30 maleimide-PEG ( -[3-(3-maleimido-1) -oxopropyl)amino]propyl-methoxy-polyoxyethylene) from NOF, product ref.
Sunbright ME-120MA), the SEQ ID NO:6 repeat protein and a 5 kDa non-proteinaceous PEG cluster (by e-
example, a 5 kDa maleimide-PEG (-[3-(3-maleimido-1-oxopropyl)amino]propyl-methoxy-polyoxyethylene) from NOF, product ref.
Sunbright ME-050MA) and the SEQ ID NO:7 repeat protein and a 2 kDa non-proteinaceous PEG cluster (eg, a 2 kDa maleimide-PEG 5 ( -[3-(3-) maleimido-1-oxopropyl)amino]propyl-methoxy-polyoxyethylene) from NOF, product ref.
Sunbright ME-020MA), was produced.
The repeat PEGylated proteins were then separated from the non-PEGylated repeat proteins and excess PEG by standard chromatographic techniques known to the skilled artisan. Therefore, SEQ ID NOs:2, 3, 5, 6 and 7 were conjugated at their C-terminal cysteine thiol to a PEG maleimide ( -[3-(3-maleimide-1-oxopropyl)amino]propyl - -methoxy-polyoxyethylene). The following structure was produced as follows:
15 wherein X is SEQ ID NO: 2, 3, 5, 6 or 7; and n is a positive integer.
权利要求:
Claims (15)
[1]
1. Recombinant binding protein, comprising an ankyrin repeat domain and a polyethylene glycol grouping of molecular weight of at least 5 kDa, wherein said ankyrin domain binds to VEGF-Axxx with Kd below 10-9 M and inhibit the binding of VEGF-Axxx to VEGFR-2.
[2]
The binding protein of claim 1, which has an apparent molecular weight of at least 100 kDa when analyzed at a concentration of 0.1 mM in PBS at room temperature by size exclusion chromatography using globular proteins as standards of molecular weight.
[3]
The binding protein of claim 1, wherein the capping module at the N-terminus of said ankyrin repeat domain comprises an Asp residue at position 5.
[4]
The binding protein of claim 1, wherein said ankyrin repeat domain competes for binding to VEGF-Axxx with the ankyrin repeat domains of SEQ ID NO:1 or 3.
[5]
The binding protein of claim 1, wherein said ankyrin repeat domain is selected from the group consisting of the ankyrin repeat domains of SEQ ID NO:1 to 7.
[6]
The binding protein according to any one of claims 1 to 5, wherein said ankyrin repeat domain is conjugated at its C-terminus by means of a peptide bond to a polypeptide linker and a residue of Cys at the C-terminus, where the thiol of said Cys 25 at the C-terminus is further conjugated to a polyethylene glycol coupled to maleimide.
[7]
The binding protein of claim 6, wherein the maleimide-coupled polyethylene glycol is -[3-(3-maleimido-1-oxopropyl)amino]propyl-methoxy-polyoxyethylene. 30
[8]
The binding protein of claim 1, wherein said binding protein is an ankyrin repeat protein selected from the group consisting of the ankyrin repeat proteins of SEQ ID NO:2, 3, 5, 6 or 7.
[9]
The binding protein according to any one of claims 6 to 8, wherein said polyethylene glycol group has a molecular weight around 20 kDa. 5
[10]
The binding protein of claim 1, which comprises the ankyrin repeat protein of SEQ ID NO:3, wherein the Cys thiol at the C-terminus of said ankyrin repeat protein is further conjugated to a polyethylene glycol coupled with maleimide.
[11]
The binding protein of claim 10, wherein said maleimide-coupled polyethylene glycol is -[3-(3-maleimido-1-oxopropyl)amino]propyl-methoxy-polyoxyethylene and wherein the group of polyethylene glycol has a molecular weight of at least 10 kDa.
[12]
A pharmaceutical composition, which comprises the binding protein of any one of claims 1 to 11, and optionally a pharmaceutical acceptable carrier and/or diluent.
[13]
A pharmaceutical composition as claimed in claim 12 for use in the treatment of an eye disease.
[14]
A pharmaceutical composition according to claim 12, for use in treating an eye disease by intravitreal injection. 20
[15]
A method of treating pathological angiogenesis in a mammal, including man, which comprises administering to a patient in need thereof an effective amount of a compound as defined in any one of claims 1 to 11.
类似技术:
公开号 | 公开日 | 专利标题
US10646542B2|2020-05-12|Modified binding proteins inhibiting the VEGF-A receptor interaction
JP6329503B2|2018-05-23|Binding protein that inhibits VEGF-A receptor interaction
AU2014243418A1|2014-10-30|Modified binding proteins inhibiting the VEGF-A receptor interaction
同族专利:
公开号 | 公开日
SI2563400T1|2019-11-29|
WO2011135067A1|2011-11-03|
MX347535B|2017-04-28|
US20130116197A1|2013-05-09|
HUE045272T2|2019-12-30|
IL222749A|2020-08-31|
NZ602943A|2014-07-25|
JP2013531616A|2013-08-08|
RS59408B1|2019-11-29|
RU2012151205A|2014-06-10|
EP2563400B1|2019-07-10|
KR101680064B1|2016-11-28|
CA2797996C|2017-10-03|
RU2605309C2|2016-12-20|
US9849158B2|2017-12-26|
EP2563400A1|2013-03-06|
CO6640296A2|2013-03-22|
ME03559B|2020-07-20|
US20210069293A1|2021-03-11|
US20160263185A1|2016-09-15|
KR20130076821A|2013-07-08|
PT2563400T|2019-09-27|
CA2797996A1|2011-11-03|
TW201141509A|2011-12-01|
US10646542B2|2020-05-12|
AU2011247563A1|2013-01-10|
ES2745491T3|2020-03-02|
PL2563400T3|2020-02-28|
US20140221295A1|2014-08-07|
ZA201207594B|2013-06-26|
SG185068A1|2012-12-28|
CN103002920B|2018-03-02|
MY162564A|2017-06-30|
CN103002920A|2013-03-27|
AU2011247563B2|2014-07-10|
DK2563400T3|2019-09-16|
TWI510246B|2015-12-01|
AR081361A1|2012-08-29|
US8710187B2|2014-04-29|
HRP20191660T1|2019-12-13|
LT2563400T|2019-10-10|
EP3593821A1|2020-01-15|
US20180125932A1|2018-05-10|
US9289466B2|2016-03-22|
UA111820C2|2016-06-24|
MX2012012638A|2013-06-28|
IL222749D0|2012-12-31|
JP6010528B2|2016-10-19|
CL2012003025A1|2013-08-23|
引用文献:
公开号 | 申请日 | 公开日 | 申请人 | 专利标题

EP1026240A3|1988-09-02|2004-04-07|Dyax Corp.|Generation and selection of recombinant varied binding proteins|
AU733674B2|1996-10-25|2001-05-24|Gilead Sciences, Inc.|Vascular endothelial growth factor nucleic acid ligand complexes|
US5270170A|1991-10-16|1993-12-14|Affymax Technologies N.V.|Peptide library and screening method|
US5348867A|1991-11-15|1994-09-20|George Georgiou|Expression of proteins on bacterial surface|
IL117645A|1995-03-30|2005-08-31|Genentech Inc|Vascular endothelial cell growth factor antagonists for use as medicaments in the treatment of age-related macular degeneration|
CA2196496A1|1997-01-31|1998-07-31|Stephen William Watson Michnick|Protein fragment complementation assay for the detection of protein-protein interactions|
CN101665536B|1997-04-07|2013-07-03|基因技术股份有限公司|Anti-vegf antibodies|
EP1325932B9|1997-04-07|2006-07-19|Genentech, Inc.|Anti-vegf antibodies|
WO1998048008A1|1997-04-23|1998-10-29|Plueckthun Andreas|Methods for identifying nucleic acid molecules encoding peptides that interact with target molecules|
CA2350417C|1998-12-02|2010-02-09|Phylos, Inc.|Dna-protein fusions and uses thereof|
US7270972B1|1999-04-02|2007-09-18|Ajinomoto Co., Inc.|Process for producing subunit peptide originating in polymer protein|
MEP3208A|1999-06-08|2010-02-10|Regeneron Pharma|Modified chimeric polypeptides with improved pharmacokinetic properties|
JP5291279B2|2000-09-08|2013-09-18|ウニヴェルジテート・チューリッヒ|A collection of repetitive proteins containing repetitive modules|
US7696320B2|2004-08-24|2010-04-13|Domantis Limited|Ligands that have binding specificity for VEGF and/or EGFR and methods of use therefor|
US8921528B2|2004-06-01|2014-12-30|Domantis Limited|Bispecific fusion antibodies with enhanced serum half-life|
WO2006137938A2|2004-11-08|2006-12-28|The University Of Texas At Austin|Antibody fragments for protection from pathogen infection and methods of use thereof|
NZ591733A|2004-11-12|2012-10-26|Bayer Schering Pharma Ag|Recombinant newcastle disease virus comprising a transgene encoding a fusion protein comprising a binding domain and a heterologous domain comprising an enzyme|
DE102005002978B4|2005-01-21|2013-04-25|Merz Pharma Gmbh & Co. Kgaa|Recombinant expression of proteins in a disulfide-bonded, two-chain form|
RU2412249C2|2005-07-08|2011-02-20|Юниверсити Оф Цюрих|Phage display with application of cotranslational translocation of fused polypeptides|
EP2402754B9|2006-03-06|2015-03-11|Amunix Operating Inc.|Unstructured recombinant polymers and uses thereof|
CA2646048A1|2006-03-30|2007-11-08|Novartis Ag|Compositions and methods of use for antibodies of c-met|
AU2007267798A1|2006-05-26|2007-12-06|Ipsen Pharma S.A.S.|Methods for site-specific pegylation|
AT516814T|2007-02-02|2011-08-15|Squibb Bristol Myers Co|10FN3 DOMAIN FOR THE TREATMENT OF DISEASES ACCOMPANIED BY UNWANTED ANGIOGENESIS|
DK2369005T3|2007-06-21|2013-06-24|Univ Muenchen Tech|Biologically active proteins with increased stability in vivo and / or in vitro|
CA2700391A1|2007-09-24|2009-04-02|University Of Zuerich|Designed armadillo repeat proteins|
DK2358746T3|2008-11-03|2020-12-21|Molecular Partners Ag|BINDING PROTEINS TO INHIBIT THE VEGF-A RECEPTOR INTERACTION|
TWI510246B|2010-04-30|2015-12-01|Molecular Partners Ag|Modified binding proteins inhibiting the vegf-a receptor interaction|
EP2643348A2|2010-11-26|2013-10-02|Molecular Partners AG|Improved capping modules for designed ankyrin repeat proteins|
CN104105704A|2011-04-29|2014-10-15|詹森生物科技公司|IL4/IL13 binding repeat proteins and uses|
WO2014001442A1|2012-06-28|2014-01-03|Molecular Partners Ag|Designed ankyrin repeat proteins binding to platelet-derived growth factor|
EP2874349B1|2012-08-01|2018-07-04|Huawei Technologies Co., Ltd.|Communication path processing method and apparatus|
EP2738180A1|2012-11-30|2014-06-04|Molecular Partners AG|Binding proteins comprising at least two binding domains against HER2.|
CN105209483B|2013-05-31|2021-07-27|分子组合公司|Designed ankyrin repeat proteins binding to hepatocyte growth factor|TWI510246B|2010-04-30|2015-12-01|Molecular Partners Ag|Modified binding proteins inhibiting the vegf-a receptor interaction|
EP2643348A2|2010-11-26|2013-10-02|Molecular Partners AG|Improved capping modules for designed ankyrin repeat proteins|
PL2846836T3|2012-05-07|2020-01-31|Allergan, Inc.|Method of treating amd in patients refractory to anti-vegf therapy|
WO2014001442A1|2012-06-28|2014-01-03|Molecular Partners Ag|Designed ankyrin repeat proteins binding to platelet-derived growth factor|
AU2012101677B4|2012-07-03|2012-12-20|Novartis Ag|Device|
JOP20200175A1|2012-07-03|2017-06-16|Novartis Ag|Syringe|
EP2738180A1|2012-11-30|2014-06-04|Molecular Partners AG|Binding proteins comprising at least two binding domains against HER2.|
CN105209483B|2013-05-31|2021-07-27|分子组合公司|Designed ankyrin repeat proteins binding to hepatocyte growth factor|
EP3010525A1|2013-06-20|2016-04-27|Novartis AG|Use of a vegf antagonist in treating choroidal neovascularisation|
WO2014203183A1|2013-06-20|2014-12-24|Novartis Ag|Use of a vegf antagonist in treating macular edema|
EP3010542A1|2013-06-20|2016-04-27|Novartis AG|Treatment of polypoidal choroidal vasculopathy|
MX2016000385A|2013-07-11|2016-04-29|Novartis Ag|Use of a vegf antagonist in treating retinopathy of prematurity.|
JP2016528202A|2013-07-11|2016-09-15|ノバルティス アーゲー|Use of VEGF antagonists in the treatment of choroidal retinal neovascularization and permeability disorders in pediatric patients|
SG11201510315TA|2013-07-12|2016-01-28|Ophthotech Corp|Methods for treating or preventing ophthalmological conditions|
KR20160070164A|2013-11-05|2016-06-17|알러간, 인코포레이티드|Method of treating conditions of the eye with an anti-vegf darpin|
WO2015109898A1|2014-01-24|2015-07-30|上海恒瑞医药有限公司|Vegf and pdgfrβ bispecific fusion protein and use thereof|
US9388239B2|2014-05-01|2016-07-12|Consejo Nacional De Investigation Cientifica|Anti-human VEGF antibodies with unusually strong binding affinity to human VEGF-A and cross reactivity to human VEGF-B|
EP3492495A1|2014-05-12|2019-06-05|Formycon AG|Pre-filled plastic syringe containing a vegf antagonist|
KR20160075107A|2014-12-19|2016-06-29|주식회사 이노테라피|PEGylated VEGF-Trap and Method for Preparation Thereof|
MX2017012485A|2015-04-02|2018-04-11|Molecular Partners Ag|Designed ankyrin repeat domains with binding specificity for serum albumin.|
KR101676690B1|2015-08-04|2016-11-16| 에빅스젠|Tetrapeptide having effect of inhibiting VEGF-induced angiogenesis and use thereof|
RU2734958C2|2015-11-18|2020-10-26|Формикон Аг|Pre-filled pharmaceutical package containing a liquid formulation based on a vegf antagonist|
BR112018009951A8|2015-11-18|2019-02-26|Sio2 Medical Products Inc|ophthalmic drug|
MX2018006171A|2015-11-18|2018-12-19|Formycon Ag|Pre-filled plastic syringe containing a vegf antagonist.|
EP3407868A1|2016-01-26|2018-12-05|Formycon AG|Liquid formulation of a vegf antagonist|
EP3423835A1|2016-02-29|2019-01-09|Maintect GmbH|Predictive markers useful in the treatment of wet age-related macular degeneration|
MX2019000727A|2016-07-20|2019-05-02|Aerpio Therapeutics Inc|Humanized monoclonal antibodies that target ve-ptp .|
US10717772B2|2016-09-22|2020-07-21|Molecular Partners Ag|Recombinant binding proteins targeting HER2 and serum albumin, and their uses|
CN108623694B|2017-03-24|2021-09-17|北京市肿瘤防治研究所|Derivative of vascular endothelial cell growth factor receptor antagonistic peptide F56 and application thereof|
EP3630043A1|2017-05-24|2020-04-08|Formycon AG|Sterilizable pre-filled pharmaceutical packages comprising a liquid formulation of a vegf-antagonist|
WO2018218013A2|2017-05-24|2018-11-29|Sio2 Medical Products, Inc.|Sterilizable pharmaceutical package for ophthalmic formulations|
WO2018215580A1|2017-05-24|2018-11-29|Formycon Ag|Method for sterilizing prefilled plastic syringes containing a vegf antagonist|
WO2019020777A1|2017-07-26|2019-01-31|Formycon Ag|Liquid formulation of a vegf antagonist|
WO2020068653A1|2018-09-24|2020-04-02|Aerpio Pharmaceuticals, Inc.|MULTISPECIFIC ANTIBODIES THAT TARGET HPTP - βAND VEGF|
AR119080A1|2019-06-04|2021-11-24|Molecular Partners Ag|MULTISPECIFIC PROTEINS|
WO2021116462A1|2019-12-11|2021-06-17|Molecular Partners Ag|Designed ankyrin repeat domains with altered surface residues|
US20210347835A1|2020-05-06|2021-11-11|Molecular Partners Ag|Novel ankyrin repeat binding proteins and their uses|
WO2021229067A1|2020-05-14|2021-11-18|Molecular Partners Ag|Multispecific proteins|
法律状态:
2021-07-20| B06F| Objections, documents and/or translations needed after an examination request according [chapter 6.6 patent gazette]|
2022-02-08| B06U| Preliminary requirement: requests with searches performed by other patent offices: procedure suspended [chapter 6.21 patent gazette]|
优先权:
申请号 | 申请日 | 专利标题
EP10161685|2010-04-30|
EP10161685.2|2010-04-30|
PCT/EP2011/056824|WO2011135067A1|2010-04-30|2011-04-29|Modified binding proteins inhibiting the vegf-a receptor interaction|
[返回顶部]