![]() ALBUMIN BINDING POLYPEPTIDE, FUSION OR CONJUGATE PROTEIN, POLYNUCLEOTIDE, METHOD TO PRODUCE A POLYPE
专利摘要:
albumin-binding polypeptide, fusion protein or conjugate, polynucleotide, method for producing a polypeptide, composition, method for preparing a composition, and method for increasing the solubility of a compound is a class of modified polypeptides that have a binding affinity for albumin. it is also about new methods and uses that explore the binding of these and other compounds to albumin in different contexts, some of which have significance for the treatment or diagnosis of diseases in mammals, including humans. 公开号:BR112013000452A2 申请号:R112013000452-5 申请日:2011-07-08 公开日:2020-02-11 发明作者:Ekblad Caroline;Abrahmsen Lars 申请人:Affibody Ab; IPC主号:
专利说明:
ALBUMIN BINDING POLYPEPTIDE, FUSION OR CONJUGATE PROTEIN, POLYNUCLEOTIDE, METHOD TO PRODUCE A POLYPEEPTIDE, COMPOSITION, METHOD FOR THE PREPARATION OF A COMPOSITION, AND, METHOD FOR INCREASING THE SOLUBILITY OF A TECHNICAL FIELD The present disclosure relates to a class of modified polypeptides that have a binding affinity for albumin. It also refers to new methods and uses that explore the binding of these and other compounds to albumin in different contexts, some of which have significance for the treatment of disease in mammals, including humans. FUNDAMENTALS Serum albumin Serum albumin is the most abundant protein in mammalian sera (40 g / 1; approximately 0.7 mM in humans) and one of its functions is to bind molecules such as lipids and bilirubin (Peters, Advances in Protein Chemistry 37: 161, 1985 ). Serum albumin is devoid of any enzymatic or immunological function. In addition, human serum albumin (HSA) is a natural carrier involved in endogenous transport and delivery of numerous natural as well as therapeutic molecules (Sellers and KochWeser, Albumin Structure, Function and Uses, eds Rosenoer et al, Pergamon, Oxford, p 159, 1977). The half-life of serum albumin is directly proportional to the size of the animal, where, for example, human serum albumin has a half-life of 19 days and serum rabbit albumin has a half-life of about 5 days ( McCurdy et al, J Lab Clin Med 143: 115, 2004). SAH is widely distributed throughout the body, in particular, in the interstitial and blood compartments, where it is mainly involved in maintaining osmolarity. 2/74 Structurally, albumins are single-chain proteins that comprise three homologous domains and a total of 584 or 585 amino acids (Dugaiczyk et al, Proc Natl Acad Sei USA 79:71, 1982). Albumin contains 17 disulfide bridges and a simple reactive thiol cysteine at position 34, but lacks N-linked and O-linked carbohydrates (Peters, 1985, supra-, Nicholson et al, Br J Anaesth 85: 599, 2000 ). Fusion or association with HSA results in increased in vivo protein half-life Several strategies have been reported to covalently couple proteins directly to serum albumin or to a peptide or protein that will allow in vivo association with serum albumin. Examples of the latter approach have been described, for example, in WO 91/01743, WO 01/45746 and Dennis et al (J Biol Chem 277: 35035-43, 2002). The first document describes, among others, the use of peptides or albumin-binding proteins derived from streptococcal G protein (SpG) to increase the half-life of other proteins. The idea is to fuse the bacterially-derived albumin-binding peptide / protein to a peptide / protein of therapeutic interest, which has been shown to have rapid elimination from the blood. The thus generated fusion protein binds serum albumin in vivo, and benefits from its longer half-life, which increases the network half-life of the fused peptide / protein of therapeutic interest. WO 01/45746 and Dennis et al refer to the same concept, but here, the authors use relatively short peptides to bind serum albumin. The peptides were selected from a library of peptides displayed on phage. In Dennis et al, initial work is mentioned in which the enhancement of an immune response to a recombinant fusion of albumin that links the streptococcal G protein domain to the 3/74 Type 1 human complement was found. Patent application No. US 2004/0001827 (Dennis) also discloses the use of constructs that comprise peptide ligands, again identified by phage display technology, that bind to serum albumin and that are conjugated to bioactive compounds for targeting tumor. Bacterial receptor protein albumin binding domains Streptococcal G protein (SpG) is a bifunctional receptor present on the surface of certain streptococcal filaments and capable of binding both IgG and serum albumin (Bjôrck et al, Mol Immunol 24: 1113, 1987). The structure is highly repetitive with several different domains structurally and functionally (Guss et al, EMBO J 5: 1567, 1986), more precisely, three Ig binding domains and three serum albumin binding domains (Olsson et al, Eur J Biochem 168: 319, 1987). The structure of one of the three serum albumin binding domains in SpG was determined, showing a fold fold of three helices (Kraulis et al, FEBS Lett 378: 190, 1996, Johansson et al, J. Biol. Chem. 277: 8114-20, 2002). An archetype of 46 amino acids was defined as ABD (albumin binding domain) and was subsequently also designated as G148GA3 (GA for G protein related albumin binding). In, for example, document No. WO 09/016043, albumin binding variants of the 46 amino acid ABD archetype are disclosed. Bacterial albumin binding domains other than those in protein G have also been identified, some of which are structurally similar to those of protein G. Examples of proteins that contain such albumin binding domains are the PAB, PPL, MAG and ZAG proteins (Rozak et al, Biochemistry 45: 3263-3271, 2006). Structure studies and 4/74 function of such albumin-binding domains have been performed and reported, for example, by Johansson et al. (Johansson et al, J Mol Biol 266: 859-865, 1997). In addition, Rozak et al reported the creation of artificial variants of G148-GA3, which were selected and studied in relation to the specificity and stability of different species (Rozak et al, Biochemistry 45: 3263-3271, 2006), while Jonsson et al developed artificial variants of G148-GA3 that have a greatly improved affinity for human serum albumin (Jonsson et al, Prot Eng Des Sei 21: 515-27, 2008). For some of the variants, a higher affinity was achieved at the cost of reduced thermal stability. In addition to the proteins that contain three helices described above, there are also other unrelated bacterial proteins that bind albumin. ABD and immunization Recently, few B and T cell epitopes have been experimentally identified within the region of streptococcal G protein filament albumin 148 (G148) (Goetsch etal, Clin Diagn Lab Immunol 10: 125-32, 2003). The authors behind the study were interested in using G148 T cell epitopes in vaccines, that is, to utilize the inherent immunostimulatory property of the albumin-binding region. Goetsch et al additionally found in a B cell epitope, that is, a region bound by antibodies after immunization, following G148. In pharmaceutical compositions for human administration, no immunoresponse is desired. Therefore, the G148 albumin binding domain is therefore unsuitable for use in such compositions due to its immunostimulatory properties mentioned above. DESCRIPTION The disadvantages and deficiencies above the technique 5/74 are overcome or alleviated by, in a first aspect, an albumin-binding polypeptide that comprises an amino acid sequence selected from i) LAX 3 AKX 6 X 7 ANXio eldx 14 ygvsdf YKRLIX 26 KAKTVEGVEALKX 39 X 4 o ILX 43 X 44 LP in which independently of each other X 3 is selected from E, S, Q and C; X e is selected from E, S and C; X 7 is selected from A and S; Xio is selected from A, S and R; X14 is selected from A, S, Ce K; X 26 is selected from D eE; X 39 is selected from D eE; X 40 is selected from A and E; X 43 is selected from A eK; X 44 is selected from A, S and E; L at position 45 is present or absent; and P in position 46 is present or absent; and ii) an amino acid sequence that has at least 95% identity to the sequence defined in i). The class of polypeptides related to the sequence defined above that has a binding affinity for albumin is derived from a common parental polypeptide sequence, which folds into a cluster domain of three alpha helices. More specifically, polypeptides, as described above, are derived from a model construction based on a complex structure between serum albumin and the albumin binding domain G148-GA3 (Lejon et al, J Biol Chem 279: 42924 -8, 2004), as well as analyzes of binding and structural properties of various mutational variants of the common parental polypeptide sequence. The amino acid sequence i) defined above 6/74 comprises amino acid substitutions when compared to the parental polypeptide sequence that results in a class of polypeptides that is expected to fold into an almost identical three-helix cluster domain. Although the parental polypeptide sequence already comprises a binding surface for interaction with albumin, that binding surface is modified by some of the substitutions, according to the definition above. Substitutions according to the definition above provide an improved albumin binding ability when compared to the parental polypeptide sequence. The binding polypeptides according to the first aspect of the disclosure exhibit a set of characteristics that, for example, make them suitable for use as fusion partners or conjugates for therapeutic molecules for human administration. The binding polypeptides according to the present disclosure demonstrate, for example, in comparison with related albumin binding polypeptides such as the G148-GA3 albumin binding domain and the albumin binding polypeptides disclosed in WO 09/016043 , at least five of the following six characteristics: • Polypeptides exhibit a different surface compared to, for example, G148-GA3 and other bacterially derived albumin binding domains. The difference can decrease or eliminate any risk of antibody reactions in a subject, such as a human being, who has previously been exposed to such bacterial proteins. • Polypeptides comprise fewer potential T cell epitopes than, for example, G148-GA3 and other mutational variants related to, but different from, the common parental polypeptide sequence, and therefore exhibit low immunogenicity when administered to a subject, how 7/74 a human being. • Polypeptides exhibit lower reactivity with circulating antibodies when administered to a subject, such as a human. Thus, through amino acid substitutions on the surface of polypeptides exposed to circulating antibodies, that is, on the surface of the polypeptide not involved in the binding interaction with albumin, antibody cross-reactivity is reduced when compared to, for example, antibody cross-reactivity. caused by G148-GA3 as measured in a human serum test suite. • Polypeptides have a high albumin binding ability, either in terms of a higher binding affinity, as defined by a K Dz value or in terms of a slower dissociation rate, as defined by a k O value ff / which, for example, known naturally occurring albumin-binding polypeptides, such as albumin-binding domains derived from bacterial proteins. • Polypeptides comprise less amino acid residues that are associated with polypeptide stability problems, for example, known naturally occurring albumin binding polypeptides, such as albumin binding domains derived from bacterial proteins. Thus, polypeptides comprise, for example, no methionine or tryptophan susceptible to oxidation and only one asparagine. • Polypeptides have a higher structural stability, as defined by a melting point of about 55 ° C, than previous albumin-binding polypeptides, such as those disclosed in WO 09/016043. In one embodiment, the polypeptide binding to 8/74 albumin according to the first aspect exhibits all six characteristics of those listed above. In another embodiment, the albumin-binding polypeptide according to the first aspect exhibits, when bound to albumin, a more hydrophilic profile than, for example, the previous albumin-binding polypeptides, such as those disclosed in WO 09 / 016043. The surface of the albumin-binding polypeptide that is exposed to the environment when the polypeptide interacts with albumin comprises a smaller amount of amino acid residues that impart surface hydrophobicity. As the skilled person will realize, the function of any polypeptide, such as the albumin-binding capacity of the polypeptides according to the first aspect, is dependent on the tertiary structure of the polypeptide. However, it is possible to make changes in the amino acid sequence in an α-helical polypeptide without affecting its structure (Taverna and Goldstein, J Mol Biol 315 (3): 479-84, 2002; He et al, Proc Natl Acad Sci USA 105 (38): 14412-17, 2008). Thus, the modified variants of i), which are such that the resulting sequence is at least 95% identical to the sequence belonging to the class defined by i), are also encompassed by the first aspect. For example, it is possible that an amino acid residue belonging to certain functional groups of amino acid residues (for example, hydrophobic, hydrophilic, polar etc.) could be exchanged for another amino acid residue of the same functional group. The term% identical or% identity, as used in the specification and the claims, is calculated as follows. The query sequence is aligned to the target sequence using the CLUSTAL W algorithm (Thompson, J.D., Higgins, D.G. and Gibson, T.J., Nucleic Acids Research, 22: 4673-4680 (1994)). A comparison is made on the window that corresponds to the shortest of the aligned strings. More The short 9/74 of the aligned sequences may in some cases be the target sequence, such as the albumin-binding polypeptide disclosed herein. In other cases, the query string may constitute the shortest of the aligned strings. The query string can, for example, consist of at least 10 amino acid residues, such as at least 20 amino acid residues, such as at least 30 amino acid residues, such as at least 40 amino acid residues, for example, 45 amino acid residues. The amino acid residues at each position are compared and the percentage of positions in the query sequence that have identical matches in the target sequence is reported as% identity. In a r polypeptide nodality in Link in albumin accordingIn another with the first aspect, X 6 is a polypeptide modality AND.in Link in albumin accordingIn another with this aspect, X 3 is S. modality of the polypeptide in Link in albumin accordingIn another with this aspect, X 3 is E. modality of the polypeptide in Link in albumin accordingIn another with this aspect, X 7 is A. modality of the polypeptide in Link in albumin accordingIn another with this aspect, X i4 is S. modality of the polypeptide in Link in albumin accordingIn another with this aspect, X 14 is C. modality of the polypeptide in Link in albumin accordingIn another with this aspect, Χ χ0 is A. modality of the polypeptide in Link in albumin accordingIn another with this aspect, Χ ϊ0 is S. modality of the polypeptide in Link in albumin accordingIn another with this aspect, X 2 6 is D. modality of the polypeptide in Link in albumin accordingIn another with this aspect, X 2 6 is E. modality of the polypeptide in Link in 10/74 albumin according to this aspect, X39 is D. in Link in In another polypeptide mode albumin accordingIn another with this aspect, X 39 is E. modality of the polypeptide in Link in albumin accordingIn another with this aspect, X 4 is A. modality of the polypeptide in Link in albumin accordingIn another with this aspect, X 43 is A. modality of the polypeptide in Link in albumin accordingIn another with this aspect, X 44 is A. modality of the polypeptide in Link in albumin accordingIn another with this aspect, X 44 is S. modality of the polypeptide in Link in albumin according with that aspect, the residue L at position 45 is present.In another polypeptide mode in Link in albumin according with that aspect, the residue P in position 46 is present.In another polypeptide mode in Link in albumin according with that aspect, the residue P in position 46 is absent.In another modality, 0 polypeptide in Link in albumin according with this aspect is subject to the condition in that X 7 is not L, E, nor D. The albumin-binding polypeptide according to the first aspect can be prepared for conjugation with a suitable conjugation partner by replacing surface-exposed amino acid residues with, for example, either a cysteine or a lysine. Such substitutions can be introduced into the N terminal helix, i.e. helix one, of the polypeptide, which is the helix furthest from serum albumin when the albumin-binding polypeptide is bound to serum albumin. In this way, a lysine residue at position X 14 of the sequence defined in i) can be used to 11/74 enable site-targeted conjugation. This can, moreover, be advantageous when the molecule is produced by chemical peptide synthesis, since the orthogonal protection of the epsilon-amine group of said lysine can be used. In addition, a cysteine residue can be introduced into the amino acid sequence to enable site-directed conjugation. For example, a cysteine residue can be introduced in any of the X 3 , X 6 and / or X 14 positions according to the definition above. The coupling of a conjugation partner to the epsilon-amine of a lysine or to the thiol group of a cysteine represents two chemically different alternatives for obtaining site-directed conjugation using an amino acid residue within the amino acid sequence i). As the skilled person understands, there are other chemical alternatives for preparing an amino acid sequence for conjugation and as such are also within the scope of the present disclosure. An example of such a chemistry is a click-like chemistry made possible by the introduction of tyrosine as presented by Ban et al (J Am Chem SOC 132: 1523 to 5, 2009). The terms albumin binding and binding affinity for albumin as used in this specification refer to a property of a polypeptide that can be tested, for example, by using surface plasmon resonance technology, as in a Biacore instrument . For example, as described in the examples below, albumin binding affinity can be tested in an experiment in which albumin or a fragment thereof is immobilized on a sensor chip in the instrument, and the sample containing the polypeptide to be tested is passed to the chip. Alternatively, the polypeptide to be tested is immobilized 12/74 on an instrument's sensor chip, and a sample containing albumin or a fragment of it is passed to the chip. In this sense, albumin may be a mammalian serum albumin, like human serum albumin. The knowledgeable person can then interpret the results obtained by such experiments to establish at least one qualitative measure of the polypeptide binding affinity for albumin. If a quantitative measure is desired, for example, to determine a K D value for the interaction, surface plasmon resonance methods can also be used. Connection values can, for example, be defined on a Biacore2000 instrument (GE Healthcare). Albumin is properly immobilized on a measurement sensor chip and samples of the polypeptide, whose affinity is to be determined, are prepared by serial dilution and injected. K D values can then be calculated from the results using, for example, the Langmuir connection model 1: 1 of the BIAevaluation 4.1 software provided by the instrument manufacturer (GE Healthcare). In one embodiment, the albumin-binding polypeptide according to this aspect binds to albumin so that the k of f value of the interaction is at most 5 x 10 5 s 1 , at most 5 x 10 ' 6 s' 1 . As described above, albumin-binding polypeptides, as defined by amino acid sequence i), are derived from a common parental polypeptide sequence that folds into a clustering domain of three alpha helices. In one embodiment, the three helix domain of this parental polypeptide sequence forms part of the G protein from the G148 filament of Streptococcus, in particular, the GA3 domain. In another embodiment, the amino acid sequence of the albumin-binding polypeptide is selected from 13/74 any one of SEQ ID NO: 1-144 and SEQ ID NO: 164-203, as selected from any one of SEQ ID NO: 1-144. More specifically, the amino acid sequence is selected from SEQ ID NO: 4-5, SEQ ID NO: 7-8, SEQ ID NO: 10-11, SEQ ID NO: 13-14, SEQ ID NO: 16- 17, SEQ ID NO: 19-20, SEQ ID NO: 2223, SEQ ID NO: 25-26, SEQ ID NO: 28-29, SEQ ID NO: 31-32, SEQ ID NO: 34-35, SEQ ID NO: 37-38, SEQ ID NO: 41-42, SEQ ID NO: 49-50, SEQ ID NO: 164-170 and SEQ ID NO: 192-203. Thus, the amino acid sequence can be selected from SEQ ID NO: 4-5, SEQ ID NO: 7-8, SEQ ID NO: 10-11, SEQ ID NO: 13-14, SEQ ID NO: 1617 , SEQ ID NO: 19-20, SEQ ID NO: 22-23, SEQ ID NO: 25-26, SEQ ID NO: 28-29, SEQ ID NO: 31-32, SEQ ID NO: 34-35, SEQ ID NO: 37-38, SEQ ID NO: 41-42 and SEQ ID NO: 49-50. In one embodiment, the albumin-binding polypeptide according to that aspect further comprises one or more additional amino acid residues positioned at the N and / or C terminus of the sequence defined in i). These additional amino acid residues may play a role in enhancing albumin binding by the polypeptide and in improving the conformational stability of the folded albumin binding domain, but may serve equally well for other purposes, related, for example, to one or more more among production, purification, in vivo or in vitro stabilization, coupling, identification or detection of the polypeptide, as well as any combination thereof. Such additional amino acid residues may comprise one or more amino acid residues added for chemical coupling purposes, for example, to a chromatographic resin to obtain an affinity matrix or to a chelation half for complexing with a radiometal. The amino acids that directly precede or follow the alpha helix at the N or C end of the amino acid sequence i) can, therefore, in one mode affect stability Conformational 14/74. An example of an amino acid residue that can contribute to improved conformational stability is a serine residue positioned at the N-terminus of amino acid sequence i) as defined above. The N-terminal serine residue can in some cases form a canonical S-X-X-E cover box, by involving hydrogen agglutination between the gamma oxygen of the serine side chain and the NH of the polypeptide main chain of the glutamic acid residue. Such N-terminal coverage can contribute to stabilization of the first alpha helix of the three-helix domain that constitutes the albumin-binding polypeptide according to the first aspect of the disclosure. Thus, in one embodiment, the additional amino acids comprise at least one serine residue at the N-terminus of the polypeptide. The amino acid sequence is, in other words, by one or more serine residues. In another embodiment of the albumin-binding polypeptide, the additional amino acids comprise a glycine residue at the N-terminus of the polypeptide. It is understood that the amino acid sequence i) can be preceded by one, two, three, four or any suitable number of amino acid residues. Thus, the amino acid sequence can be preceded by a single serine residue, a single glycine residue or a combination of the two, such as a glycine-serine (GS) combination or a glycine-serine-serine (GSS) combination. Examples of albumin-binding polypeptides that comprise additional N-terminal amine residues are shown in SEQ ID NO: 145-163, as in SEQ ID NO: 145-148 and SEQ ID NO: 162-163. In yet another embodiment, the additional amino acid residues comprise a glutamic acid at the N-terminus of the polypeptide as defined by sequence i). Similarly, C-terminal coverage can be exploited to improve stability of the third alpha helix 15/74 of the three helix domain that make up the albumin-binding polypeptide. A proline residue, when present at the C-terminus of the amino acid sequence defined in i), can, at least partially, function as a cover residue. In such a case, a lysine residue that follows the proline residue at the C-terminus may contribute to further stabilization of the third helix of the albumin-binding polypeptide by the hydrogen agglutination between the epsilon amine group of the lysine residue and the carbonyl groups of the amino acids located two and three residues before lysine in the polypeptide backbone, for example, when both L45 and ο P46 are present, the carbonyl groups of the leucine and alanine residues of the amino acid sequence defined in i). Thus, in one embodiment, the additional amino acids comprise a lysine residue at the C-terminus of the polypeptide. As discussed above, the additional amino acids can be related to the production of the albumin-binding polypeptide. In particular, when an albumin-binding polypeptide according to a modality in which ο P4 6 is present is produced by chemical peptide synthesis, one or more optional amino acid residues that follow the C-terminal proline can provide advantages. Such additional amino acid residues can, for example, prevent the formation of unwanted substances, such as diketopiperazine in the dipeptide stage of synthesis. An example of such an amino acid residue is glycine. Thus, in one embodiment, the additional amino acids comprise a glycine residue at the C-terminus of the polypeptide, which directly follows the proline residue or which follows a glycine and / or lysine residue as considered above. Alternatively, polypeptide production may benefit from amidation of the C-terminal proline residue of amino acid sequence i), when 16/74 present. In that case, the C-terminal proline comprises an additional amine group on the carboxylic carbon. In a modality of the polypeptides described in this document, in particular, which end at their C-terminus with proline or another amino acid known to racemize during peptide synthesis, the aforementioned addition of a glycine to C-terminus or proline amidation, when present, it can also oppose potential problems with racemization of the C-terminal amino acid residue. If the polypeptide, amidated in this way, is designed to be produced by recombinant means, rather than by chemical synthesis, amidation of the C-terminal amino acid can be performed by various methods known in the art, for example, through the use of amidation PAM enzyme . Examples of albumin-binding polypeptides that comprise additional C-terminal amino acid residues are shown in SEQ ID NO: 145-152, as in SEQ ID NO: 148-150. The knowledgeable person is aware of methods to achieve the C-terminal modification, such as by different types of pre-produced matrices for peptide synthesis. In another embodiment, the additional amino acid residues comprise the C and / or N terminal cysteine residue of the polypeptide. Such a cysteine residue can precede and / or directly follow the amino acid sequence as defined in i) or it can precede and / or follow any other additional amino acid residues as described above. Examples of albumin-binding polypeptides that comprise a cysteine residue at the C and / or N-terminus of the polypeptide chain are shown in SEQ ID NO: 149-150 (C-terminus) and SEQ ID NO: 151-152 (N-terminus ). By adding a cysteine residue to the polypeptide chain, a thiol group for site-directed conjugation of the albumin-binding polypeptide can be obtained. Alternatively, a selenocysteine residue can be introduced at the C-terminus 17/74 of the polypeptide chain, in a manner similar to the introduction of a cysteine residue, to facilitate site-specific conjugation (Cheng et al, Nat Prot 1: 2, 2006). In one embodiment, the albumin-binding polypeptide comprises no more than two cysteine residues. In another embodiment, the albumin-binding polypeptide comprises no more than a cysteine residue. In another embodiment, the additional amino acid residues of the albumin-binding polypeptide comprise a tag for purification or detection of the polypeptide, such as a hexaistidyl (Hisg) tag or a myc tag (c-Myc) or a FLAG tag for interaction with antibodies specific for labeling and / or to be used for purification. The knowledgeable person is aware of other alternatives. In yet another embodiment, the albumin-binding polypeptide according to this aspect binds to human serum albumin. In other embodiments, the albumin-binding polypeptide binds albumin from species other than the human species, such as albumin from mice, rats, dogs and cynomolgus monkeys. The additional amino acid residues discussed above can also constitute one or more polypeptide domains with any desired function, such as the same binding function as the first, albumin binding domain or other binding function or a therapeutic function or an enzymatic or fluorescent function or mixtures thereof. Consequently, in another related aspect, a conjugate or fusion protein is provided which comprises i) a first half consisting of a binding polypeptide according to the first aspect 18/74 as described in this document; and ii) a second half consisting of a polypeptide that has a desired biological activity. A conjugate or fusion protein comprising a binding polypeptide according to the first aspect of the disclosure and a second half can increase the in vitro and / or in vivo half-life of the second half, when compared to the in vivo half-life of the second half alone. As a consequence, when a conjugate or fusion protein according to this aspect is administered to a subject, such as a human subject, in vivo exposure to the second half may increase, which may lead to the enhanced potency of the biological activity of the second half, when compared to the potency of the in vivo exposure of the second half when administered alone. The desired biological activity can, for example, be a therapeutic activity, a binding activity or an enzymatic activity. When the desired biological activity is therapeutic activity, the second half showing this activity can be a therapeutically active polypeptide. Non-limiting examples of therapeutically active polypeptides are biomolecules, such as molecules selected from the group consisting of human endogenous enzymes, hormones, growth factors, chemokines, cytokines and lymphokines, and biologically active non-human proteins, such as the proteins of the group that it consists of bacterial toxins (eg, staphylococcal and streptococcal superantigens and pseudomonas exotoxin), enzymes (eg, RNase and beta-lactamase) and activating proteins (eg, streptocinase). Non-limiting examples of therapeutically active biomolecules that may be useful in a fusion or conjugated to the albumin-binding polypeptide are selected from the group consisting of IL-2, GLP-1, BNP (peptide 19/74 Alb-beta-natriuretic), IL-1-RA (interleukin-1 receptor antagonist), KGF (keratinocyte growth factor), Stemgen®, growth hormone (GH), G-CSF, CTLA-4, myostatin, Factor VII, Factor VIII and Factor IX. Defective blood vessels pierced with tumor tissue make their vasculature (endothelial barrier) permeable to macromolecules, whereas in healthy tissue blood vessels, only small molecules can pass through the endothelial barrier. Likewise, the permeability of the blood-brain barrier to albumin in inflamed joints of patients with rheumatoid arthritis is markedly increased. Thus, fusion proteins or conjugates according to this aspect are likely to permeate blood vessels in tumor tissue and the hematoarticular barrier in inflamed joints. When said desired biological activity of the second half is binding activity, said second half can be a binding polypeptide capable of selectively interacting with a target molecule. Such a binding polypeptide can, for example, be selected from the group consisting of antibodies and fragments and domains thereof that substantially retain antibody binding activity; microbodies, maxibodies, avimers and other small disulfide-bound proteins; and framework-derived binding proteins selected from the group consisting of staphylococcal protein A and domains thereof, other three helix domains, lipocalins, ankyrin repeating domains, cellulose binding domains, crystalline γ, fluorescent green protein, antigen 4 associated with human cytotoxic lymphocyte T, protease inhibitors such as Kunitz domains, PDZ domains, SH3 domains, peptide aptamers, staphylococcal nuclease, tendamistates, fibronectin type III domain, transferin, zinc fingers 20/74 and conotoxins. In some embodiments, the target molecule for binding the target binding polypeptide can be selected from the group consisting of amyloid peptide β (Αβ) from Alzheimer's disease; other disease-associated amyloid peptides; toxins, such as bacterial toxins and snake venom; blood clotting factors, such as von Willebrand factor; interleukin, such as IL-13; myostatin; proinflammatory factors, such as TNF-oi, TNF-oi receptor, IL-1, IL-8 and IL-23; complement factors, such as C3 and C5; hypersensitivity mediators, such as histamine and IgE; tumor-related antigens, such as CD19, CD20, CD22, CD30, CD33, CD40, CD52, CD70, cMet, HER1, HER2, HER3, HER4, CAIX (carbonic anhydrase IX), CEA, IL-2 receptor, MUC1, PSMA, TAG-72; and other biological molecules such as G-CSF, GM-CSF, growth hormone (GH), insulin and somatostatin. As a skilled person understands, the albumin-binding polypeptide according to the first aspect can be useful in a fusion protein or as a conjugated partner to any other half. Therefore, the above lists of therapeutically active polypeptides, binding polypeptides and target molecules should not be construed as limiting in any way. In an embodiment of a fusion protein or conjugate according to the present disclosure, the second half is conjugated to the albumin-binding polypeptide by means of a lysine or cysteine residue added to the C or N-terminus of the albumin-binding polypeptide or through a lysine or cysteine residue at the position within the albumin-binding polypeptide, as at a position selected from X 3 , X 6 and X 14 . If the conjugation site is one within the amino acid sequence i) of the albumin-binding polypeptide, such as a cysteine at position X i4 , none 21/74 additional amino acid needs to be added to the albumin-binding polypeptide in order to enable conjugation to the second half. A conjugation site within the polypeptide chain as defined by i) may furthermore protect the polypeptide against cross-reactive antibodies, in particular, the portion of the polypeptide close to the conjugation site. Without wishing to be bound by theory, when the conjugate, via the albumin-binding polypeptide, is bound to serum albumin in vivo, that is, located in the serum albumin-binding pocket, the second half, conjugated in the position inside the, for example, helix one of the domain of three helices that make up the albumin-binding polypeptide, points out from the serum albumin to which the albumin-binding polypeptide is attached. In addition, a conjugation site within the albumin-binding polypeptide may impair the presentation of the portion of the peptides otherwise derived from that portion of the polypeptide to T cells due to, for example, effects on processing in the antigen presenting the cell, fitting impaired potential peptide in the peptide binding groove of the class II MCH molecule, and remodeled peptide surface available to the T cell receptor (due to the highlighted conjugated portion). Thus, the immunogenicity of the portion of the conjugate close to the conjugation site is expected to be further reduced after conjugation. Due to the high affinity between the albumin-binding polypeptide of the present disclosure and serum albumin, a conjugate or fusion protein comprising such albumin-binding polypeptide can be considered as an indirect complex with serum albumin. A conjugate or fusion protein according to the present disclosure thereby provides an alternative to the frequently used method of exploring direct fusions or conjugates with serum albumin. 22/74 Such direct conjugates with serum albumin are often heterogeneous, regardless of which method is used for coupling. When a specific molecule is coupled to the serum albumin via an amine group of a lysine residue, any one of a large number of lysines on the surface of the serum albumin molecule can be targeted, which gives a random conjugation site and a random product. Although coupling of thiol by means of the single unpaired cysteine to human serum albumin (at position 34, Peters, 1985, supra) appears to offer an alternative method for obtaining a direct conjugate, such a methodology often does not lead to a homogeneous product. Only 20% to 60% of the molecules in commercially available serum (human) albumin exhibit a free thiol group, while the rest are blocked by thiol compounds such as cysteine, homocysteine or glutathione. In contrast, the conjugation of the three helix domain of the binding polypeptide according to the present disclosure can be performed in a site specific manner. This can be achieved, as discussed above, or by coupling to one or more cysteines, a selenocysteine or a designated lysine (orthogonally protected during synthesis). According to that aspect of the present disclosure, the second half having the desired biological activity can either be conjugated to the three helix domain of the albumin-binding polypeptide or produced as a fusion protein therewith. A non-limiting example of a conjugate according to the present disclosure is given below. Glucagon 1-like peptide (GLP-1), or a derivative thereof, is a small polypeptide that may suitably be present as a second half in a conjugate with the albumin-binding polypeptide. Conjugation of GLP-1 to the albumin-binding polypeptide can be performed in any of the 23/74 polypeptide sequence positions, as described above. The conjugate can thus be produced in a non-biological process and is expected to exhibit a significantly enhanced potency when compared to the potency of GLP-1 alone. Conjugation can be used with both small polypeptides and small proteins, such as GLP-1, or with large polypeptides or proteins. A conjugate according to the present disclosure can typically comprise a non-amino acid spacer moiety, such as polyethylene glycol (PEG). Other polypeptides or proteins can be combined with an amino acid sequence of the albumin-binding polypeptide in the form of a fusion protein. Such a fusion protein may, in addition, comprise one or more spacer amino acid residues between the first and the second halves. As described above, the albumin-binding polypeptide, according to the first aspect, binds serum albumin from various species, including mouse, rat, dog and cynomolgus monkeys. In this way, a conjugate or fusion protein, according to the present disclosure, can contribute to intensify the biological effect of a second half, not only in a human subject, but also in animal models. Several endogenous proteins have been produced as direct fusions with human serum albumin, examples of such proteins include G-CSF, GH, interferons, CD4, IL-2, insulin, glucagon, GLP-1, antibody Fab fragments and protease inhibitors such as proteins derived from the Kunitz domain. However, such direct mergers may not be fully evaluated in animal models. This is due to the fact that human serum albumin does not interact properly with the endogenous neonatal Fc receptor (FcRn), for example, in the commonly used animal models, mouse and rat, and that 24/74 this interaction is an important factor that contributes to the long circulation time of serum albumin. As described above, a conjugate or fusion protein according to the present disclosure can, in the presence of serum albumin, combine with albumin and function as an indirect fusion with albumin. This makes a conjugate or fusion protein comprising a binding polypeptide, according to the first aspect, useful in pre-clinical model species, as long as the second half is biologically active in the selected species. In one embodiment, a fusion protein or conjugate is provided that comprises an additional portion consisting of a polypeptide that has an additional desired biological activity, which may be the same or different from that of the second portion. A specific example of such a conjugate or fusion protein comprises a therapeutically active polypeptide as defined above as a second portion and a binding polypeptide as defined above as an additional portion. Regarding the above description of conjugates or fusion proteins that incorporate an albumin-binding polypeptide according to the first aspect, it should be noted that the designation of first, second and additional portions is made for the purpose of evidence to distinguish between polypeptide or albumin-binding polypeptides according to the present description on the one hand, and portions that exhibit other functions on the other hand. These designations are not intended to refer to the actual order of the different domains in the polypeptide chain of the conjugate or fusion protein. Thus, for example, said first portion may, without restriction, appear at the N-terminal end, in the middle, or at the C-terminal end of the conjugate or fusion protein. In a related aspect, a 25/74 albumin-binding polypeptide, conjugate or fusion protein as defined in the present description, which further comprise an organic molecule, as a cytotoxic agent. Non-limiting examples of cytotoxic agents that can be fused or conjugated to an albumin-binding polypeptide according to the first aspect, or combined with a conjugate or fusion protein according to the second aspect, are selected from calicheamicin, auristatin, doxorubicin , maytansinoid, taxol, ecteinascidin, geldanamycin, methotrexate and their derivatives, and combinations thereof. Previously, attempts have been made to treat various disorders with direct albumin conjugates. Such direct albumin conjugates have been explored, for example, with doxorubicin against cancer (Kratz et al, J Med Chem 45: 5,523 to 33, 2002) and methotrexate against rheumatoid arthritis (Wunder et al, J Immunol 170: 4,793 to 4,801, 2003). It should be understood that the albumin-binding polypeptide, either alone or as a moiety in a conjugate or fusion protein, through its high albumin-binding capacity, provides an indirect means of building albumin complexes, and thus it can provide an alternative treatment method compared to the attempts mentioned above. The above aspects further cover polypeptides in which the albumin-binding polypeptide according to the first aspect, or the albumin-binding polypeptide as comprised in a conjugate or fusion protein according to the second aspect, endowed with a identification group, as an identification selected from the group consisting of fluorescent metals and dyes, chromophoric dyes, chemiluminescent compounds and bioluminescent proteins, enzymes, radionuclides and particles, for example, for polypeptide detection purposes. In In particular, the description encompasses a radio tagged polypeptide consisting of a radioquelate of an albumin-binding polypeptide, conjugate or fusion protein as described herein and a radionuclide, such as a radioactive metal. In embodiments in which the labeled albumin-binding polypeptide comprises an albumin-binding polypeptide according to the first aspect of the description and an identification, the labeled polypeptide can, for example, be used for indirectly labeling serum albumin. Due to the strong association between the labeled polypeptide and serum albumin, the labeled polypeptide can be used, for example, to study vascular permeability and blood accumulation. In other embodiments, the labeled albumin-binding polypeptide is present as a portion in a conjugate or fusion protein that also comprises a second portion that has a desired biological activity. The identification may, in some cases, be coupled only to the albumin-binding polypeptide, and in some cases, to the albumin-binding polypeptide and to the second portion of the conjugate or fusion protein. When reference is made to a labeled polypeptide, this should be understood as a reference to all aspects of polypeptides as described herein, including proteins and fusion conjugates that comprise an albumin-binding polypeptide and a second and, optionally, moieties additional. Thus, a labeled polypeptide can contain only the albumin-binding polypeptide and, for example, a therapeutic radionuclide, which can be chelated or covalently coupled to the albumin-binding polypeptide, or contain the albumin-binding polypeptide, a therapeutic radionuclide and a second portion as a 27/74 small molecule that has a desired biological activity as a therapeutic efficacy. In embodiments in which the albumin-binding polypeptide, the fusion conjugate or protein is radio-tagged, such a radio-tagged polypeptide may comprise a radionuclide. A majority of radionuclides are metallic in nature and metals typically do not have the ability to form stable covalent bonds with elements presented in proteins and peptides. For this reason, the labeling of proteins and peptides with radioactive metals is performed with the use of chelators, that is, multidentified ligands, which form non-covalent compounds, called chelates, with metal ions. In an embodiment of the albumin-binding polypeptide, conjugate or fusion protein, the incorporation of a radionuclide is possible by providing a chelation environment, through which the radionuclide can be coordinated, chelated or complexed with respect to the polypeptide. An example of a chelator is the polyamino-polycarboxylated type of chelator. Two classes of such polyamine polycarboxylated chelators can be distinguished: macrocyclic and acyclic chelators. In one embodiment, the albumin-binding polypeptide, conjugate or fusion protein comprises a chelation environment provided by a polyamino-polycarboxylated chelator conjugated to the albumin-binding polypeptide through a thiol group of a cystine residue or an epsilon amine group of a lysine residue. The macrocyclic chelators most commonly used for radioisotopes of indium, gallium, yttrium, bismuth, radioaactinide and radiolanthanide are different derivatives of DOTA (1,4,7,10-tetraazacyclododecane-l, 4,7,10tetraacetic acid). In one embodiment, the chelation environment of the albumin-binding polypeptide, conjugate or protein of 28/74 merger is provided by DOTA or a derivative thereof. More specifically, a group of chelation polypeptides covered by the present description is produced by reacting DOTA-derived 10-maleimidoethylacetamide 1,4,7,10-tetraazacyclododecane-1,4,7-tris-acetic acid (maleimidomonoamide-DOTA) with , for example, a thiol group of the albumin-binding polypeptide, for example, as described in Example 5. The high kinetic inertia, that is, the slow rate of dissociation of DOTA metal, favors stable fixation of a radionuclide. However, high temperatures are necessary for labeling due to a slow association rate. For this reason, DOTA derivatives are widely used for labeling short peptides, such as the albumin binding polypeptides of the present description, which exhibit binding functionality after incubation at temperatures required for the labeling reaction. The most commonly used acyclic polyamine polycarboxylated chelators are different derivatives of DTPA (diethylenetriamine-pentaacetic acid). Therefore, polypeptides that have a chelation environment provided by diethylenetriamineapentaacetic acid or derivatives thereof are also covered by the present description. It has been found that semi-rigid variants of the modified main structure of DTPA provide adequate stability for labeling with 90 Y of, for example, Zevalin®. Although acyclic chelators are less inert, and consequently less stable than macrocyclic ones, their labeling is fast enough even at room temperature. For this reason, they can be used for labeling conjugates or fusion proteins according to the present description. Detailed protocols for coupling polyamine polycarboxylated chelators to proteins and peptides from 29/74 targeting were published by Cooper et al (Nat Prot 1: 314 to 7, 2006) and by Sosabowski and Mather (Nat Prot 1: 972 to 6, 2006). An albumin-binding polypeptide, conjugate or fusion protein according to the aspects described herein coupled to a polyamino-polycarboxylated chelator can be used to provide a radio-labeled polypeptide consisting of a radioquelate of the albumin-binding polypeptide, conjugate or protein fusion coupled to the chelator and a radionuclide suitable for medical imaging, in which the radionuclide is selected from the group consisting of 61 Cu, 64 Cu, 66 Ga, Ê7 Ga, 68 Ga, 110m In, 11: L In, 44 Sc and 86 Y, or with a radionuclide suitable for therapy, where the radionuclide is selected from the group consisting of 225 Ac, 212 Bi, 213 Bi, 67 Cu, 16S Ho, 177 Lu, 212 Pb, 149 Pm, 153 Sm , 227 Th and 90 Y, the radionuclide being complexed with the albumin-binding polypeptide through a chelator. In the same modalities, the polypeptide can also be radio-tagged with non-metallic radioisotopes using the so-called indirect labeling. Thus, for labeling with, for example, 18 F, 76 Br, different isotopes of iodine and 211 At, intermediate binding molecules are used for labeling. Such a binding molecule should contain two functional portions, one that provides fast and effective radio tagging, and the other that allows fast and efficient coupling to proteins, for example, to amino groups, or, preferably, to the thiol group of an exclusive cystine, as in Χ ϊ4 position of the albumin-binding polypeptide. For example, a malemide group reacts with thiol groups to form a stable thioether bond. The linker molecule can first be reacted with radioidentification and subsequently with the thiol or selenethiol group of the protein. In another aspect, a polynucleotide is provided that 30/74 encodes an albumin-binding polypeptide or a fusion protein as described herein. Also covered is a method of producing an albumin-binding polypeptide or fusion protein as described above, which comprises expressing the polynucleotide, an expression vector comprising the polynucleotide and a host cell comprising the expression vector. The albumin-binding polypeptide of the present description can alternatively be produced by non-biological peptide synthesis with the use of amino acids and / or amino acid derivatives that have protected reactive side chains, in which the non-biological peptide synthesis comprises gradually coupling the amino acids and / or the amino acid derivatives to form a polypeptide according to the first aspect that has protected reactive side chains, remove the protective groups from the reactive side chains of the polypeptide, and wrap the polypeptide in aqueous solution. In this way, normal amino acids (eg, glycine, alanine, phenylalanine, isoleucine, leucine and valine) and pre-protected amino acid derivatives are used to sequentially construct a sequence of polypeptides, either in solution or on a solid support in an organic solvent. A specific example of peptide synthesis on solid support is described in Example 5. When a complete polypeptide sequence is constructed, the protecting groups are removed and the polypeptide is allowed to be folded into an aqueous solution. Each polypeptide according to the present description folds reversibly into a three-helix cluster domain with no added factors, and therefore folds spontaneously. The conjugate according to the second aspect can be 31/74 produced by a method comprising producing an albumin-binding polypeptide according to any of the methods described above, such as through non-biological peptide synthesis, and conjugating the produced albumin-binding polypeptide with a second and / or additional portion as defined in connection with the second aspect. In one embodiment of a conjugate or fusion protein, a conjugate or fusion protein is further provided as defined herein for use in therapy, for example, for use as a medicament. Such a conjugate or fusion protein may exhibit an in vivo half-life that is longer than the in vivo half-life of the polypeptide that has a desired biological activity per se. The conjugate or fusion protein may, in addition, extract none or a reduced immunoresponse upon administration to the mammal, as a human, as compared to the immunoresponse extracted upon administration to the mammal of the polypeptide which has a desired biological activity per se. Alternatively expressed, this provides a method to decrease the immunogenicity of a polypeptide that has a desired biological activity, by fusing or conjugating such a polypeptide to an albumin-binding polypeptide, conjugate or fusion protein according to revealed aspects in this document. In addition, this may allow intensification of the biological activity of a second portion. In another embodiment, a conjugate or fusion protein according to the present description is provided for use in diagnosis, for example, for use as a diagnostic agent. The present description also relates to different aspects of the use of the albumin-binding polypeptide described above, as well as several methods for 32/74 treatment, diagnosis and detection in which the polypeptide is useful due to its, among others, binding characteristics. When referring to the albumin-binding polypeptide in the following description of these uses and methods, it is understood that this term encompasses the albumin-binding polypeptide alone, but also all those molecules based on that polypeptide described above which, for example, incorporates the albumin-binding polypeptide as a portion in a conjugate or fusion protein, and / or are conjugated in an identification, 10 a chelator, a therapeutic and / or diagnostic agent, and / or are provided with additional amino acid residues such as a label or for other purposes. As explained above, such fusion proteins, derivatives, etc., form a part of the present description. Another set of aspects is related to the provision of new means to increase the solubility in aqueous solution of an unsatisfactorily soluble compound, through the conjugation of it in an albumin-binding polypeptide, conjugate or fusion protein. The following 20 unsatisfactorily soluble compound complex and an albumin-binding polypeptide, alone or incorporated as a portion in a conjugate or fusion protein, has the ability to associate with albumin in vivo or in vitro, the association of which increases solubility in aqueous solution. Thus, in a form of this additional aspect, a composition is provided, which comprises a compound which, per se, has a solubility in water not greater than 100 pg / ml; coupled to an albumin-binding polypeptide, conjugate 30 or fusion protein as described herein, the compound and albumin-binding polypeptide, conjugate or fusion protein are covalently coupled. 33/74 In one embodiment, the compound has, per se, a solubility of no more than 10 pg / ml. In yet another embodiment, said solubility is not greater than 1 pg / ml. In some embodiments, the compound may be a pharmaceutically active compound, for example, a cytotoxic agent. Non-limiting examples of cytotoxic agents are those selected from calicheamicin, auristatin, doxorubicin, maytansinoid, taxol, ecteinascidin, geldanamycin and their derivatives, and combinations thereof. 10 Alternatively, the cytotoxic agent may be a synthetic chemotoxin produced by organic synthesis and not derived from a naturally occurring compound. In addition to the unsatisfactorily soluble compound and the albumin-binding polypeptide, conjugate or fusion protein, the composition according to that aspect of the description may, in some embodiments, also comprise a binding polypeptide with an affinity for a clinically relevant target. This binding polypeptide is suitably different from the 20 albumin binding polypeptide, and can be coupled covalently or non-covalently to the other components of the composition of the invention. As non-limiting examples, the binding polypeptide with an affinity for a clinically relevant target can be selected from the group consisting of antibodies and fragments and 25 domains thereof which substantially retain antibody binding activity; micro-bodies, maxibodies, avimers and other small disulfide-bound proteins; and binding proteins derived from a framework selected from the group consisting of staphylococcal protein A and domains thereof, 30 other three helix domains, lipocalins, anquirin repeat domains, cellulose binding domains, crystalline γ, green fluorescent protein, antigen 4 associated with human cytotoxic T lymphocyte, inhibitors of 34/74 protease as Kunitz domains, PDZ domains, SH3 domains, aptamer peptides, staphylococcal nucleasse, tendamistats, fibronectin type III domain, transferrin, zinc fingers and conotoxins. The composition according to the above aspect of the present description has an ability to associate with albumin in vivo or in vitro, by providing in the composition an albumin-binding polypeptide, either alone or as present in a conjugate or fusion protein . In 10 certain cases, it may be beneficial to form a complex of the composition with albumin outside a living organism, that is, to add exogenous albumin to the composition. Such a composition can be lyophilized, providing a formulation that is suitable for storage at room temperature. Accordingly, the present description also provides a composition as defined above which further comprises albumin, such as human serum albumin. The present description also provides the composition according to the above aspect for use as a medicament, i.e., for use in therapy, in cases where the compound is a therapeutically active compound. Suitably, the delivery of an albumin-binding polypeptide, conjugate or fusion protein and, optionally, albumin, does not adversely affect the therapeutic effectiveness of the active compound, so the composition of the invention will be useful in those therapeutic settings or prophylactics in which the compound per se is indicated. In another embodiment, the composition according to the above aspect is provided for use as a diagnostic agent, i.e., for use in diagnosis. A related aspect of the present description provides a method of preparing a composition as described immediately above. The method comprises 35/74 provide a compound which per se has a solubility in water not greater than 100 pg / ml; and covalently coupling the compound to an albumin-binding polypeptide, conjugate or fusion protein according to the aspects as described herein, thus forming a composition comprising a covalent complex of compound and polypeptide of binding of albumin, conjugate or fusion protein. In embodiments of the present description in which albumin is included in the composition, the method may comprise the additional step of mixing said albumin-binding compound compound and polypeptide complex, albumin fusion protein, thereby forming a composition comprising a non-covalent complex i) of the covalent complex of albumin-binding compound and polypeptide, conjugate or fusion protein, and ii) of albumin. The relative proportions of the two components of this non-covalent complex can, for example, be 1: 1, so that one unit of the unsatisfactorily soluble compound complex and albumin-binding polypeptide, conjugate or fusion protein is associated with a molecule of albumin. In one embodiment, the method further comprises lyophilizing the non-covalent complex to obtain a lyophilized composition. In another approximately related aspect, the present description provides a method of increasing the aqueous solubility of a compound, which comprises providing a compound which, per se, has a solubility in water not greater than 100 pg / ml; coupling the compound covalently to an albumin-binding polypeptide, conjugate or fusion protein according to the aspects as described herein, thus forming a covalent complex of compound and conjugated albumin-binding polypeptide or protein 36/74 fusion; and mixing said albumin-binding polypeptide compound and polypeptide complex to albumin under conditions that promote the non-covalent association of the albumin-binding polypeptide to albumin; the water solubility of the compound in said complex being superior to the water solubility of the compound per se. In these aspects of the method related to the solubility of an unsatisfactorily soluble compound, the 10 optional features of the various components are as described in connection with the aspect of the immediately preceding composition. Although the invention has been described with reference to several exemplary modalities, it will be understood by the person skilled in the art that various changes can be made and equivalents can be replaced by elements of the same without departing from the scope of the invention. In addition, many modifications can be made in order to adapt a particular situation or molecule to the teachings of the invention without deviating from its essential scope. Therefore, it is intended that the invention is not limited to any particular modality contemplated for carrying out that invention, but that the invention will include all modalities that fall within the scope of the appended claims. FIGURES Figure 1 is a listing of the amino acid sequences of examples of the albumin-binding polypeptides of the present description (SEQ ID NO: 1-152, SEQ ID NO: 155-203), the GA3 domain of G148 strain of Streptococcus strain 30 extended by an N-terminal glycine residue (SEQ ID NO: 153) and an albumin-binding polypeptide derived from G148-GA3 as previously described by Jonsson et al (supra, SEQ ID NO: 154). 37/74 Figure 2 shows the result of the binding analysis performed on a Biacore instrument to investigate the binding of the albumin-binding polypeptide PEP07912 (SEQ ID NO: 157) to human serum albumin. Three different concentrations of purified protein (40 nM, gray fat line; 10 nM, black line; and 2.5 nM, gray line) were injected into a surface with 955 RU of immobilized human serum albumin. Figures 3A to C show the result of * 10 binding analysis performed by ELISA to investigate the binding of albumin binding polypeptides PEP07913 (SEQ ID NO: 153), PEP06923 (SEQ ID NO: 154), PEP07271 (SEQ ID NO: 155), PEP07912 (SEQ ID NO: 157), PEP07554 (SEQ ID NO: 156), PEP07914 (SEQ ID NO: 158), PEP07968 (PEP07911 DOTA conjugated , SEQ ID NO: 159) and 15 PEP07844 (SEQ ID NO: 161), the IgG molecules present in individual normal human serum 126, where A) shows the median OD value, B) shows the percentage of negative serum (defined as OD <0.15), and C) shows the percentage of positive serum (defined as OD> 1.0). Figures 4A to B are chromatograms showing the analysis of purified chemically produced albumin-binding polypeptide PEP07834 (SEQ ID NO: 160), where A) shows the absorbance signal at 220 nm, in white subtracted, and B) shows the absorbance signal at 280 nm, blank * 25 subtracted. The two peaks appeared at both wavelengths. ▼ Figures 5A to B are spectrograms that show the mass spectrometric analysis of the two peaks identified in Figure 4A) and B). A) is the spectrogram of the first peak, i.e., the monomer of PEP07834 (SEQ ID NO: 160), and B) is the spectrogram of the dimer of PEP07834. Figures 6A to C are diagrams showing an immunogenicity assessment of HIV-binding polypeptides. 38/74 albumin PEP07913 (SEQ ID NO: 153), PEP07912 (SEQ ID NO: 157), PEP07914 (SEQ ID NO: 158) and PEP07968 (PEP07911 DOTA conjugate, SEQ ID NO: 159) in a cell proliferation assay T CD3 + CD4 + . A) shows the number of individuals that respond to albumin-binding polypeptides compared to recombinant human albumin in a cohort of 52 Caucasian donors. B) shows the median stimulus indices (SI) for PEP07913, PEP07912, PEP07914 and PEP07968 compared to the negative control containing recombinant human albumin. C) shows the number of individuals who respond against all proteins in the study as compared to the buffer control. Figures 7A to C show the result of binding analysis performed on a Biacore instrument to investigate the binding of albumin binding polypeptides A) PEP07986 (SEQ ID NO: 163), B) PEP08296 (PEP08185 DOTA conjugate, SEQ ID NO: 148) and 0) PEP06923 (SEQ ID NO: 154) to albumin of different species. The sensograms shown correspond to the protein injected at a concentration of 40 nM on surfaces immobilized with human albumin (113 0 RU), thin gray line; cinomolgo monkey (1046 RU), thick gray line; rat (831 RU), thick light gray line; dog (1053 RU), thin black line; and mouse (858 RU), thick black line. Figure 8 shows the inhibitory effect of Z x -PP013 (open circles), Z Y -PP013 (open squares) and Z neg -PP013 (closed triangles) on cytokine-induced TF-1 cell proliferation in the presence of molar excess of five times HSA. Figure 9 shows the maximum binding responses obtained by Biacore analysis of PEP07986 (SEQ ID NO: 163) stored at 4, 25 or 40 ° C for one week, two weeks, one month and three months as indicated, at a concentration of 2 mg / ml, injected in immobilized HSA (704 RU) at a concentration of 10 nM. The untreated samples of time time = 0 are 39/74 shown as references. Figure 10 shows the result of binding analysis performed on a Biacore instrument to investigate the binding of albumin-binding polypeptide PEP08296 (PEP08185 DOTA conjugate, SEQ ID NO: 148) to human serum albumin before and after heat treatment. Two concentrations of PEP08296 (0.8 nM, gray lines; 4 nM, black lines) were injected into a surface with 724 RU of immobilized human serum albumin. The solid lines are anterior heat treatment and dotted lines, posterior heat treatment for 10 minutes at 90 ° C. Figures 11A to B show the overlapping of two CD spectra of PEP08296 (PEP08185 DOTA conjugate, SEQ ID NO: 148) before and after heat treatment for 12 minutes at 90 ° C. A) Sample incubated in PBS pH 7.2. B) Sample incubated in PBS pH 4.0. Figure 12 shows the maximum intensity projection image (MIP) of the entire 68 Ga-PEP08296 body distribution in a healthy mouse, added during 1.5 hours of data collection immediately after intravenous injection (tail vein). Circulating radioactivity in the main vessels (for example, the jugular (long arrow) and femoral (short arrow)), the heart (H), liver (L), spleen (S), kidney (K) and bladder (B) are promptly outlined. Figure 13 shows a gel filtration chromatogram of PEP07986 (SEQ ID NO: 163) injected at a concentration of 42 mg / ml, solid black line. A chromatogram of ovalbumin (Mw 43 kDa) injected at a concentration of 5 mg / ml, broken gray line, is included for comparison, confirming that the peak for PEP07986 is not an aggregate, which would be expected in the volume of empty space eluted in an instant before ovalbumin. The invention will now be illustrated 40/74 additionally through the non-limiting description of experiments conducted in accordance with it. Unless otherwise specified, conventional methods of chemistry and molecular biology have been used throughout. EXAMPLES Example 1: Cloning, expression, purification and characterization of albumin-binding polypeptides In this example, ten different albumin-binding polypeptides, PEP07913 (SEQ ID NO: 153), PEP07912 (SEQ ID NO: 156), PEP07914 (SEQ ID NO: 158), PEP07968 (PEP07911 DOTA conjugate, SEQ ID NO: 159) , PEP06923 (SEQ ID NO: 154), PEP07271 (SEQ ID NO: 155), PEP07554 (SEQ ID NO: 156), PEP07844 (SEQ ID NO: 161), PEP07986 (SEQ ID NO: 163) and PEP08296 (PEP08185 DOTA conjugate, SEQ ID NO: 148), their amino acid sequences are shown in Figure 1 and in the attached sequence listing, they have been cloned, purified and characterized. Material and methods Cloning of albumin-binding polypeptide variants Mutations in G148-GA3 were generated using site-directed mutagenesis with the appropriate oligonucleotides to obtain the desired albumin-binding polypeptide variants. The gene fragments were amplified by PCR with primers adding specific endonuclease sites as well as an MGSS Nterminal sequence that precedes the albumin-binding polypeptide variants. The fragments were cleaved with Ndel and Notl, purified and linked to a cloning vector, plasmid pAY02556 (containing an origin of replication of pBR322, a kanamycin resistance gene and a T7 promoter for expression of the gene of interest), restricted with the same enzymes. The ligations were transformed into TOPIO E. coli cells 41/74 electrocompetent. The transformed cells were spread on TBAB plates (30 g / 1 blood tryptose based agar) supplemented with 50 pg / ml kanamycin, followed by incubation at 37 ° C overnight. The colonies were scanned using PCR and the sequencing of amplified fragments was performed using biotinylated oligonucleotide and a Terminator v3.1 BigDye® Cycle Sequencing Kit (Applied Biosystems), used according to the manufacturer's protocol. The sequencing reactions were purified by binding to spheres coated with magnetic streptavidin using a Magnatrix 8000 (NorDiag AB), and analyzed on an ABI PRISM® 3100 Genetic Analyzer (PE Applied Biosystems). All albumin-binding polypeptide variants were subcloned as monomers and the constructs encoded by the expression vectors were MGSS- [PP ###], where PP ### corresponds to the amino acid residues that make up the sequence of the polypeptide-binding polypeptide. albumin. In addition, the G148-GA3, PP007 (SEQ ID NO: 7), PP013 (SEQ ID NO: 13) and PP037 (SEQ ID NO: 37) gene fragments were amplified by PCR with primers adding well-specific endonuclease sites such as a hexahistidine sequence, a TEV protease sites and a glycine residue before the amino acid residues that make up the albumin-binding polypeptide sequence. Polypeptides PEP07913 (SEQ ID NO: 153), PEP07912 (SEQ ID NO: 157), PEP07914 (SEQ ID NO: 158) and PEP07968 (SEQ ID NO: 159) correspond to the albumin-binding polypeptides G148GA3, PP007 (SEQ ID NO: 7), PP013 (SEQ ID NO: 13) and PP037 (SEQ ID NO: 37) with added glycine residues. The fragments were cleaved with XbaT and NotT, purified and linked to a cloning vector, plasmid pAY02512 (containing an origin of replication of pBR322, a kanamycin resistance gene and 42/74 a T7 promoter for expression of the gene of interest. The cloning site is preceded by a sequence encoding a peptide containing a hexahistidine tag followed by a dividing site for the TEV protease), restricted with the same enzymes. The ligation, transformation and sequence verification were carried out as described above. The four variants of G148-GA3 albumin-binding polypeptide, PP007, PP013 and PP037 were subcloned as monomers and the constructs encoded by the expression vectors were MGSSHHHHHHLQSSGVDLGTENLYFQG- [PP ###]. The expression vector encoding MGSSHHHHHHLQSSGVDLGTENLYFQG- [PP013] was further modified by site-directed mutagenesis with the use of oligonucleotides, resulting in the insertion of a serine residue before the amino acid residues that constitute the sequence of the albumin-binding polypeptide, to obtain the construct MGSSHHHHHHLQSSGVDLGTENLYFQGS- [PP013]. This construct was further modified by 1) site-directed mutagenesis to replace the serine residue at position 14 (in PP013) with a cystine residue, generating MGSSHHHHHHLQSSGVDLGTENLYFQGS- [PP049], and 2) adding a C-terminal glycine residue, generating MGSSHHHHHHLQSSGVDLGTENLYFQGS- [PP049] -G. The addition of Cterminal glycine was performed by PCR amplification with primers including nucleotides that encode the glycine residue and specific endonuclease sites. The fragment was cleaved with Xbal and Notl, purified and linked to a cloning vector, plasmid pAY02641 (containing a pBR322 origin of replication, a kanamycin resistance gene and a T7 promoter for expression of the gene of interest), restricted with the same enzymes. The ligation, transformation and sequence verification were carried out as described above. Protein expression 43/74 The albumin-binding polypeptide variants were expressed in E. coli BL21 (DE3) with an MGSS N-terminal extension or with a His 6 N-terminal tag followed by a TEV-protease recognition site and a glycine residue. One colony of each variant of albumin-binding polypeptide was used to inoculate 4 ml of TSB + YE medium supplemented with kanamycin at a concentration of 50 pg / ml. Cultures were grown at 37 ° C for approximately 5 hours. 3 ml of each culture was used to inoculate 800 ml of TSB + YE supplemented with kanamycin at a concentration of 50 pg / ml in parallel bioreactors (Greta system, Belach Bioteknik AB). Cultures were performed at 37 ° C, with aeration at 800 ml / minute and a stirring profile to maintain dissolved oxygen levels above 30%, at an OD600 of 2, which was followed by the addition of IPTG to a concentration end of 0.5 mM. Cultivation was continued for five hours, after which cultivation was cooled to 10 ° C, aeration was stopped and agitation was reduced to 300 rpm. Cell pellets were harvested by centrifugation (15,600 xg, 4 ° C, 20 minutes) and stored at -20 ° C until purification. Purification of albumin-binding polypeptide variants with a His 6 tag and a TEV-protease site Frozen cell pellets that store soluble hexahistidine-labeled polypeptides PEP07913 (SEQ ID NO: 153), PEP07912 (SEQ ID NO: 156), PEP07914 (SEQ ID NO: 158), PEP07968 (SEQ ID NO: 159), PEP07986 (SEQ ID NO: 163) and PEP08185 (SEQ ID NO: 148) were suspended in 35 ml of binding buffer (20 mM sodium phosphate, 0.5 M NaCl, 20 mM imidazole, pH 7.4) with an addition of 1000 U of Benzonase® (1.01654.001, Merck) and broken by ultrasonication. For each of the polypeptides, the ultrasonic suspension was clarified by centrifugation (1 hour, 37,000 x g, 4 ° C) and the supernatant i · TM was loaded onto a His GraviTrap column 44/74 (11-0033-99, GE Healthcare). The column was washed with 10 ml of washing buffer (20 mM sodium phosphate, 0.5 of 14 NaCl, 60 mH imidazole, pH 7.4), before eluting the polypeptide with 3 ml elution buffer (20 ml4 of sodium phosphate, 0.5 M NaCl, 0.5 M imidazole, pH 7.4). The buffer was changed to a divage buffer (50 mM Tris-HCl, 150 mM NaCl, pH 8) using a PD-10 desalination column (17-085101, GE Healthcare). The amount of polypeptide product was determined by measuring the absorbance at 280 nm before adding DTT to a final concentration of 5 mM. TEV protease with His s tag was added to the divage buffer at a mass ratio of 1:10 to the polypeptide product. The divage was carried out overnight under slow mixing at 4 ° C. Imidazole was added to the mixture divagem at a concentration of 20 mM before loading the mixture onto a His GraviTrap ™ column (11-0033-99, GE Healthcare) to remove His tags are cleaved, TEV protease with His6 tag and product not cleaved with Hisg label. For each variant, the stream, containing the albumin-binding polypeptide variant, was further purified by reverse phase chromatography (RPC), as follows. The flow fraction was loaded into 1 ml of RPC Resource 15 column (GE Healthcare), previously equilibrated with RPC A Buffer (0.1% TFA in water). After washing the column with 10 column volume (CV) RPC A Buffer, bound polypeptides were eluted with a linear gradient from 0 to 50% RPC B Buffer (0.1% TFA in acetonitrile) over 10 CV. The flow rate was 2 ml / minute and the absorbance at 280 nm was monitored. Fractions containing albumin-binding polypeptide variant were identified by SDS-PAGE analysis and clustered. The albumin-binding polypeptide variants purified by RPC were further purified by 45/74 gel filtration in 120 ml Superdex 75 (GE Healthcare) packed in an XK16 column (GE Healthcare). The current buffer was IxPBS, and the flow rate was 2 ml / minute. Fractions containing pure albumin-binding polypeptide variant were agglomerated and concentrated to approximately 1.3 mg / ml. Finally, the concentrate was purified from trace amounts of remaining endotoxins using 1 ml columns of AffinityPak Detoxi-Gel Endotoxin removal gel (Pierce, prod # 20344), according to the manufacturer's recommendations. The albumin-binding polypeptide variants PEP07911 and PEP08185 were conjugated to Mal-DOTA prior to the RPC purification step, as follows. The buffer of the flow fraction passing through the IMAC-FT purification step was exchanged for 0.2 M NaAc, pH 5.5, using a disposable PD-10 desalination column (GE Healthcare). Maleimido-mono-amide-DOTA (Macrocyclic, cat. No. B-272) was added in molar excess of 5 times and incubated for 60 minutes at 30 ° C with continuous shaking. The resulting polypeptides were denoted PEP07968 and PEP08296, respectively. Purification of His 6 unlabeled albumin-binding polypeptide variants THE pellets of frozen cells that harbor variants in polypeptide albumin binding soluble PEP06923 (SEQ ID NO: 154), PEP07271 (SEQ ID NO: 155), PEP07554 (SEQ ID NO: 156) and PEP07844 (SEQ ID NO: 161) were suspended in 20 mN Tris-HCl, pH 8 and disrupted by ultrasonication. For each of the polypeptide variants, the ultrasonic suspension was clarified by centrifugation (30 min, 32,000 x g, 4 ° C) and the supernatant was loaded onto an HSA-Sepharose column (GE Healthcare). After washing with TST buffer (25 mM Tris-HCl, 1 mM EDTA, 200 mH NaCl, Tween 20 at 46/74 0.05%, pH 8.0), followed by 5 mN NH 4 Ac, pH 5.5, the bound albumin-binding polypeptide variant was eluted with 0.5 M HAc, pH 3.2. The albumin-binding polypeptide variants were further purified by reverse phase chromatography (RPC), as follows. For each of the variants, the eluate from the HSA affinity purification step was loaded onto a 1 ml Resource 15 RPC column (GE Healthcare), previously equilibrated with RPC Buffer A (0.1% TFA in water). After washing the column with 10 CV RPC A Buffer, the bound polypeptides were eluted with a linear gradient of 0 to 50% RPC B Buffer (0.1% TFA in acetonitrile) over 10 CV. The flow rate was 2 ml / min and the absorbance at 280 nm was monitored. Fractions containing pure albumin-binding polypeptide variants were identified by SDS-PAGE analysis and pooled. Finally, the buffer was changed to IxPBS (2.68 mM KC1, 137 mM NaCl, 1.47 mM KH 2 PO 4 , 8.1 mM Na 2 HPO 4 , pH 7.4) using a disposable PD-10 desalination column (GE Healthcare). Characterization of purified albumin-binding polypeptide variants The concentration was evaluated by measuring the / s ® absorbance at 280 nm using a NanoDrop ND-1000 Spectrometer. The proteins were further analyzed with SDS-PAGE and LC-MS. For SDS-PAGE analysis, approximately 10 pg of each variant of albumin-binding polypeptide was mixed with NuPAGE LDS Sample Buffer (Invitrogen), incubated at 70 ° C for 15 minutes and loaded in NuPAGE 4 Gels at 12% Bis -Tris (Invitrogen). The gels were activated with NuPAGE MES SDS Continuous Buffer (Invitrogen) in an XCell II SureLock Electrophoresis Cell (Novex) that used the Rigid Pre-staining Standard (Invitrogen) as a weight marker 47/74 molecular and that used PhastGel BlueR (GE Healthcare) for staining. To verify the identity of the albumin-binding polypeptide variants, LC / MS analyzes were performed using the Agilent 1100 LC / MSD system, equipped with API-ESI and a single quadruple mass analyzer. Approximately 10 pg of each of the purified albumin-binding polypeptide variants was loaded onto a Zorbax 300SB-C8 Narrow Hole column (2.1 x 150 mm, 3.5 pm, Agilent Technologies) at a flow rate of 0 , 5 ml / min. Polypeptides were eluted using a linear gradient of 10 to 70% solution B for 15 minutes at 0.5 ml / min. The separation was carried out at 30 ° C. The ion signal and absorbance at 280 and 220 nm were monitored. The molecular weights of the purified albumin-binding polypeptide variants were confirmed by MS. Results The expression levels of the albumin-binding polypeptide variants were 10 to 30 mg of product / g of cell pellet, as estimated from the SDS-PAGE analysis. For all variants, the purity, as determined by SDS-PAGE analysis, exceeded 95% and the LC / MS analysis verified the correct molecular weights. After purification, between 1 and 8 mg of pure polypeptide were obtained for each of the ten variants of albumin binding polypeptide. Example 2: Affinity determination for albumin-binding polypeptides In this example, PEP06923 (SEQ ID NO: 154), PEP07271 (SEQ ID NO: 155), PEP07844 (SEQ ID NO: 161), PEP07912 (SEQ ID NO: 157), PEP07913 (SEQ ID NO: 153), PEP07914 ( SEQ ID NO: 158) and PEP07968, (PEP07911 conjugated to DOTA, SEQ ID NO: 159), 48/74 synthesized or expressed and purified in Example 1, were characterized by affinity to human serum albumin (HSA) using a Biacore instrument. PEP07913 corresponds to the amino acid sequence of G148-GA3 with the addition of an N-terminal glycine residue, while PEP07271, PEP07844, PEP07912, PEP07914 and PEP07968 correspond to the albumin-binding polypeptides of PP001 (SEQ ID NO: 1), PP043 (SEQ ID NO: 43), PP007 (SEQ ID NO: 7), PP013 (SEQ ID NO: 13) and PP037 (SEQ ID NO: 37) with different N-terminal amino acid additions. Material and methods The biosensor analysis in a Biacore2000 instrument (GE Healthcare) was performed with HSA (Albucult®, Novozymes), immobilized by coupling amine to the carboxylated dextran layer on the surfaces of CM-5 fragments (research degree; GE Healthcare) according with the manufacturer's recommendations. The fragment's surface 1 was activated and deactivated as a reference cell (blank surface) during the injections, and while surface2 comprised HSA immobilized at 731 resonance units (RU) and surface 4 comprised HSA immobilized at 955 RU. The purified albumin-binding polypeptide variants were diluted in 2.5 nM HBS-EP (GE Healthcare) buffer 10nMe40nMe injected at a constant flow rate of 50 μΐ / min for 5 minutes, followed by injection of HBS- EP for 60 minutes. The surfaces were regenerated with an injection of 25 μΐ of HCl, 10 mM. Affinity measurements were performed in two sets; in the first HBS-EP set, PEP06923, PEP07271, PEP07912, PEP07913, PEP07914 and PEP07968 were injected (fragment surface 2) and in the second HBS-EP set, PEP06923, PEP07844, PEP07912 and PEP07914 were injected (fragment surface 4). PEP06923 was injected twice in each cycle as a control. The results were analyzed using BiaEvaluation software (GE Healthcare). At 49/74 curves of the blank surface were subtracted from the curves of the binder surfaces. Results The Biacore 2000 instrument has a technical limitation, hindering measurements of very high affinity. Therefore, the purpose of the Biacore study was not to determine the exact kinetic parameters of the affinity of albumin-binding polypeptide variants for HSA. However, the results provide a quantitative estimate of the relative affinities of these polypeptides for albumin. After subtracting the reference surface and buffer injection, the curves were fitted to a 1: 1 connection model (Langmuir) using the BIAevaluation software with correction for mass transfer and with the RUmax set as a local parameter. The curves were shown in Figure 2. The relative values K Dz k a (k on ) and k d (k off ) were estimated and are shown in the Tables below. Table 1: Kinetic parameters (k a , k d and K D ) of albumin-binding polypeptides for HSA, I the set k a (Ms' 1 ) k d (s' 1 ) K D (M) PEP07913 5.7 x 10 5 9.3 x IO ' 4 1.6 x IO ' 9 PEP06923 (1) 2.9 x 10 7 2.9 x 10 ' 5 9.9 x IO "13 PEP06923 (2) 2.6 x 10 7 2.8 x IO ' 5 1.1 x IO '12 PEP07271 3.9 x 10 6 2.9 x IO ' 5 7.5 x IO '12 PEP07912 4.6 X 10 6 2.8 X IO ' 5 6.2 x IO '12 PEP07914 3.5 x 10 6 2.5 x IO ' 5 7.2 x 10 '12 PEP07968 3.0 x 10 6 2.7 x IO ' 5 9.0 x IO '12 Tabe: La 2: The kinetic parameters (k a, k d and K D) albumin binding polypeptides HSA, all 2 k a (Ms' 1 ) k d (s' 1 ) K D (M) PEP06923 (1) 2.0 x 10 7 2.6 X IO ' 5 1.3 x IO '12 PEP06923 (2) 2.1 x 10 7 2.5 x IO ' 5 1.2 x IO '12 PEP07912 5.4 X 10 s 2.8 X IO ' 5 5.2 x IO '12 PEP07914 3.8 X 10 5 2.6 x IO ' 5 6.9 x 10 '12 50/74 PEP07844 5.4 x 10 s 2.3 x 10 ' 5 4.4 x IO '12 As shown in Tables 1 and 2, PEP07271 (SEQ ID NO: 155), PEP07844 (SEQ ID NO: 161), PEP07912 (SEQ ID NO: 157), PEP07914 (SEQ ID NO: 158) and PEP07968 (PEP07911 conjugated to DOTA, SEQ ID NO: 159) all appear to have approximately the same affinity for HSA, greatly exceeding the affinity of parent G148-GA3 (PEP07913; SEQ ID NO: 153). The HSA affinity of these polypeptides is slightly lower compared to PEP06923 (SEQ ID NO: 154), despite similar dissociation. Example 3: Determination of the melting temperature (Tm) for albumin binding polypeptides In this example, the albumin-binding polypeptide variants PEP07913 (SEQ ID NO: 153), PEP06923 (SEQ ID NO: 154), PEP07271 (SEQ ID NO: 155), PEP07554 (SEQ ID NO: 156), PEP07912 (SEQ ID NO: 157), PEP07914 (SEQ ID NO: 158), PEP07968 (PEP07911 conjugated to DOTA, SEQ ID NO: 159), PEP07844 (SEQ ID NO: 161), and PEP07986 (SEQ ID NO: 163), expressed and purified as described in Example 1, and the albumin polypeptide variant PEP07975 (PEP07834 conjugated to DOTA, SEQ ID NO: 160), produced as described in Example 5, were analyzed by CD analysis. PEP07913 corresponds to the G148-GA3 sequence that has an N-terminal glycine residue, PEP06923 is a high affinity derivative designed by Jonsson et al, supra, while PEP07271, PEP07554, PEP07912, PEP07914, PEP07968, PEP07844 and PEP07975 are Examples of the albumin-binding polypeptides of PP001 (SEQ ID NO: 1), PP007 (SEQ ID NO: 7), PP013 (SEQ ID NO: 13), PP037 (SEQ ID NO: : 37) and PP043 (SEQ ID NO: 43) which have different amino acid additions of N-terminal amino according to the present disclosure. Material and methods 51/74 The purified albumin-binding polypeptide variants were diluted in IxPBS, to final concentrations between 0.4 and 0.5 mg / ml. The analysis of circular dichroism (CD) was performed in a Jasco J-810 spectropolarimeter in 5 cells with an optical path length of 1 mm. In variable temperature measurements, absorbance was measured at 221 nm from 20 ° C to 90 ° C, with a temperature gradient of 5 ° C / min. Results The melting temperatures (Tm) of the different variants of albumin-binding polypeptide were calculated to determine the midpoint of the transition on the CD vs. temperature plotting. The results are summarized in Table 3 below. Table 3. Tm values determined from albumin-binding polypeptide variants tested Variant SEQ ID NO n ° N-terminal sequence 3 Tm (° C) PEP07913 SEQNO: 153 ID GL 61 PEP06923 SEQNO: 154 ID GSSL 57 PEP07271 SEQNO: 155 ID GSSL 65 PEP07554 SEQNO: 156 ID GSSL 58 PEP07912 SEQNO: 157 ID GL 53 PEP07914 SEQNO: 158 ID GL 59 PEP07968 SEQ NO: 159 1 ID GL 53 PEP07975 SEQ ΝΟ ^ όΟ 1 ' 2 ID AL 50 PEP07844 SEQNO: 161 ID GSSL 65 PEP07986 SEQNO: 163 ID GSL 61 77 Õ peptide is conjugated with maleimide-DOTA in cysteine 52/74 2) The peptide is amidated at the C-terminus 3) Leucine (underlined) is the residue at position 1 of the amino acid sequence of the albumin-binding polypeptide as defined in the first aspect of the present disclosure The PEP07968 polypeptide is identical to PEP07912, except that the former has a cysteine residue at position 14 conjugated to DOTA maleimide and the latter a serine residue. Thus, modification of DOTA should not affect the melting temperature. In addition, PEP07975 is conjugated to DOTA with the use of Ci4 and is identical to PEP07968 except for the C-terminal amide (resulting from the peptide synthesis in Example 5) and because it has an N-terminal alanine instead of a glycine. In addition, comparing PEP07912 and PEP07554 reveals that a Nterminal serine generates a higher melting temperature than a glycine in the same position (difference of 5 ° C in Tm). Thus, all variants of albumin-binding polypeptide according to the present disclosure show Tm above 55 ° C, except for PEP07912 and DOTA-conjugated variants. Taking into account the importance of the N-terminal portion, all tested albumin-binding polypeptides are superior to that derived from the prior art of Jonsson et al, that is, PEP06923. Example 4: Serum response analysis The percentage of IgG containing human serum, which can bind to a set of albumin-binding polypeptides as disclosed in the present invention was analyzed by ELISA. In total, 149 serum samples corresponding to 127 individuals were screened. Material and methods ELISA plates (96 area half-well plates (Costar, cat. N ° 3690)) were coated with 50 μΐ / well of Albucult® 53/74 (Novozymes) diluted to 8 pg / ml in coating buffer (Sigma, cat. No. 3041). The plates were coated overnight for three days at 4 ° C. On the day of analysis, the plates were washed twice with tap water and blocked for 2 hours with 100 μΐ of phosphate buffered saline (PBS) containing 0.05% casein (PBSC). The plates were emptied and 50 μΐ / well of the albumin-binding polypeptides PEP07913 (SEQ ID NO: 153), PEP06923 (SEQ ID NO: 154), PEP07271 (SEQ ID NO: 155), PEP07912 (SEQ ID NO: 157) , PEP07554 (SEQ ID NO: 156), PEP07914 (SEQ ID NO: 158), PEP07968 (PEP07911 conjugated to DOTA, SEQ ID NO: 159) and PEP07844 (SEQ ID NO: 161), diluted to 2 pg / ml in PBSC , were added according to a pre-produced plate design. After incubation for two hours at room temperature (RT), the plates were washed in PBSC four times using an automatic ELISA washer. The 149 serum samples from 129 subjects were diluted 50 times in PBSC by adding 24 μΐ of serum to 1174 μΐ of PBSC. 50 μΐ of the diluted serum was added per well according to the pre-produced plate design. Each serum sample was tested as a singlet. Positive and negative controls were included on each plate and for each albumin-binding polypeptide. Albumin binding antibodies (50 μΐ, 0.5 μΐ / ml of immunoglobulin solution prepared internally from serum of primates immunized with PEP06923) were added as a positive control and 50 μΐ of PBSC was used as a negative control. The plates were incubated for one hour at RT and subsequently washed four times in PBSC using an automatic ELISA washer. Bound IgG was detected with 50 μΐ / well of anti-human IgG (Southern Biotech, cat # 2040-05) diluted 10,000 times in PBSC. After washing four times in PBSC using an automatic ELISA washer, 50 μΐ / well of substrate was added (Pierce cat. No. 34021). The reaction was stopped after 10 to 15 minutes by 54/74 adding 50 μΐ of H 2 SO 4 to each well, before measuring absorbance using a multi-well plate reader (Victor3, Perkin Elmer). Results Of the 149 sera screened for IgG binding to albumin binding polypeptides, 23 were negative for all eight polypeptides (OD value <0.1), that is, they did not show IgG bound to the polypeptides. The analysis was performed with 126 sera that were positive for one or more albumin-binding polypeptides. The average absorbance was calculated (Figure 3A) and the percentage of sera with an OD value of <0.15 (Figure 3B) or> 1.0 (Figure 3C). The highest mean OD value and the highest percentage of IgG-bound serum were obtained with PEP07913 (SEQ ID NO: 153), PEP06923 (SEQ ID NO: 154) and PEP07844 (SEQ ID NO: 161), while the lowest reactivity was found against PEP07968 (PEP07911 conjugated to DOTA, SEQ ID NO: 159), PEP07914 (SEQ ID NO: 158) and PEP07954 (SEQ ID NO: 156). Thus, the most reactive albumin-binding polypeptides were the parental G14 8-GA3 (PEP07 913, SEQ ID NO: 153) and the previously enhanced affinity derivative (PEP06923, SEQ ID NO: 154), which has helix 1 retained from G148-GA3. The third of the most reactive polypeptides (PEP07844, SEQ ID NO: 161) contains the original lysine at position 14 in helix 1. The residue is intended for conjugation and will therefore not be exposed in context. The identical albumin-binding polypeptide variant, except for having an alanine at position 14 (PEP07554, SEQ ID NO: 156), is one of the least reactive. Example 5: Chemical synthesis of an albumin-binding polypeptide conjugated to DOTA Material and methods Albumin binding polypeptide PEP07834 (SEQ 55/74 ID NO: 160) was synthesized by solid phase peptide synthesis (SPPS, as described by Quibell, M. & Johnson, T., in Fmoc Solid Phase Peptide Synthesis-A Practical Approach, WC Chan, PD White Eds, Oxford University Press 2000, 115 to 135) in a 433 A Peptide Synthesizer reactor (Applied Biosystems, Foster City, CA) on a 0.1 mmol scale, that is, with a possible theoretical yield of 0.1 mmol of peptide, with the use of standard Fmoc chemistry. A weak acid Fmoc amide resin was used as a solid support throughout the synthesis (Rink Amide MBHA Resin LL (100 to 200 mesh), carrying 0.39 mmol of amide / g of resin (Novabiochem)). According to the sequence below, 47 amino acid residues were coupled to the amide resin by the acylation reactions in the reactor for 10 minutes at room temperature (RT) and mixing. The acylation reactions were carried out with a 10-fold molecular excess of Fmoc protected amino acids in NMP (N-methylpyrrolidone, Merck), activated with 1 eq of 2- (IH-benzotriazol-l-yl) -1,1 hexafluorophosphate, 3,3 tetramethylamino (HBTU, IRIS Biotech), 1 eq of 1hydroxybenzotriazole (HOBt, IRIS Biotech) and 2 eq of diisopropylethylamine (DIEA, Applied Biosystems). In addition, all reactive amino acid side chains were protected with standard side chain protecting groups (tert-butyl (tBu) for Asp, Glu, Ser, Tr and Tir, tert-butyloxycarbonyl (Boc) for Lis, 2,2,4 , 6,7 pentamethyldihydrobenzofuran-5-sulfonyl (Pbf) for Arg, and trityl (Trt) for Asn and Cis) before activation and coupling. In order to decrease the amount of incomplete couplings that lead to truncated peptides, a smaller amount of selected amino acid residues were subjected to acylation coupling twice, without Fmoc deprotection as described below between the first and the second couplings. The amino acid sequence of the albumin-binding polypeptide 56/74 synthesized PEP07834 was ALASAKEAAN AELDCYGVSD FYKRLIDKAK TVEGVEALKD AILAALPNH 2 (SEQ ID NO: I6O-NH2). The underlined amino acid residues were doubly coupled. Any remaining unreacted amino groups in the resin-bound peptides were covered with acetic anhydride (0.5 M acetic anhydride (AlfaAesar), 0.125 M DIEA, 0.015 M HOBt in NMP) for 5 minutes. After each coupling, the deprotection of the N-terminal Fmoc group in the resin-bound peptides was performed by treatment with 20% piperidine (Sigma-Aldrich) in NMP for 10 minutes. After the synthesis was completed, the peptides were cleaved from the solid support and simultaneously the side chain protection groups were cleaved by treatment with TFA / EDT / H2O / TIS (94: 2.5: 2.5: 1) (TFA: trifluoroacetic acid (Apollo) , EDT: 1,2-ethanedithiol (Aldrich), TIS: triisopropylsilane (Aldrich)) at RT for 2 hours with occasional mixing. After the TFA treatment, the peptides were extracted three times using 20% acetonitrile (Merck) in water and tert-butyl methyl ether (Merck). The aqueous phases were combined, filtered and lyophilized. The crude peptides were analyzed and purified by semi-prepared RP-HPLC (Reprosil GOLD Cl8 300, 250 * 10 mm, 5 pm particle size) and a gradient of 32 to 55% B (A: 0.1% TFA-H2O ; B: 0.1% TFA-CH3CN) for 25 minutes at a flow rate of 2.5 ml min ' 1 , followed by lyophilization. The synthetic yield was determined by calculating the areas integrated under the 220 nm signal peaks from crude analysis on RP-HPLC. The correct molecular weight was verified using liquid chromatography electrospray ionization mass spectrometry (LC-ESI-MS) in an LC / MS 6520 Accurate Mass Q-TOF (Agilent Technologies). THE 57/74 product purity was checked using RP-HPLC (Reprosil GOLD C18 300, 250 * 4.6 mm, 3 pm particle size) using a gradient of 35 to 55% B over 25 minutes at a flow rate of 1.0 ml min ' 1 . Conjugation with DOTA mg of PEP07834-amide (SEQ ID NO: 160-amide) was reduced with 20 mM DTT at 40 ° C for 30 minutes. The excess DTT was removed by buffer exchange on a PD-10 column (GE Healthcare) for 0.2 M ammonium acetate, pH 5.5. The coupling was performed with a 5-fold molar excess of chelator, maleimide-mono-amide-DOTA (Macrocyclics, Cat. N ° B- 72) solution in water (1 mg / ml). The mixture was incubated for 1 hour at 30 ° C with continuous shaking. Purification of unconjugated chelators was performed on a semi-prepared RPC column (Zorbax 300SB C18, 9.4x250 mm, 5 μτη). The degree of coupling of the purified material was analyzed by HPLC-MS on a Zorbax 00SB C8 150 x 2.1 mm, 3.5 pm analytical column. Only PEP07834 conjugated to maleimide-DOTA, denoted PEP07975, was detected by the method. Results Based on the elution profile of the crude material, the synthetic yield of the albumin-binding polypeptide PEP07834-amide (SEQ ID NO: 160-amide) was determined to be 8%. The molecular weight found was 4,952.9 Da, which is in good agreement with the theoretical molecular weight calculated for 4,952.6 Da. When analyzing the purified product, approximately 10 to 15% of the protein was revealed to be a disulfide-bound homodimer (Figures 4 and 5). The binding activity of the DOTA-conjugated peptide (PEP07975) was confirmed as described in Example 2 (data not shown) and the melting temperature determined as described in Example 3. Example 6: 58/74 Immunogenicity testing of albumin-binding polypeptides PEP07913 (SEQ ID NO: 153), PEP07912 (SEQ ID NO: 157), PEP07914 (SEQ ID NO: 158), and PEP07968 (DOTA conjugate PEP07911, SEQ ID NO: 159) were screened for their ability to induce proliferation of T cells in peripheral blood mononuclear cells (PBMC) of 52 Caucasian human subjects (obtained from CRI-Labo Medische Analyze, Gent, Belgium). PEP07913 corresponds to the sequence of G148-GA3 which has an N-terminal glycine residue, while PEP07912, PEP07914 and PEP07968 are Examples of the PP007 albumin-binding polypeptides (SEQ ID NO: 7), PP013 (SEQ ID NO: 13 ) and PP037 (SEQ ID NO: 37) which have different amino acid additions of N-terminal amino according to the present disclosure. Materials and methods PBMCs, prepared according to standard cellular biological methods, were added to a 96-well round bottom plate (Falcon) treated with tissue culture (TC) in an amount of 300,000 PBMCs / well. The cells were stimulated by adding 100 μΐ / well of albumin-binding polypeptides PEP07913, PEP07912, PEP07914 and PEP07968 in AIMV (Invitrogen) medium additionally containing 900 pg / ml (3-fold molar excess) of recombinant human albumin (Albucult® , Novozymes). This corresponded to a final concentration of albumin-binding polypeptide of 30 pg / ml. Stimulation was done eight times, that is, the same albumin-binding polypeptide was added to eight wells in identical amounts and under the same conditions. In the positive control wells, cells were stimulated with either 30 pg / ml Lapa Californian Hemocyanin (KLH, Calbiochem) or 30 pg / ml tetanus toxoid (TT, Statens Serum Institut). In negative control wells, only AIMV medium with or without 900 pg / ml albumin 59/74 was added. Cell proliferation was assessed after seven days of culture using the Alexa Fluor 488 Click-iT EdU flow cytometry assay kit (Invitrogen). 1 μΜ / well of EdU incorporation marker was added on day six. On day seven, the cells were washed, dissociated from the plate, washed again and stained for 30 minutes with the anti-CD3-PerCP reagent (Becton Dickinson) and the anti-CD4Alexa647 reagent (Becton Dickinson). After staining, the cells were washed, fixed (BD cellfix, BD biosciences), permeabilized (using saponin) and stained for EdU by adding the Click-iT reagent according to the manufacturer's protocol (Invitrogen). After the staining was completed, the cells were washed again and analyzed using flow cytometry (FACSCantoII, BD Biosciences). To assess the number of cells in proliferation, a fixed number of fluorospheres (Invitrogen) was added to each well prior to analysis. All staining and washing procedures were performed directly on the 96-well plate. The raw FACSCantoII data were synchronized hierarchically in CD3 + CD4 + T cells and the number of cells synchronized as well as their fluorescence intensity of the EdU-Alexa Flour 488 embedding marker were recorded. The average values of the number of cells in proliferation / octuplicate of wells treated with protein were compared to the positive and negative controls and the resulting ratios, described as stimulation indices (SI), were calculated. Based on the SI and the variation between the replicates, the SI limit values were set at 2.0 and 0.5 for stimulation and inhibition, respectively. Results The albumin-binding polypeptides PEP07913, PEP07912, PEP07914 and PEP07968 were evaluated for their 60/74 immunogenic potential in the presence of a 3-fold excess of recombinant human albumin in a target human population using an in vitro PBMC proliferation assay. In comparison to the albumin control PEP07913 induce proliferation of CD3 + CD4 + cells from 52 donors 6, PEP07 912 in 5 of 52 donors and PEP07914 PEP07968 and in 1 of 52 donors (Figure 6A). The mean stimulation index (SI) for all 52 donors was not significantly different for PEP07914 and PEP07968 compared to the negative control that contained recombinant human albumin (p = 0.79 and 0.48, respectively, Figure 6B). The SI for PEP07913 was significantly higher (p = 0.002) while the SI for PEP07912 was higher, but not significant (p = 0.03, Figure 6B). As compared to the buffer only, the number of individuals who responded was 10 for PEP07912, 7 for PEP07912, 2 for PEP07914, 1 for PEP07968, 2 for recombinant human albumin, and 49 and 51 for the two positive controls TT and KLH, respectively (Figure 6C). Albumin-binding polypeptides were classified according to their immunogenicity in the following order: PEP07913> PEP07912> PEP07914> PEP07968. Both PEP07914 and PEP07968 were defined as non-immunogenic. The above results thus demonstrate that the immunogenic potential of the albumin-binding polypeptides of the present disclosure is low, compared to the positive controls. Example 7: Affinity of albumin-binding polypeptides to albumin of different species In this example, PEP06923 (SEQ ID NO: 154), PEP07986 (SEQ ID NO: 163) and PEP08296, (PEP08185 conjugated to DOTA, SEQ ID NO: 148), expressed and purified as described in Example 1, were characterized by affinity to albumin from 61/74 humans (HSA), cynomolgus monkey (CSA), rat (RSA), mouse (MSA) and dog (DSA) using a Biacore instrument. Material and methods The biosensor analysis in a Biacore2000 instrument (GE Healthcare) was performed with HSA (Albucult®, Novozymes), CSA (internally purified from cynomolgus serum), RSA (Sigma-Aldrich, Cat. N ° A6272), MSA (SigmaAldrich, Cat. No. A3559) and DSA (MP Biomedicals, Cat. No. 55925), immobilized by coupling amine to the carboxylated dextran layer on the surfaces of CM-5 fragments (research grade; GE Healthcare) according to the recommendations of the manufacturer. In fragment 1, surface 1 was activated and deactivated and used as a reference cell (blank surface) during injections, while surface 2 comprised HSA immobilized at 1,130 resonance units (RU), surface 3 comprising immobilized CSA at 1,046 RU, surface 4 comprised RSA immobilized at 831 RU. In fragment 2, surface 1 was used as a blank surface, while surface 3 comprised MSA immobilized at 858 RU. In fragment 3, surface 1 was used as a blank surface, while surface 2 comprised DSA immobilized at 1,053 RU. For the affinity analysis for HSA, CSA, and RSA (fragment 1), the purified albumin-binding polypeptide variants were diluted in continuous buffer HBS-EP (GE Healthcare) at 40 nM, 10 nM and 2.5 nM; for affinity analysis for MSA (fragment 2) the albumin-binding polypeptide variants were diluted to 1,280 nM, 640 nM, 160 nM and 40 nM and for affinity analysis for DSA (fragment 3) the polypeptide-binding variants albumin were diluted to 1,280 nM, 640 nM, 160 nM, 40 nM and 10 nM. The albumin-binding polypeptides were injected into a 62/74 constant flow rate of 50 μΐ / min for 5 minutes, followed by injection of HBS-EP for 60 minutes. The surfaces were regenerated with an injection of 25 μΐ of HCl, 10 mM. All samples were run in duplicates. The results were analyzed using BiaEvaluation software (GE Healthcare). The white surface curves were subtracted from the binder surface curves. Results Biacore 2000 instrument has a technical limitation, hindering measurements of very high affinity. Therefore, the purpose of the Biacore study was not to determine the exact kinetic parameters of the affinity of albumin-binding polypeptide variants for HSA, CSA, RSA, MSA and DSA respectively. However, the results provide a quantitative estimate of the relative affinities of these closed polypeptides for albumin of different species. After subtracting the reference surface and buffer injection, the curves were fitted to a 1: 1 connection model (Langmuir) using the BIAevaluation software with correction for mass transfer and with the RUmax set as a local parameter. Representative connection curves are shown in Figure 7. PEP07986 and PEP08296 (PEP08185 conjugated to DOTA) bind with high affinity (K D in the range from below picomolar to below nanomolar) to human serum albumin as well as to the albumin of the pre-clinical model species frequent rat, cynomolgus monkey, mouse and dog . The relative affinities for the different species can be classified as RSA> HSA / CSA> MSA / DSA, that is, the values of K D classified as K d .r S to K d - H sa / K d -csa <K d _ MS a / K d - DS a · The affinities in terms of K D values are the same or slightly lower (but in the same order of magnitude) as the affinity obtained for PEP06923 (polypeptide not 63/74 inventive). Example 8: In vitro activity of protein Z variants fused to an albumin-binding polypeptide In this example, cytokine-specific Z protein variants comprising polypeptides (derived from the B-domain of staphylococcal protein A) genetically fused to the albumin-binding polypeptide variant PP013 (SEQ ID NO: 13) have been tested for their functionality, being blocked to Cytokine-induced proliferation of TF-1 cells in the presence of human serum albumin. The proliferation of TF-1 cells is dependent on the presence of any one of several different types of cytokines and the proliferative response can be inhibited by blocking reagents such as the corresponding cytokine-specific protein Z variant. PP013 fused to a protein Z variant with specificity for an irrelevant protein was used as a negative control. Materials and methods Z cloning - PP013 fusion proteins The gene fragments of Z protein variants with specificity for cytokine X or Y respectively, or for an irrelevant protein (negative control), were amplified by PCR using primers that add specific PstI and AccT endonuclease sites. The fragments were cleaved with PstI and Accl, purified and ligated into an expression vector, plasmid pAY02747, restricted with the same enzymes. pAY02747 contains a pBR322 origin of replication, a kanamycin resistance gene and a T7 promoter for expression of the gene of interest. The cloning site is preceded by a sequence encoding the amino acids MGSSLQ and followed by a sequence encoding VDSS-PP013, where PP013 is the albumin-binding polypeptide revealed with the 64/74 SEQ ID NO: 13. The ligation, transformation and sequence verification were carried out as described above. The encoded proteins were: 1) MGSSLQ-Z x -VDSS-PP013 (denoted Z x -PP013) 2) MGSSLQ-ZY-VDSS-PP013 (denoted ZY-PP013) 3) MGSSLQ-Zneg-VDSS-PP013 (denoted Zneg-PP013) Protein expression Z x -PP013, Zy-PP013 and Zneg-PP013 were expressed in E. coli BL21 (DE3) cells. The transformation colonies of each fusion variant were used to inoculate the 50 ml starting cultures of TSB + YE medium supplemented with kanamycin to a concentration of 50 pg / ml. The cultures were grown at 37 ° C overnight with agitation, 100 rpm. The starting cultures were then used to inoculate 900 ml of TSB + YE medium supplemented with kanamycin to a concentration of 50 pg / ml. Cultures were grown for approximately 1.5 h and an OD600 of> 1.1, whereby IPTG was added to a final concentration of 0.2 mM. The cultivation continued for five hours. Cell pellets were harvested by centrifugation (15,600 g, 4 ° C, 20 minutes) and stored at -20 ° C until purification. Protein purification The frozen cell pellets housing Z x -PP013, Z Y -PP013 and Z neg -PP013 soluble fusion protein variants were resuspended in 50 mM Tris-HCl, 150 mM NaCl, pH 8 and 1000 U Benzonase * ( Merck Cat. No. 1,01654,0001) has been added. The cells were disrupted by ultrasonication and for each of the fusion protein variants, the ultrasonic suspension was clarified by centrifugation (15 min, 37,000 g, 4 ° C). 20x TST buffer (20x [25 mM Tris-HCl, 1 mM EDTA, 200 mM NaCl, 0.05% Tween 20, pH 8.0]) was added to a resulting volume in 1 x TST buffer in the suspension clarified. Each sample of the fusion protein variant was 65/74 loaded on an HSA-Sepharose column (GE Healthcare). After washing with TST buffer, followed by 5 mM NH 4 Ac, pH 5.5, the bound fusion protein variant was eluted with 0.5 M HAc, pH 2.5. The fusion protein variants were further purified by reverse phase chromatography (RPC), as follows. For each of the variants, the eluate from the HSA affinity purification step was loaded onto a 1 ml Resource 15 RPC column (GE Healthcare), previously equilibrated with RPC Buffer A (0.1% TFA in water). After column washing with 10 CV of RPC Buffer A and 5 CV of RPC B Buffer (0.1% TFA in acetonitrile), the bound fusion proteins were eluted with a linear gradient of 10 to 50% RPC B Buffer for 20 CV. The flow rate was 2 ml / min and the absorbance at 280 nm was monitored. Fractions containing pure fusion protein variants were identified by SDS-PAGE analysis and pooled. Finally, the buffer was changed to IxPBS (2.68 mM KC1, 137 mM NaCl, 1.47 mM KH 2 PO4, 8.1 mM Na 2 HPO4, pH 7.4) using a column disposable PD-10 desalination system (GE Healthcare). To verify the identity of the fusion protein variants, SDS-PAGE and LC / MS analyzes were performed as described in Example 1. In vitro cell assay for Z - PP013 fusion proteins The TF-1 cell line (CLS Cat. No. 300434) was propagated as recommended by the supplier in RPMI 1640 medium + 10% fetal calf serum (Gibco) with the addition of 2 ng / ml rhGM-CSF (Miltenyi) . On the day of the experiment, the cells were washed in RPMI 1640 + 10% fetal calf serum to remove GM-CSF. The ability of Z x -PP013 and Zy-PP013 to block cytokine-induced proliferation was analyzed by mixing 66/74 of the molecules Z x -PP013, Z Y -PP013 and Z ne g-PP013 with cytokines X and Y respectively and with a five-fold molar excess of HSA (Albucult®, Novozymes). The molecules were titrated in a series of two-fold dilutions with a fixed concentration of cytokine (4.9 pM) and a five-fold molar excess of HSA. Titration was carried out in 96-well plates in a volume of 100 μΐ. 25,000 cells were added per well (100 μΐ) and the plates were incubated at 37 ° C, 5% CO 2 for three days. To measure proliferation, 19 μΐ of CCK-8 cell proliferation reagent (Sigma) diluted twice in RPMI 1640 medium + 10% fetal calf serum, was added per well. The color reaction was monitored after 4 hours using the 96-well plate reader (Victor3; PerkinElmer). Results As shown in Figure 8, both Z x -PP013 and Z Y -PP013 inhibited the respective cytokine-induced proliferation in the presence of HSA while Z neg -PP013, the negative control, did not affect the proliferation of TF-1. Thus, the experiment shows that the function of the Z molecules was maintained when incorporated into a fusion protein that contains the albumin-binding polypeptide and also when the fusion proteins were bound to albumin. Example 9 Long-term stability of an albumin-binding polypeptide In this example, the stability of PEP07986 (SEQ ID NO: 163), expressed and purified as described in Example 1, was investigated after storage at 4, 25, and 40 ° C for up to three months. The state of the polypeptide after storage was investigated by measuring its binding to HSA using a Biacore instrument. Material and methods Lyophilized PEP07986 was dissolved in the 67/74 Sterile NaPi (20 mM sodium phosphate, 150 mM sodium chloride, pH 7.2) at a concentration of 2 mg / ml. A reference sample (time = 0) was removed and stored at -80 ° C. The 105 μΐ aliquots were stored in sterile threaded cap 330 eppendorf tubes sealed with parafilm at 4, 25 and 40 ° C. After a week, two weeks, a month and three months, a sample stored at each temperature was cooled to 4 ° C, centrifuged for 5 minutes at 13,000 rpm and then stored at -80 ° C awaiting Biosensor analysis. Biosensor analysis was performed essentially as described in Example 2, but with HSA (Albucult®, Novozymes), immobilized at 704 resonance units (RU) and the albumin-binding polypeptide variant was diluted to 10 nM and injected at constant flow rate of 20 μΐ / minute for 10 minutes, followed by the injection of HBS-EP for 10 minutes. Results The HSA binding of PEP07986 (SEQ ID NO: 163) was retained after storage at 4, 25, and 40 ° C for at least three months. The maximum binding responses for HSA obtained for ο PEP07986 stored under the various conditions are shown in Figure 9. Example 10: Stability of an albumin-binding polypeptide under extreme conditions In this example, the biosensor and circular dichroism (CD) analysis of the albumin-binding polypeptide PEP08296 (PEP08185 conjugated by DOTA, SEQ ID NO: 148) after heat treatment (90 ° C) in low pH buffer (~ 4 , 0) is described. Since such extreme reaction conditions have to be used, for example, for labeling 68 Ga of DOTA-modified proteins, the influence of the low pH and high heat treatment on the structural identity of the polypeptide and its ability to 68/74 binding to HSA was investigated by measuring melting temperature (Tm), refolding properties and binding to HSA. Material and methods Biosensor analysis of heat stability The biosensor analysis in a Biacore 2000 (GE Healthcare) instrument was performed with HSA (Albucult, Novozymes) immobilized by the amine coupling in the carboxylated dextran layer on the surface of the CM-5 fragment (research grade; GE Healthcare) according to manufacturer's recommendations. The fragment's surface 1 was activated and deactivated and used as a reference cell (blank surface) during injections, while surface 2 comprised HSA immobilized at 724 resonance units (RU). Ο PEP08296 (50 μΐ, 100 μg) in a 15 ml Falcon tube was diluted with 450 μΐ of 0.2 M sodium acetate (NaAc) pH 5.5 to a final peptide concentration of 0.2 mg / ml . After adding 1.5 ml of 0.05 M HCl (similar to the conditions and volume used to elute a 68 Ge / 68 Ga generator) the sample was incubated for 10 minutes at 90 ° C or RT (control ) and then transferred to RT. 6 ml of 0.1 M sodium citrate was added to neutralize the pH. The PEP08296 heat treated (0.8 and 4 nM) was injected at a constant flow rate of 50 μΐ / minute for 5 minutes, followed by dissociation in HBS-EP for 15 minutes. The surfaces were regenerated with an injection of 25 μΐ of 10 mM HCl. The results were analyzed using the BIAevaluation software (GE Healthcare). The blank surface curves were subtracted from the binding surface curves. Determination of the melting temperature (Tm) Ο PEP08296 was dissolved in PBS at a final concentration of 0.5 mg / ml. PBS with a pH of approximately 4.0 was prepared by adding 9.5 μΐ of 100 mM HCl to 100 μΐ of PBS. The analysis of circular dichroism (CD) was performed 69/74 as described in Example 3. CD analysis of heat stability To investigate the structural reversibility of PEP08296 after heat treatment, two CD spectra between 195 and 250 were recorded per sample at 20 ° C. After the first spectrum, a VTM cycle with heating at 90 ° C was performed as described above followed by the collection of the second CD spectrum between 195 and 250 nm at 20 ° C. In addition, PEP08296 was incubated in PBS buffer pH 4.0 or PBS buffer pH 7.2 for 12 minutes at 90 ° C in a thermomixer (500 rpm, mixing interval 10 s on, 30 s off). After incubation, the samples were cooled on ice followed by centrifugation at 13,000 rpm for 1 minute and a CD spectrum between 195 and 250 nm was recorded at 20 ° C Results Biosensor analysis was used to investigate whether heat treatment in combination with low pH, that is, common conditions required for the 68 Ga labeling of the polypeptide, would affect the ability of PEP08296 to bind to HSA. Figure 10 shows the result of this binding analysis performed with a Biacore 2000 instrument. Two different concentrations of PEP08296, 0.8 nM and 4 nM, were injected onto a surface with 724 RU of immobilized human serum albumin Heat treatment for 10 minutes at 90 ° C, pH 4.0, slightly reduced the binding capacity of PEP08296 to HSA, indicating a potential structural change of the molecule. CD was used to further investigate the potential structural change of the molecule. Similar CD spectra before and after heating would prove that a sample is structurally reversible. In the first experiment, the samples were heated with a temperature gradient from 20 ° C to 90 ° C. The CD spectra before and after the heat treatment were similar in the Tm determination experiment with 70/74 the typical lows at 207 and 221 nm indicating that ahelicity, that is, short time heating to 90 ° C in either pH 4 or pH 7.2 buffer had no effect on the structure of PEP08296. However, pretreatment of PEP08296 for 12 minutes at 90 ° C showed a slightly reduced alpha helix content of PEP08296 if incubated at pH 4.0, but no change in alpha helix content if incubated at pH 7.2. The typical overlays of the two CD spectra before and after heating are shown in Figure 11. The results of the melting temperature determination (Tm) are summarized in Table 4. Table 4. Tm of PEP08296 Designation Tm (° C) PEP08296 at pH 7.2 59 PEP08296 at pH 4.0 62 Example 11: Radioisotopic ventriculography imaging using a Ga-labeled albumin-binding polypeptide In the experiments that constitute this example, the entire body distribution of Ga-labeled PEP08296 (DOTA conjugated PEP08185, SEQ ID NO: 148) in rats was followed by dynamic imaging over 1.5 hours. Due to the strong association between the labeled polypeptide and serum albumin, the labeled polypeptide can be used, for example, to study tissue permeability and radioisotopic ventriculography. Material and methods 68 Ga labeling of PEP08296 68 Ga was eluted as 68 GaCl3 from the 68 Ge / S8 Ga generator (Obninsk, Russia) with 0.1 M HC1, converted to S8 GaCl4- with concentrated HCl, collected in a column of anion exchange (Chromafix-HCO 3 ) and subsequently eluted as water at 18 ΜΩ, as previously described (Velikyan et al 71/74 (2008), Nucl Med Biol 35: 529 to 536). The labeling was carried out essentially as described in Tolmachev et al. (EJNMMI 37: 1356 to 1367, 2010). The concentrated 68 Ga eluate (150 to 200 μΐ) was added to PEP08296 (£ 100 pg in 0.2 M sodium acetate buffer pH 5.5) and the pH was adjusted to 3.5 to 4 using sodium acetate (1.25 M) or HCl (0.1 M). The labeling mixture was incubated at 90 ° C for 15 minutes before cooling and the labeled protein was isolated by size exclusion purification on a NAP-5 column eluted with physiologically buffered saline. The identity and radiochemical purity of the 68 Ga-labeled protein was evaluated by radio-HPLC using UV (210 nm) and radioactivity detectors in a superdex Peptide 10/300 GL column (GE Healthcare) eluted with physiologically buffered saline . Small animal PET One rat (277 g) was anesthetized with isoflurane (initially 5%, then 2% mixed with air / O 2 7: 3), controlled by an EZ vaporizer using Microflex non-rebreathing masks from Euthanex Corporation and kept in a heating pad (37 ° C) while located within a FocusP20 microPET system (Siemens, CTI Concorde Microsystems). 68 Ga-PEP08296, 33 MBq, was dispensed in a syringe, diluted with 0.5 ml saline and injected through the tail vein. Data were acquired from the entire body by moving the bed in a constant bed movement protocol for 1.5 hours. The data were processed with MicroPET Manager and corrected for random decay and dead time. The images were reconstructed using the standard 2D filtered overhead projection using a ramp filter and evaluated using Research Workplace software (Siemens Medical Solutions). 72/74 Results The basic distribution patterns (Figure 12) for PEP08296 were very similar to those of radioisotope-labeled albumin such as 68 Ga-DOTA, 64 Cu-D0TA and 1: L C (see, for example, Hoffend et al (2005), Nucl Med Biol 32: 287 to 292 and Lu et al (2008), [1- 1: L C] Butanol and [Methyl- 1: L C] Albumin for Blood Flow and Blood Pool Imaging, published in the XIth Turku PET Symposium, May 24-27, 2008). In summary, high concentrations of radioactivity were observed in major blood vessels throughout the scan. The organs with large volumes of blood (liver, spleen and kidney) were also clearly delineated, as was the radioactivity of cardiac radioisotopic ventriculography. The radioactivity in the urinary bladder increased during the observation period, this observation of renal elimination being consistent with previous observations. with labeled albumin based trackers and with that of the albumin metabolism itself. The general distribution pattern of radioactivity and very slow plasma clearance after intravenous injection of 68 Ga-PEP08296 is consistent with its expected strong and very fast binding to albumin. These results, therefore, support additional applications of the radiorastrander as an in vivo radioisotopic ventriculography imaging agent for use with positron emission tomography studies of tissue permeability, both during disease development and during therapeutic intervention. Example 12: Solubility of an albumin-binding polypeptide The solubility of PEP07986 (SEQ ID NO: 163) in the physiological buffer was investigated by consecutive concentrations of the sample using ultrafiltration, followed by concentration measurement and investigation of the aggregation state. Concentrations determined by absorbance readings 73/74 direct at 280 ntn were consistent with the concentrations determined by gel filtration, showing a solubility of more than 42 mg / ml with no aggregate detected. Material and methods Lyophilized PEP07986 was dissolved in the NaPi buffer (20 mM sodium phosphate, 150 mM sodium chloride, pH 7.2) at a concentration of 3 mg / ml. The Amicon Ultra centrifuge filter units, 3 kDa cuts, (Millipore, Cat. NUFC800324) were pre-rinsed with 2 ml of NaPi buffer by centrifugation at 4,000 g for 20 minutes in a mobile bucket rotor centrifuge (Multifuge , Heraeus). 1,620 μΐ of PEP07986 at 3 mg / ml were applied to a first centrifugal filter unit and centrifugation was performed at 4,000 g, 20 ° C, for 7 minutes. A 25 μΐ sample was removed (sample UF 1) for further analysis and the rest of the sample was transferred to a second centrifugal filter unit. Centrifugation and sample removal were repeated three times with spin times of 8, 9 and 20 minutes respectively (sample UF 2, 3 and 4 respectively). Absorbance readings were performed using a NanoDrop® ND-1000 Spectrophotometer and diluting UF samples 1 to 4 in NaPi buffer 2, 4, 6 and 12 times respectively. The concentrations were calculated using the extinction coefficient 1 Abs 280 = 1,955 mg / ml. Gel filtration was performed on an 1100 HPLC system (Agilent Technologies) using a Superdex75 10/300 GL column (GE Healthcare) that was equilibrated in the NaPi buffer. 10 μΐ of each UF sample was applied to the column; the NaPi buffer was used as a continuous buffer and the flow rate was 0.5 ml / minute. A chromatogram of the standard molecular weight ovalbumin (GE Healthcare) injected at a concentration of 5 mg / ml was also collected. Concentrations were determined by integrating the area under the curve. 74/74 Results The concentrations determined by the direct absorbance readings at 280 nm and the concentrations determined by the gel filtration are shown in Table 5. The 5 solubility of PEP07986 (SEQ ID NO: 163) is at least 42 mg / ml in the physiological buffer (phosphate of 20 mM sodium, 150 mW sodium chloride, pH 7.2). No aggregates were detected by gel filtration, as shown in Figure 13. Table 5. Concentrations determined after 10 consecutive concentrations of PEP07986 (SEQ ID NO: 163) Concentrations (mg / ml) determined by Sample Spectrophotometer Gel filtration UF2 12.1 12.4 UF3 22.2 22.1 UF4 42.7 42.6 1/4
权利要求:
Claims (15) [1] 1 . ALBUMIN BINDING POLYPEPTIDE, characterized by comprising an amino acid sequence selected from among i) LAX 3 AKX 6 X7ANX 10 eldx 14 ygvsdf ykrlix 26 kakt VEGVEALKX39X40 ILX43X44LP in which independently of each other X3 is selected among E, S, Q and C; x 6is selected among E, S and C; X7 is selected between A and S; X10 is selected among A, S and R; X14 is selected among A, S, C and K; X26 is selected between D and E; X39 is selected between D and E; X40 is selected between A and E; X43 is selected between A and K; X44 is selected among A, S and E; L at position 45 is present or absent; and P in position 46 is present or absent; and ii) an amino acid sequence that has at least 95% identity to the sequence defined in i). [2] 2 . ALBUMIN BINDING POLYPEPTIDE, according to claim 1, characterized in that it binds to albumin in such a way that the k off value of the interaction is at most 5 x 10 5 s for example, at most 5 x 10 6 s 1 . [3] 3. ALBUMIN BINDING POLYPEPTIDE, according to claim 1, characterized in that the amino acid sequence is selected from any one of SEQ ID NO: 1 to 144 and SEQ ID NO: 164 to 203, for example, selected from from any one of SEQ ID NO: 1 to 144. [4] 4. ALBUMIN BINDING POLYPEPTIDE, according to claim 3, characterized in that the sequence of 2/4 amino acids to be selected from any of SEQ ID NO: 4 to 5, SEQ ID NO: 7 to 8, SEQ ID NO: 10 to 11, SEQ ID NO: 13 to 14, SEQ ID NO: 16 to 17, SEQ ID NO: 19 to 20, SEQ ID NO: 22 to 23, SEQ ID NO: 25 to 26, SEQ ID NO: 28 to 29, SEQ ID NO: 31 to 32, SEQ ID NO: 34 to 35, SEQ ID NO: 37 to 38, SEQ ID NO: 41 to 42 and SEQ ID NO: 49 to 50. [5] 5. ALBUMIN BINDING POLYPEPTIDE, according to any one of the preceding claims, characterized in that it binds to human serum albumin. [6] 6. FUSION OR CONJUGATE PROTEIN, characterized by understanding i) a first chemical moiety consisting of an albumin-binding polypeptide, as defined in any of the preceding claims; and ii) a second chemical moiety consisting of a polypeptide that has a desired biological activity. [7] 7. ALBUMIN, FUSION PROTEIN OR CONJUGATE BINDING POLYPEPTIDE, as defined in any of the preceding claims, characterized by an identification. [8] 8. POLINUCLEOTIDE, characterized by encoding an albumin-binding polypeptide or a fusion protein, as defined in any of the preceding claims. [9] 9. METHOD FOR PRODUCING A POLYPEPTIDE, as defined in any of the preceding claims, characterized in that it comprises the expression of a polynucleotide as defined in claim 8. [10] 10. METHOD FOR PRODUCING A POLYPEPTIDE, as defined in any one of claims 1 to 8 or according to claim 9, characterized by the synthesis of non-biological peptide that uses and / or derivatives of amino amino acids that have protected reactive side chains, being that synthesis 3/4 non-biological peptide comprises stepwise coupling of amino acids and / or amino acid derivatives to form a polypeptide, as defined in any of claims 1 to 5, which has protected reactive side chains, removal of side chain protecting groups reactive polypeptides, and folding the polypeptide in aqueous solution. [11] 11. FUSION PROTEIN OR CONJUGATE, according to claim 6, or as defined in any of claims 7 to 10, characterized by use as a medicament. [12] 12. FUSION PROTEIN OR CONJUGATE, according to any one of claims 6 or 11, or as defined in any one of claims 7 to 10, characterized by use in diagnosis. [13] 13. COMPOSITION, characterized by comprising a compound that alone has a solubility in water of not more than 100 pg / ml; coupled to an albumin-binding polypeptide, a fusion protein or conjugate, as defined in any of claims 1 to 6, wherein the compound and albumin-binding polypeptide, fusion protein or conjugate are covalently linked. [14] 14. METHOD FOR PREPARING A COMPOSITION, as defined in claim 13, characterized by comprising a) provide a compound which alone has a solubility in water of not more than 100 pg / ml; and b) covalently coupling the compound to an albumin-binding polypeptide, fusion protein or conjugate, as defined in any one of claims 1 to 6, thus forming a composition that 4/4 comprises a covalent complex of the compound and an albumin-binding polypeptide, fusion protein or conjugate. [15] 15. METHOD FOR INCREASING THE SOLUBILITY OF A COMPOUND, characterized by understanding to provide a compound that alone has a solubility in water of no more than 100 pg / ml; covalently coupling the compound to an albumin-binding polypeptide, fusion protein or conjugate, as defined in any one of claims 1 to 6, thereby forming a covalent complex of compound and albumin-binding polypeptide; and mixing said complex of albumin-binding compound and polypeptide, fusion protein or conjugated with albumin under conditions that promote non-covalent association of the polypeptide of Link albumin with serum albumin human;through which compound water solubility in said complex it is composed by itself. bigger that the water solubility of 1/19 SEQ ID NO: I δ z O0 U » SEQ ID NO 02 I SEQ ID NO: 3 I I tON OI O3S SEQ ID NO: 5 | I 9 = ON OI Q3S 1 1 hON 01 03S I I SEQ ID NOt8 1 Tl Õ 2Q0 UI m SEQIDNO: 10 | SEQ ID NO: 11 | SEQ ID NO: 12 | 1 εμοΝ οι oas | | SEQ ID NO: 14 | 1 SEQ ID NO: 1S | SEQIDNOrie j | SEQ ID NO: 17 | | SEQ ID NO: 18 | I SEQ ID NO: 19 | I SEQ ID NO: 20 | I SEQ ID NO21 | CU Tl o 2 athe ω UI »No z g 5 uu 40 T1 o 2 aσ ui Ci) I «ON 0103S 1 1 SEQ ID NO ^ 6 | 1 SEQ ID NO: 27 | | SEQ ID NO08 | I 6K-0N 01D3S 1 OC ON ai DIG <r Õ 2 Oσ U1 <0 ¢ 4 m Ô 2 Q 0 UI co | SEQ ID NO: 33 | | SEQ ID NO: 34 | I SEQ ID NO: 35 1 | SEQ ID NO: 36 | Amount of amino acids _______________________________ EASAKEAAHA EU> AYGVSDF XKIÜrXDKftKT VEGVEALEDA IIAALP____________________________ 6iI LASAKESANS ELDAYGVSDF YKRLIDKAKT VEGVEALKDA ILAALP | T.ARAKEEAHA ELDAYGVSDF YKRLIDKAKT VEGVEALKDA ILAALP____________________________ i1 LASAKSAANS ELDAYGVSDF YKRLIDKAKT VEGVEALKDA ILAALP____________________ j LASAKEAANA ELDSYGVSDF YKRLIDKAKT VEGVEALEDA ILAALP | LASAKEAANS ELDSYGVSDF YKRLIDKAKT VEGVEALEDA ILAALP_____________________________1 LASAKESANS ELDSYGVSDF YKRLIDKAKT VEGVEALEDA ILAALP___________________________ LASAKESANA ELDSYGVSDF YKRLIDKAKT VEGVEALKDA ILAALP _____________________________________________________ ___________ | tI i LASAK3AANS ELDSYGVSDF YKRLIDKAKT VEGYEALEDA IIAAU_____________________________j LAEAKEAANA ELDSYGVSDF YKRLIDKAKT VEGVEALKDA ILAALP___________________________ i LAEAKEAANS ELDSYGVSDF YKRLIDKAKT VEGVEALKDA ILAALP I I LAEAKESANS ELDSYGVSDF YKRLIDKAKT VEGVEALKDA ILAALP____________________________J | LAEAKESANA ELDSYGVSDF YKRLIDKAKT VEGVEALKDA ILAALP | 1 LAEAKSÀANA ELDSYGVSDF YKRLIDKAKT VEGVEALKDA ILAALP_____________________________ I LAEAKSAANS ELDSYGVSDF YKRLIDKAKT VEGVEALEDA ILAALP_____________________!| T. AR ACT! IANS ELDAYGVSDF YKRLIDKAKT VEGVEALKDA ILAALP 1 LAEAKESANS ELDAYGVSDF YKRLIDKAKT VEGVEALKDA ILAALP___________________________ j0 I, a * rmti νατπ 3ΑΡ3Λ amoi'mi .aasASXWts vNvvswavi | | LAEAKSÀANS ELDAYGVSDF YKRLIDKAKT VEGVEALKDA ILAALP | 1 LAQAKEAANA ELDAYGVSDF YKRLIDKAKT VEGVEALEDA ILAALP_____________________________1 I LAOAKEAANS EMAYGVSDF YKRLIDKAKT VEGVEALKDA ILAALP___________________________ I LAQAKESANS ELDAYGVSDF YKRLIDKAKT VEGVEALKDA ILAALP ............ 1 I LAQAKESANA ELDAYGVSDF YKRLIDKAKT VEGVEALEDA ILAALP____________________________I [LAQAKSAANA ELDAYGVSDF YKRLIDKAKT VEGVEALEDA ILAALP____________________________I mii vaynraADSA jawagrraax josadasctih wmsreDn I I T.EQRCTiaNS ELDSYGVSDF YKRLIDKAKT VEGVEALEDA ILAALP____________________________ I LAQAKESANS ELDSYGVSDF YKRLIDKAKT VEGVEALEDA ILAALP____________________________1 1 LAQAKESANA ELDSYGVSDF YKRLIDKAKT VEGVEALEDA ILAALP____________________________I I LAQAKSÀANA ELDSYGVSDF YKRLIDKAKT VEGVEALEDA ILAALP _ 1 | dTHfll WÍTV3AD3A. JDíTMaiTEXA dGSADASOTa SNVYSEttOri] o 0) 75 s HP cu (D Λ -H □ I PP0O1 I I PP002 I I 600dd | The£Q_ I PPD05 1 1CL PP007 I ppnns 1 1Q_ 1 OlOdd | I PP011 1 CXJÍ I PP0I3 I I PP014 1 1 PP015 1 PP016 £ CL PP018 PP019 £ £ L &£CL SIg ppuaa £ a. & PPD2Ô 1 I PP027 I 8£ £ L I PPD29! 1CL. PP031 1 PPQ32 I PP033 PPD34 PPD35 secHd 1
类似技术:
公开号 | 公开日 | 专利标题 US20190375799A1|2019-12-12|Polypeptides JP2018183171A|2018-11-22|Albumin binding polypeptide JP6959616B2|2021-11-02|Site-specific RI-labeled antibody with IgG-binding peptide KR20180042442A|2018-04-25|Radiolabeled her2 binding peptides EP3463486A1|2019-04-10|Pet imaging with pd-l1 binding polypeptides Honarvar et al.2013|Evaluation of backbone-cyclized HER2-binding 2-helix Affibody molecule for In Vivo molecular imaging JP6440618B2|2018-12-19|HER3 binding polypeptide AU2018211539A1|2019-07-25|Compositions and methods for cancer imaging and radiotherapy WO2022013376A1|2022-01-20|Serum stable binding proteins for human her2 for theranostic applications WO2021122729A1|2021-06-24|Anti-her2 polypeptides derivatives as new diagnostic molecular probes WO2021122726A1|2021-06-24|Anti-her2 polypeptides derivatives as new diagnostic molecular probes IL284097D0|2021-08-31|Novel folr1 specific binding proteins for cancer diagnosis and treatment
同族专利:
公开号 | 公开日 IL223989A|2018-10-31| HK1209761A1|2016-04-08| JP2013534421A|2013-09-05| LT2933262T|2018-07-10| KR101884654B1|2018-08-02| US20160108095A1|2016-04-21| CN110437320A|2019-11-12| ES2676403T3|2018-07-19| EP2933262B1|2018-05-02| CA2804002C|2021-07-20| WO2012004384A2|2012-01-12| PL2933262T3|2018-10-31| JP5827994B2|2015-12-02| DK2590993T3|2015-06-29| CA2804002A1|2012-01-12| CN110437321A|2019-11-12| TR201809437T4|2018-07-23| US20140162956A1|2014-06-12| NZ604805A|2014-09-26| US10329331B2|2019-06-25| AU2011275766A1|2013-01-10| US20190375799A1|2019-12-12| ZA201300994B|2020-05-27| DK2933262T3|2018-06-25| RU2577964C2|2016-03-20| HK1182719A1|2013-12-06| MX2013000314A|2013-01-29| JP2016053047A|2016-04-14| ES2540114T3|2015-07-08| EP2590993B1|2015-05-06| AU2011275766B2|2015-10-22| CN103003296A|2013-03-27| MY161679A|2017-05-15| EP2933262A1|2015-10-21| JP6486810B2|2019-03-20| KR20130034043A|2013-04-04| RU2013102953A|2014-08-20| EP2590993A2|2013-05-15| NO2933262T3|2018-09-29| PL2590993T3|2015-10-30| WO2012004384A3|2012-03-29| US9211344B2|2015-12-15|
引用文献:
公开号 | 申请日 | 公开日 | 申请人 | 专利标题 SE509359C2|1989-08-01|1999-01-18|Cemu Bioteknik Ab|Use of stabilized protein or peptide conjugates for the preparation of a drug| SE9400088D0|1994-01-14|1994-01-14|Kabi Pharmacia Ab|Bacterial receptor structures| US6362225B1|1999-01-21|2002-03-26|George Andreakos|Target therapies for treating common viral infections| PT1240337E|1999-12-24|2007-01-31|Genentech Inc|Methods and compositions for prolonging elimination half-times of bioactive compounds| US7288265B1|2000-10-16|2007-10-30|Lectec Corporation|Treating viral infection at smallpox vaccination site| US7316819B2|2001-03-08|2008-01-08|Unigene Laboratories, Inc.|Oral peptide pharmaceutical dosage form and method of production| US20040001827A1|2002-06-28|2004-01-01|Dennis Mark S.|Serum albumin binding peptides for tumor targeting| US7772188B2|2003-01-28|2010-08-10|Ironwood Pharmaceuticals, Inc.|Methods and compositions for the treatment of gastrointestinal disorders| EP1631308B1|2003-05-30|2013-07-31|Amylin Pharmaceuticals, LLC|Novel methods and compositions for enhanced transmucosal delivery of peptides and proteins| JP5634008B2|2004-04-06|2014-12-03|アフィボディ・アーベー|New uses and methods| US20050282756A1|2004-06-18|2005-12-22|Mehta Nozer M|Oral delivery of peptide pharmaceutical compositions| CN101128487B|2004-12-02|2012-10-10|杜门蒂斯有限公司|Bispecific domain antibodies targeting serum albumin and GLP-1 or PYY| US8093207B2|2005-12-09|2012-01-10|Unigene Laboratories, Inc.|Fast-acting oral peptide pharmaceutical products| WO2008021088A2|2006-08-08|2008-02-21|The Regents Of The University Of Californina|Salicylanilides enhance oral delivery of therapeutic peptides| JP2010505435A|2006-10-11|2010-02-25|アブリンクスエン.ヴェー.|Amino acid sequences that bind to serum proteins essentially independently of pH, compounds containing them, and uses thereof| JP5718638B2|2007-07-31|2015-05-13|アフィボディ・アーベー|Novel compositions, methods and uses| EP2077272A1|2007-12-21|2009-07-08|Affibody AB|Polypeptide libraries with a predetermined scaffold| CA2719272C|2008-03-26|2020-08-18|Oramed Ltd.|Methods and compositions for oral administration of proteins| CN101294178A|2008-05-26|2008-10-29|江南大学|Method for improving hyaluronic acid volume of production of fermentation production with two-stage cultivation method| CN101294187B|2008-06-06|2013-07-31|暨南大学|Method for sustained-releasing polypeptide with biological activity and application thereof| NZ591497A|2008-08-18|2012-11-30|Entera Bio Ltd|A compositions comprising a protein, a protease inhibitor and N-caprylate or N-decanoate for diabetes mellitus treatment| WO2010054699A1|2008-11-17|2010-05-20|Affibody Ab|Conjugates of albumin binding domain| DK2437767T3|2009-06-01|2015-09-28|Medimmune Llc|MOLECULES WITH EXTENDED half-lives and uses thereof| EP2933262B1|2010-07-09|2018-05-02|Affibody AB|Polypeptides| US20120027058A1|2010-07-29|2012-02-02|The Regents Of The University Of Michigan|Portable, wireless multi-channel impedance analyzer| DK2621515T3|2010-09-28|2017-07-17|Aegerion Pharmaceuticals Inc|Chimeric seal-human leptin polypeptide with increased solubility| CN103347532A|2011-02-04|2013-10-09|爱奇司治疗公司|Orally bioavailable peptide drug compositions and methods thereof| DK2729160T3|2011-07-08|2019-07-01|Aegerion Pharmaceuticals Inc|MANIPULATED POLYPEPTIDES WHICH HAVE IMPROVED EFFECT TIME AND REDUCED IMMUNOGENICITY| EP2830665A1|2012-03-28|2015-02-04|Affibody AB|Oral administration| KR102123457B1|2012-09-25|2020-06-16|애피바디 에이비|Albumin binding polypeptide| EP3083675B1|2013-12-20|2018-03-07|Affibody AB|Engineered albumin binding polypeptide|EP2933262B1|2010-07-09|2018-05-02|Affibody AB|Polypeptides| US20120165650A1|2010-12-22|2012-06-28|General Electric Company|Her2 binders| KR102030531B1|2011-05-21|2019-10-10|마크로제닉스, 인크.|Deimmunized serum-binding domains and their use for extending serum half-life| US9382304B2|2011-07-08|2016-07-05|Amylin Pharmaceuticals, Llc|Engineered polypeptides having enhanced duration of action with reduced immunogenicity| DK2729160T3|2011-07-08|2019-07-01|Aegerion Pharmaceuticals Inc|MANIPULATED POLYPEPTIDES WHICH HAVE IMPROVED EFFECT TIME AND REDUCED IMMUNOGENICITY| EP3511339A1|2012-02-20|2019-07-17|Swedish Orphan Biovitrum AB |Polypeptides binding to human complement c5| JP2020033372A|2012-03-28|2020-03-05|アフィボディ・アーベー|Oral administration| EP2830665A1|2012-03-28|2015-02-04|Affibody AB|Oral administration| KR102123457B1|2012-09-25|2020-06-16|애피바디 에이비|Albumin binding polypeptide| CN104768976B|2012-10-05|2020-09-22|阿菲博迪公司|HER3 binding polypeptides| PL2912054T3|2012-10-25|2017-11-30|Affibody Ab|Albumin binding polypeptide| WO2014064238A1|2012-10-25|2014-05-01|Affibody Ab|Method for the separation of proteins containing an albumin-binding| WO2014076179A1|2012-11-14|2014-05-22|Affibody Ab|New polypeptide| WO2014076177A1|2012-11-14|2014-05-22|Affibody Ab|New polypeptide| WO2014096163A1|2012-12-19|2014-06-26|Affibody Ab|New polypeptides| EP2968490A1|2013-03-14|2016-01-20|Daiichi Sankyo Co., Ltd.|Novel binding proteins for pcsk9| CA2902657A1|2013-03-15|2014-09-18|Affibody Ab|Fcrn binding polypeptides| US9943568B2|2013-04-18|2018-04-17|Armo Biosciences, Inc.|Methods of using pegylated interleukin-10 for treating cancer| EP3434277A1|2013-06-17|2019-01-30|Armo Biosciences, Inc.|Method for assessing protein identity and stability| WO2015028550A1|2013-08-28|2015-03-05|Affibody Ab|C5 binding polypeptides| EP3811961A1|2013-08-28|2021-04-28|IPC Research, LLC|Stable polypeptides binding to human complement c5| AU2014311432A1|2013-08-30|2016-03-03|Armo Biosciences, Inc.|Methods of using interleukin-10 for treating diseases and disorders| EP3083675B1|2013-12-20|2018-03-07|Affibody AB|Engineered albumin binding polypeptide| SG10201912019WA|2014-05-22|2020-02-27|Pieris Pharmaceuticals Gmbh|Novel specific-binding polypeptides and uses thereof| CN106573072A|2014-06-02|2017-04-19|阿尔莫生物科技股份有限公司|Methods of lowering serum cholesterol| BR112017006515A8|2014-09-29|2018-02-27|Univ Duke|bispecific molecules, compositions, method for treating or preventing HIV-1 infection in an individual with this need, and vector| EP3206713A4|2014-10-14|2018-06-27|Armo Biosciences, Inc.|Interleukin-15 compositions and uses thereof| CA2963995A1|2014-10-22|2016-04-28|Armo Biosciences, Inc.|Methods of using interleukin-10 for treating diseases and disorders| CA2972683A1|2015-01-12|2016-07-21|Affibody Ab|Il-17a-binding polypeptides| WO2016126615A1|2015-02-03|2016-08-11|Armo Biosciences, Inc.|Methods of using interleukin-10 for treating diseases and disorders| MX2017012485A|2015-04-02|2018-04-11|Molecular Partners Ag|Designed ankyrin repeat domains with binding specificity for serum albumin.| CN107847583A|2015-05-28|2018-03-27|阿尔莫生物科技股份有限公司|PEGylated Interleukin 10 for treating cancer| CA2995120A1|2015-08-25|2017-03-02|Armo Biosciences, Inc.|Methods of using interleukin-10 for treating diseases and disorders| EP3665202A1|2017-08-09|2020-06-17|Massachusetts Institute Of Technology|Albumin binding peptide conjugates and methods thereof| CN108362873B|2018-02-13|2021-06-01|苏州仁端生物医药科技有限公司|Cadmium ion detection kit and application thereof| WO2019175176A1|2018-03-13|2019-09-19|Affibody Ab|Polypeptides based on a novel scaffold| WO2021043757A1|2019-09-02|2021-03-11|Biotest Ag|Factor viii protein with increased half-life| EP3785726A1|2019-09-02|2021-03-03|Biotest AG|Factor viii protein with increased half-life| WO2021089695A1|2019-11-05|2021-05-14|Affibody Ab|Polypeptides| WO2021165226A1|2020-02-17|2021-08-26|Biotest Ag|Subcutaneous administration of factor viii| EP3878515A1|2020-03-09|2021-09-15|Hober Biotech AB|Therapeutic agent targeting her2|
法律状态:
2020-02-18| B15I| Others concerning applications: loss of priority|Free format text: PERDA DAS PRIORIDADES US 61/399,285 DE 09/07/2010 E US 61/403,561 DE 17/09/2010 REIVINDICADAS NO PCT/EP2011/061623 POR NAO ENVIO DE DOCUMENTO COMPROBATORIO DE CESSAO DAS MESMAS CONFORME AS DISPOSICOES PREVISTAS NA LEI 9.279 DE 14/05/1996 (LPI) ART. 166O, NO ART. 28 DA RESOLUCAO INPI-PR 77/2013 E ART 3O DA IN 179 DE 21/02/2017 UMA VEZ QUE DEPOSITANTE CONSTANTE DA PETICAO DE REQUERIMENTO DO PEDIDO PCT E DISTINTO DAQUELE QUE DEPOSITOU AS PRIORIDADES REIVINDICADAS. | 2020-02-27| B06F| Objections, documents and/or translations needed after an examination request according [chapter 6.6 patent gazette]| 2020-06-23| B12F| Other appeals [chapter 12.6 patent gazette]| 2020-11-17| B07D| Technical examination (opinion) related to article 229 of industrial property law [chapter 7.4 patent gazette]|Free format text: DE ACORDO COM O ARTIGO 229-C DA LEI NO 10196/2001, QUE MODIFICOU A LEI NO 9279/96, A CONCESSAO DA PATENTE ESTA CONDICIONADA A ANUENCIA PREVIA DA ANVISA. CONSIDERANDO A APROVACAO DOS TERMOS DO PARECER NO 337/PGF/EA/2010, BEM COMO A PORTARIA INTERMINISTERIAL NO 1065 DE 24/05/2012, ENCAMINHA-SE O PRESENTE PEDIDO PARA AS PROVIDENCIAS CABIVEIS. | 2021-05-11| B07E| Notification of approval relating to section 229 industrial property law [chapter 7.5 patent gazette]| 2021-10-13| B350| Update of information on the portal [chapter 15.35 patent gazette]| 2021-11-30| B06U| Preliminary requirement: requests with searches performed by other patent offices: procedure suspended [chapter 6.21 patent gazette]|
优先权:
[返回顶部]
申请号 | 申请日 | 专利标题 US39928510P| true| 2010-07-09|2010-07-09| US61/399,285|2010-07-09| US40356110P| true| 2010-09-17|2010-09-17| US61/403,561|2010-09-17| PCT/EP2011/061623|WO2012004384A2|2010-07-09|2011-07-08|Polypeptides| 相关专利
Sulfonates, polymers, resist compositions and patterning process
Washing machine
Washing machine
Device for fixture finishing and tension adjusting of membrane
Structure for Equipping Band in a Plane Cathode Ray Tube
Process for preparation of 7 alpha-carboxyl 9, 11-epoxy steroids and intermediates useful therein an
国家/地区
|