WO2014072481A1 - Albumin variants - Google Patents

Albumin variants Download PDF

Info

Publication number
WO2014072481A1
WO2014072481A1 PCT/EP2013/073426 EP2013073426W WO2014072481A1 WO 2014072481 A1 WO2014072481 A1 WO 2014072481A1 EP 2013073426 W EP2013073426 W EP 2013073426W WO 2014072481 A1 WO2014072481 A1 WO 2014072481A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
polypeptide
albumin
fragment
positions
Prior art date
Application number
PCT/EP2013/073426
Other languages
French (fr)
Inventor
Jason Cameron
Karen DELAHAY
Jens Erik Nielsen
Andrew Plumridge
Jan Terje Andersen
Original Assignee
Novozymes Biopharma Dk A/S
Novozymes A/S
Novozymes Biopharma Uk Limited
University Of Oslo
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to JP2015541165A priority Critical patent/JP6487328B2/en
Priority to EP13789000.0A priority patent/EP2917233A1/en
Priority to CA2890766A priority patent/CA2890766A1/en
Priority to CN201380069331.4A priority patent/CN105452290A/en
Priority to RU2015121693A priority patent/RU2670063C2/en
Priority to KR1020157014474A priority patent/KR20150082422A/en
Application filed by Novozymes Biopharma Dk A/S, Novozymes A/S, Novozymes Biopharma Uk Limited, University Of Oslo filed Critical Novozymes Biopharma Dk A/S
Priority to AU2013343503A priority patent/AU2013343503B2/en
Priority to MX2015005363A priority patent/MX2015005363A/en
Priority to BR112015010318A priority patent/BR112015010318A2/en
Publication of WO2014072481A1 publication Critical patent/WO2014072481A1/en
Priority to IL238185A priority patent/IL238185B/en
Priority to AU2018201136A priority patent/AU2018201136A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/76Albumins
    • C07K14/765Serum albumin, e.g. HSA
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/385Haptens or antigens, bound to carriers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/38Albumins
    • A61K38/385Serum albumin
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/31Fusion polypeptide fusions, other than Fc, for prolonged plasma life, e.g. albumin

Definitions

  • the invention relates to variants of albumin or fragments thereof or fusion polypeptides comprising variant albumin or fragments thereof having a change in binding affinity to FcRn and/or a change in half-life compared to the albumin, fragment thereof or fusion polypeptide comprising albumin or a fragment thereof.
  • the invention allows tailoring of binding affinity and/or half-life of an albumin to the requirements and desires of a user or application.
  • Albumin is a protein naturally found in the blood plasma of mammals where it is the most abundant protein. It has important roles in maintaining the desired osmotic pressure of the blood and also in transport of various substances in the blood stream. Albumins have been characterized from many species including human, pig, mouse, rat, rabbit and goat and they share a high degree of sequence and structural homology.
  • FcRn neonatal Fc receptor
  • BrambeH neonatal Fc receptor
  • FcRn is a membrane bound protein, expressed in many cell and tissue types. FcRn has been found to salvage albumin from intracellular degradation (Roopenian D. C. and Akilesh, S. (2007), Nat. Rev. Immunol 7, 715-725.). FcRn is a bifunctional molecule that contributes to maintaining a high level of IgGs and albumin in serum in mammals such as human beings.
  • FcRn-immunoglobulin (IgG) interaction Whilst the FcRn-immunoglobulin (IgG) interaction has been characterized in the prior art, the FcRn-albumin interaction is less well characterized.
  • the major FcRn binding site is localized within Dil l (381 -585), (Andersen et al (2010), Clinical Biochemistry 43,367-372).
  • a number of key amino acids have been shown to be important in binding, notably histidines H464, H510 a n d H 53 6 and Lys500 (Andersen et al (2010), Nat. Commun. 3:610. DOI:10.1038/ncomms1607).
  • HSA human serum albumin
  • HSA Human serum albumin
  • the plasma half-life of HSA has been found to be approximately 19 days.
  • a natural variant having lower plasma half-life has been identified (Peach, R. J . and Brennan, S. 0., (1991 ) Biochim Biophys Acta.1097:49-54) having the substitution D494N .
  • This substitution generated an N-glycosylation site in this variant, which is not present in the wild-type albumin. It is not known whether the glycosylation or the amino acid change is responsible for the change in plasma half- life.
  • Albumin has a long plasma half-life and because of this property it has been suggested for use i n d rug del ivery.
  • Al bu m i n has been conjugated to pharmaceutically beneficial compounds (WO2000/69902), and it was found that the conjugate maintained the long plasma half-life of albumin.
  • the resulting plasma half-life of the conjugate was generally considerably longer than the plasma half-life of the beneficial therapeutic compound alone.
  • albumin has been genetically fused to therapeutically beneficial peptides (WO 2001/79271 A and WO2003/59934) with the typical result that the fusion has the activity of the therapeutically beneficial peptide and a considerably longer plasma half-life than the plasma half-life of the therapeutically beneficial peptides alone.
  • Galliano et al (1993) Biochim. Biophys. Acta 1225, 27-32 discloses a natural variant E505K.
  • Minchiotti ei a/ (1990) discloses a natural variant K536E.
  • Minchiotti ei a/ (1987) Biochim. Biophys. Acta 916, 41 1 -418 discloses a natural variant K574N.
  • Takahashi et al (1987) Proc. Natl. Acad. Sci. USA 84, 4413-4417 discloses a natural variant D550G. Carlson et al (1992).
  • Proc. Nat. Acad. Sci. USA 89, 8225- 8229 discloses a natural variant D550A.
  • WO201 1/051489 and WO2012/150319 disclose a number of point mutations in albumin which modulate the binding of albumin to FcRn.
  • WO2010/092135 discloses a number of point mutations in albumin which increase the number of thiols available for conjugation in the albumin, the disclosure is silent about the affect of the mutations on the binding of the albumin to FcRn.
  • WO201 1/103076 discloses albumin variants, each containing a substitution in Domain III of HSA.
  • WO2012/1 12188 discloses albumin variants containing substitutions in Domain III of HSA.
  • Albumin has the ability to bind a number of ligands and these become associated (associates) with albumin. This property has been utilized to extend the plasma half-life of drugs having the ability to non-covalently bind to albumin. This can also be achieved by binding a pharmaceutical beneficial compound, which has little or no albumin binding properties, to a moiety having albumin binding properties, see review article and reference therein, Kratz (2008) Journal of Controlled Release 132, 171 -183.
  • Albumin is used in preparations of pharmaceutically beneficial compounds, in which such a preparation may be for example, but not limited to, a nanoparticle or microparticle of albumin.
  • delivery of a pharmaceutically beneficial compound or mixture of compounds may benefit from alteration in the albumin's affinity to its receptor where the beneficial compound has been shown to associate with albumin for the means of delivery. It is not clear what determines the plasma half-life of the formed associates (for example but not limited to Levemir®, Kurtzhals P et al. Biochem. J. 1995; 312:725-731 ), conjugates or fusion polypeptides but it appears to be a result of the combination of the albumin and the selected pharmaceutically beneficial compound/polypeptide.
  • Kenanova et al (2010, Protein Engineering, Design & Selection 23(10): 789-798; WO2010/1 18169) discloses a docking model comprising a structural model of domain III of HSA (solved at pH 7 to 8) and a structural model of FcRn (solved at pH 6.4).
  • Kenanova et al discloses that positions 464, 505, 510, 531 and 535 in domain III potentially interact with FcRn.
  • the present invention provides further variants having altered binding affinity to the FcRn receptor.
  • the albumin moiety or moieties may therefore be used to tailor the binding affinity to FcRn and/or half-life of fusion polypeptides, conjugates, associates, nanoparticles and compositions comprising the albumin moiety.
  • the present invention relates to albumin variants comprising an alteration at positions corresponding to positions selected among two or more of the group consisting of positions 492, 550, 573, 574 and 580 of the mature polypeptide of SEQ ID NO: 2 or equivalent positions of other albumins or fragments thereof.
  • Position 492 is located in the connector loop between subdomain Dllla and subdomain Dlllb.
  • Positions 550 , 573 , 574 and 580 are located in subdomain Dlllb.
  • Subdomain 1Mb is located proximal to the connector loop between subdomain Dllla and subdomain Dlllb.
  • the present invention also relates to isolated polynucleotides encoding the variants; nucleic acid constructs, vectors, and host cells comprising the polynucleotides; and methods of producing the variants.
  • the invention also relates to conjugates or associates comprising the variant albumin or fragment thereof according to the invention and a beneficial therapeutic moiety or to a fusion polypeptide comprising a variant albumin or fragment thereof of the invention and a fusion partner polypeptide.
  • the invention further relates to compositions comprising the variant albumin, fragment thereof, fusion polypeptide comprising variant albumin or fragment thereof or conjugates comprising the variant albumin or fragment thereof, according to the invention or associates comprising the variant albumi n or fragment thereof, accordi ng to the invention .
  • the compositions are preferably pharmaceutical compositions.
  • the invention further relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a variant albumin, fragment thereof, fusion polypeptide comprising variant albumin or fragment thereof or conjugates comprising the variant albumin or fragment thereof, or associates comprising the variant albumin or fragment thereof.
  • the invention also relates to the use of the variants, fragments, fusion polypeptides, conjugates, associates, nanoparticles and microparticles.
  • the invention also relates to a method for preparing a variant albumin, fragment thereof, fusion polypeptide comprising variant albumin or fragment thereof or conjugates comprising the variant albumin or fragment thereof, or associates comprising the variant albumin or fragment thereof.
  • Figure 1 Multiple alignment of amino acid sequences of (i) full length mature HSA (Hu_1_2_3), (ii) an albumin variant comprising domain I and domain III of HSA (Hu_1_3), (iii) an albumin variant comprising domain II and domain I II of HSA (Hu_2_3), (iv) full-length Macaca mulatta albumin (Mac_mul), (v) full-length Rattus norvegicus albumin (Rat) and (vi) full-length Mus musculus albumin (Mouse). Positions 500, 550 and 573 (relative to full length HSA) are indicated by arrows. In Figure 1 , Domains I, II and III are referred to as 1 , 2 and 3 (respectively).
  • Figure 2 Multiple alignment of amino acid sequences of mature albumin from human, sheep, mouse, rabbit and goat and immature albumins from chimpanzee ("Chimp"), macaque, hamster, guinea pig, rat, cow, horse, donkey, dog, chicken, and pig.
  • the Start and End amino acids of domains 1 , 2 and 3 are indicated with respect to mature human albumin.
  • Figure 3 conserveed groups of amino acids based on their properties.
  • FIG. 4 Representation of shFcRn-HSA docking model.
  • A-B Two orientations of the complex are shown. Albumin is shown by a space-filling diagram, FcRn is shown as a ribbon diagram.
  • the core binding interface of HSA is highlighted in pink (in grey-scale this is seen as the darkest (almost black) region; Dl (CBI)), while the area distally localized from the interface is shown as DM (orange) and DI N is split into sub-domains Dl I la (in colour, this is cyan) and Dlllb (in colour, this is blue).
  • variant means a polypeptide derived from a parent albumin by one or more (several) alteration(s), i.e., a substitution, insertion, and/or deletion, at one or more (several) positions.
  • a substitution means a replacement of an amino acid occupying a position with a different amino acid;
  • a deletion means removal of an amino acid occupying a position; and
  • an insertion means adding 1 or more, such as 1 , 2, 3, 4, 5, 6, 7, 8, 9 or 10, preferably 1 -3 amino acids immediately adjacent an amino acid occupying a position.
  • 'immediately adjacent' may be to the N-side ('upstream') or C-side ('downstream') of the amino acid occupying a position ('the named amino acid'). Therefore, for an amino acid named/numbered 'X', the insertion may be at position 'X+1 ' ('downstream') or at position 'X-1 ' ('upstream').
  • Mutant means a polynucleotide encoding a variant.
  • Wild-Type Albumin means albumin having the same amino acid sequence as naturally found in an animal or in a human being.
  • Parent Albumin means an albumin to which an alteration is made by the hand of man to produce the albumin variants of the invention.
  • the parent may be a naturally occurring (wild-type) polypeptide or an allele thereof, or even a variant thereof.
  • Albumins are proteins and constitute the most abundant protein in plasma in mammals and albumins from a long n umber of mammals have been characterized by biochemical methods and/or by sequence information.
  • Several albumins e.g., human serum albumin (HSA), have also been characterized crystallographically and the structure determined (HSA: He XM, Carter DC (July 1 992). "Atomic structure and chemistry of human serum albumin”. Nature 358 (6383): 209-15; horse albumin: Ho, J.X. et al. (2001 ). X-ray and primary structure of horse serum albumin (Equus caballus) at 0.27-nm resolution. Eur J Biochem. 215(1 ):205-12).
  • albumin means a protein having the same and/or very similar th ree dimensional (tertiary) structure as HSA or HSA domains and has similar properties to HSA or to the relevant domains. Similar three dimensional structures are for example the structures of the albumins from the species mentioned herein. Some of the major properties of albumin are i) its ability to regulate plasma volume (oncotic activity), ii) a long plasma half-life of around 19 days ⁇ 5 days, iii) binding to FcRn, iv) ligand-binding, e.g. binding of endogenous molecules such as acidic, lipophilic compounds including bilirubin, fatty acids, hemin and thyroxine (see also table
  • Albumins have generally a long plasma half-life of approximately 20 days or longer, e.g., HSA has a plasma half-life of 19 days. It is known that the long plasma half-life of HSA is mediated via interaction with its receptor FcRn, however, an understanding or knowledge of the exact mechanism behind the long half-life of HSA is not essential for the invention.
  • albumin proteins As examples of albumin proteins , more specifically albumin proteins which may be used as parent 'backbones' as a starting point for making the albumin variants according to the invention, can be mentioned human serum albumin (e.g. AAA98797 or P02768-1 , SEQ I D NO:
  • primate serum albumin such as chimpanzee serum albumin (e.g. predicted sequence XP_517233.2 SEQ I D NO: 5), gorilla serum albumin or macaque serum albumin (e.g. NP_001 182578, SEQ ID NO: 6), rodent serum albumin (such as hamster serum albumin (e.g. A6YF56, SEQ I D NO : 7), gu inea pig serum albumi n (e.g. Q6WDN9-1 , SEQ I D NO: 8), mouse serum albumin (e.g.
  • primate serum albumin such as chimpanzee serum albumin (e.g. predicted sequence XP_517233.2 SEQ I D NO: 5), gorilla serum albumin or macaque serum albumin (e.g. NP_001 182578, SEQ ID NO: 6), rodent serum albumin (such as hamster serum albumin (e.g. A6YF56, SEQ I D NO : 7), gu
  • the parent or reference albumin may be an artificial variant such as HSA.
  • K573P (SEQ ID NO: 3) or a chimeric albumin such as the N-terminal of HSA and the C-terminal of macaca albumin (SEQ I D NO: 20), N- terminal of HSA and the C-terminal of mouse albumin (SEQ ID NO: 21 ), N-terminal of HSA and the C-terminal of rabbit albumin (SEQ I D NO: 22), N-terminal of HSA and the C-terminal of sheep albumin (SEQ ID NO: 23).
  • a chimeric albumin such as the N-terminal of HSA and the C-terminal of macaca albumin (SEQ I D NO: 20), N- terminal of HSA and the C-terminal of mouse albumin (SEQ ID NO: 21 ), N-terminal of HSA and the C-terminal of rabbit albumin (SEQ I D NO: 22), N-terminal of HSA and the C-terminal of sheep albumin (SEQ ID NO: 23).
  • albumin examples include ovalbumin (e.g. P01012.pro: chicken ovalbumin; 073860. pro: turkey ovalbumin).
  • HSA as disclosed in SEQ I D NO: 2 or any naturally occurring allele thereof, is the preferred parent albumin according to the invention.
  • HSA is a protein consisting of 585 amino acid residues and has a molecular weight of 67 kDa. In its natural form it is not glycosylated.
  • natural alleles may exist having essentially the same properties as HSA but having one or more (several) amino acid changes compared to SEQ ID NO: 2, and the inventors also contemplate the use of such natural alleles as parent albumin according to the invention.
  • the parent albumin , a fragment thereof, or albumin part of a fusion polypeptide comprising albumin or a fragment thereof according to the invention preferably has a sequence identity to the sequence of HSA shown in SEQ I D NO: 2 of at least 60%, preferably at least 70%, preferably at least 80%, preferably at least 85%, preferably at least 86%, preferably at least 87%, preferably at least 88%, preferably at least 89%, preferably at least 90%, preferably at least 91 %, preferably at least 92%, preferably at least 93%, preferably at least 94%, preferably at least 95%, more preferred at least 96%, more preferred at least 97%, more preferred at least 98% and most preferred at least 99%.
  • the parent albumin maintains at least one of the major properties of albumin or a similar tertiary structure as an albumin, such as HSA.
  • the sequence identity may be over the full-length of SEQ I D NO: 2 or over a molecule consisting or comprising of a fragment such as one or more (several) domains of SEQ ID NO: 2 such as a molecule consisting of or comprising domain I II (e.g. SEQ ID NO: 27), a molecule consisting of or comprising domain II and domain I II (e.g. SEQ I D NO: 25), a molecule consisting of or comprising domain I and domain III (e.g.
  • SEQ ID NO: 24 a molecule consisting of or comprising two copies of domain III (e.g. SEQ ID NO: 26), a molecule consisting of or comprising three copies of domain III (e.g. SEQ I D NO: 28) or a molecule consisting of or comprising domain I and two copies of domain III (e.g. SEQ ID NO: 29).
  • the parent preferably comprises or consists of the amino acid sequence of SEQ I D NO: 4 (immature sequence of HSA) or SEQ ID NO: 2 (mature sequence of HSA).
  • the parent is an allelic variant of the mature polypeptide of SEQ ID NO: 2.
  • the parent albumin may be encoded by a polynucleotide that hybridizes under very low stringency conditions, low stringency conditions, medium stringency conditions, medium-high stringency conditions, high stringency conditions, or very high stringency conditions with (i) the mature polypeptide coding sequence of SEQ I D NO: 1 or (ii) the full-length complementary strand of (i) (J. Sambrook, E.F. Fritsch, and T. Maniatis, 1989, Molecular Cloning, A Laboratory Manual, 2d edition, Cold Spring Harbor, New York).
  • the polynucleotide of SEQ I D NO: 1 or a subsequence thereof, as well as the amino acid sequence of SEQ I D NO: 2 or a fragment thereof, may be used to design nucleic acid probes to identify and clone DNA encoding a parent from strains of different genera or species according to methods well known in the art. I n particular, such probes can be used for hybridization with the genomic or cDNA of the genus or species of interest, following standard Southern blotting procedures, in order to identify and isolate the corresponding gene therein. Such probes can be considerably shorter than the entire sequence, but should be at least 14, e.g. , at least 25, at least 35, or at least 70 nucleotides in length.
  • the nucleic acid probe is at least 1 00 nucleotides in length , e.g., at least 200 nucleotides, at least 300 nucleotides, at least 400 nucleotides, at least 500 nucleotides, at least 600 nucleotides, at least 700 nucleotides, at least 800 nucleotides, or at least 900 nucleotides in length.
  • Both DNA and RNA probes can be used.
  • the probes are typically labelled for detecting the corresponding gene (for example, with 32 P, 3 H, 35 S, biotin, or avidin). Such probes are encompassed by the invention.
  • a genomic DNA or cDNA library prepared from such other organisms may be screened for DNA that hybridizes with the probes described above and encodes a parent.
  • Genomic or other DNA from such other organisms may be separated by agarose or polyacrylamide gel electrophoresis, or other separation techniques.
  • DNA from the libraries or the separated DNA may be transferred to and immobilized on nitrocellulose or other suitable carrier material.
  • the carrier material is used in a Southern blot.
  • hybridization indicates that the polynucleotide hybridizes to a labelled nucleotide probe corresponding to the polynucleotide shown in SEQ I D NO: 1 , its complementary strand, or a subsequence thereof, under low to very high stringency conditions.
  • Molecules to which the probe hybridizes can be detected using, for example, X-ray film or any other detection means known in the art.
  • the nucleic acid probe may comprise or consist of the mature polypeptide coding sequence of SEQ ID NO: 1 , i.e. nucleotides 1 to 1785 of SEQ ID NO: 1 .
  • the nucleic acid probe may comprise or consist of a polynucleotide that encodes the polypeptide of SEQ ID NO: 2 or a fragment thereof.
  • very low to very high stringency conditions are defined as pre-hybridization and hybridization at 42°C in 5X SSPE, 0.3% SDS, 200 micrograms/ml sheared and denatured salmon sperm DNA, and either 25% formamide for very low and low stringencies, 35% formamide for medium and medium-high stringencies, or 50% formamide for high and very high stringencies, following standard Southern blotting procedures for 12 to 24 hours optimally.
  • the carrier material is finally washed three times each for 15 minutes using 2X SSC, 0.2% SDS at 45°C (very low stringency), 50°C (low stringency), 55°C (medium stringency), 60°C (medium-high stringency), 65°C (high stringency), or 70°C (very high stringency).
  • stringency conditions are defined as pre-hybridization and hybridization at about 5°C to about 10°C below the calculated T m using the calculation according to Bolton and McCarthy (1962, Proc. Natl. Acad. Sci. USA 48: 1390) in 0.9 M NaCI, 0.09 M Tris-HCI pH 7.6, 6 mM EDTA, 0.5% NP-40, 1 X Denhardt's solution , 1 m M sodium pyrophosphate, 1 mM sodium monobasic phosphate, 0.1 mM ATP, and 0.2 mg of yeast RNA per ml following standard Southern blotting procedures for 12 to 24 hours optimally. The carrier material is finally washed once in 6X SCC plus 0.1 % SDS for 15 minutes and twice each for 15 minutes using 6X SSC at 5°C to 10°C below the calculated T m .
  • the parent may be encoded by a polynucleotide with a sequence identity to the mature polypeptide coding sequence of SEQ ID NO: 1 of at least 60%, e.g., at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%, which encodes a polypeptide which is able to function as an albumin .
  • the parent is encoded by a polynucleotide comprising or consisting of SEQ ID NO: 1 .
  • Albumin moiety The albumin part of a fusion polypeptide, conjugate, associate, nanoparticle or composition comprising the albumin variant or fragment thereof according to the invention, may be referred to as an 'albumin moiety' or 'albumin component'.
  • a polypeptide according to the invention may comprise or consist of an albumin moiety.
  • FcRn and shFcRn The term "FcRn" means the human neonatal Fc receptor (FcRn).
  • shFcRn is a soluble recombinant form of FcRn.
  • hFcRn is a heterodimer of SEQ I D NO: 30 (truncated heavy chain of the major histocompatibility complex class l-like Fc receptor (FCGRT)) and SEQ ID NO: 31 (beta-2-microglobulin). Together, SEQ I D NO: 30 and 31 form hFcRn.
  • Isolated variant means a variant in a form or environment which does not occur in nature.
  • isolated variants include (1 ) any non- naturally occurring variant, (2) any variant that is at least partially removed from one or more (several) or all of the naturally occurring constituents with which it is associated in nature; (3) any variant modified by the hand of man relative to the polypeptide from which it is derived (e.g. the polypeptide from which it is derived as found in nature); or (4) any variant modified by increasing the amount of the variant e relative to other components with which it is naturally associated (e.g., multiple copies of a gene encoding the substance; use of a stronger promoter than the promoter naturally associated with the gene encoding the substance).
  • An isolated variant may be present in a fermentation broth sample.
  • substantially pure variant means a preparation that contains at most 10%, at most 8%, at most 6%, at most 5%, at most 4%, at most 3%, at most 2%, at most 1 %, and at most 0.5% by weight of other polypeptide material with which it is natively or recombinantly associated.
  • the variant is at least 92% pure, e.g., at least 94% pure, at least 95% pure, at least 96% pure, at least 97% pure, at least 98% pure, at least 99%, at least 99.5% pure, and 100% pure by weight of the total polypeptide material present in the preparation. Purity may be determined by SDS-PAGE or GP-HPLC.
  • the variants of the invention are preferably in a substantially pure form. This can be accomplished, for example, by preparing the variant by well-known recombinant methods and by purification methods.
  • Mature polypeptide means a polypeptide in its final form followi ng translation and any post-translational modifications , such as N-terminal processing, C-terminal truncation, glycosylation, phosphorylation, etc.
  • the mature polypeptide may be amino acids 1 to 585 of SEQ ID NO: 2, e.g. with the inclusion of alterations according to the invention and/or any post-translational modifications.
  • Mature polypeptide coding sequence means a polynucleotide that encodes a mature albumin polypeptide.
  • the mature polypeptide coding sequence may be nucleotides 1 to 1 758 of SEQ I D NO: 1 e.g. with the alterations required to encode a variant according to the invention.
  • sequence identity The relatedness between two amino acid sequences or between two nucleotide sequences is described by the parameter "sequence identity”.
  • the sequence identity between two amino acid sequences is determined using the Needleman-Wunsch algorithm (Needleman and Wunsch, 1970, J. Mol. Biol. 48: 443-453) as implemented in the Needle program of the EMBOSS package (EMBOSS: The European Molecular Biology Open Software Suite, Rice et al., 2000, Trends Genet. 16: 276-277), preferably version 5.0.0 or later.
  • the parameters used are gap open penalty of 10, gap extension penalty of 0.5, and the EBLOSUM62 (EMBOSS version of BLOSUM62) substitution matrix.
  • the output of Needle labeled "longest identity" (obtained using the -nobrief option) is used as the percent identity and is calculated as follows:
  • th e seq u en ce id entity between two deoxyribonucleotide sequences is determined using the Need leman-Wunsch algorithm (Needleman and Wunsch, 1970, supra) as implemented in the Needle program of the EMBOSS package (EMBOSS: The European Molecular Biology Open Software Suite, Rice et al., 2000, supra), preferably version 5.0.0 or later.
  • the parameters used are gap open penalty of 10, gap extension penalty of 0.5, and the EDNAFULL (EMBOSS version of NCBI NUC4.4) substitution matrix.
  • the output of Needle labeled "longest identity" (obtained using the -nobrief option) is used as the percent identity and is calculated as follows:
  • Fragment means a polypeptide having one or more (several) amino acids deleted from the amino and/or carboxyl terminus of an albumin and/or an internal region of albumin that has retained the ability to bind to FcRn. Fragments may consist of one uninterrupted sequence derived from HSA or it may comprise two or more sequences derived from HSA.
  • the fragments according to the invention have a size of more than approximately 20 amino acid residues, preferably more than 30 amino acid residues, more preferred more than 40 amino acid residues, more preferred more than 50 amino acid residues, more preferred more than 75 amino acid residues, more preferred more than 1 00 amino acid residues, more preferred more than 200 amino acid residues, more preferred more than 300 amino acid residues, even more preferred more than 400 amino acid residues and most preferred more than 500 amino acid residues.
  • a fragment may comprise or consist of one more domains of albumin such as Dl + DM , Dl + Di l l , DM + DI N , Di l l + Di l l , Dl + Di l l + Di l l, Di l l + Di l l +DI II , or fragments of such domains or combinations of domains.
  • albumin such as Dl + DM , Dl + Di l l , DM + DI N , Di l l + Di l l , Dl + Di l l + Di l l, Di l l + Di l l +DI II , or fragments of such domains or combinations of domains.
  • HSA domain I may consist of or comprise amino acids 1 to 194 ( ⁇ 1 to 15 amino acids) of SEQ I D NO: 2
  • HSA domain I I may consist of or comprise amino acids 192 ( ⁇ 1 to 15 amino acids) to 387 ( ⁇ 1 to 15 amino acids) of SEQ ID NO: 2
  • domain I II may consist of or comprise amino acid residues 381 ( ⁇ 1 to 15 amino acids) to 585 ( ⁇ 1 to 15 amino acids) of SEQ ID NO: 2.
  • ⁇ 1 to 15 amino acids means that the residue number may deviate by 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, or 15 amino acids to the C-terminus and/or to the N-terminus of the stated amino acid position. Examples of domains I, II and III are described by Dockal et al (The Journal of Biological Chemistry, 1999, Vol. 274(41 ): 29303-29310) and Kjeldsen et al (Protein Expression and Purification, 1998, Vol 13: 163-169) and are tabulated below.
  • the skilled person can identify domains I , I I and I I I in non-human albumins by amino acid sequence alignment with HSA, for example using the Needleman-Wunsch algorithm (Needleman and Wunsch, 1970, J. Mol. Biol. 48: 443-453) as implemented in the Needle program of the EMBOSS package (EMBOSS: The European Molecular Biology Open Software Suite, Rice et al., 2000, Trends Genet. 16: 276-277), preferably version 3.0.0 or later.
  • the optional parameters used are gap open penalty of 10, gap extension penalty of 0.5, and the EBLOSUM62 (EMBOSS version of BLOSU M62) substitution matrix.
  • Other suitable software includes M USCLE ((Multiple sequence comparison by log-expectation, Robert C.
  • domains have at least 70, 75, 80, 85, 90, 95, 96, 97, 98, 99, 99.5 % identity or 100% identity to Domain I, II or III of HSA (SEQ ID NO: 2).
  • allelic variant means any of two or more alternative forms of a gene occupying the same chromosomal locus. Allelic variation arises naturally through mutation, and may result in polymorphism within populations. Gene mutations can be silent (no change in the encoded polypeptide) or may encode polypeptides having altered amino acid sequences.
  • An allelic variant of a polypeptide is a polypeptide encoded by an allelic variant of a gene.
  • Coding sequence means a polynucleotide, which directly specifies the amino acid sequence of its translated polypeptide product.
  • the boundaries of the coding sequence are generally determined by an open reading frame, which usually begins with the ATG start codon or alternative start codons such as GTG and TTG and ends with a stop codon such as TAA, TAG, and TGA.
  • the coding sequence may be a DNA, cDNA, synthetic, or recombinant polynucleotide.
  • cDNA means a DNA molecule that can be prepared by reverse transcription from a mature, spliced, mRNA molecule obtained from a eukaryotic cell. cDNA lacks intron sequences that may be present in the corresponding genomic DNA.
  • the initial, primary RNA transcript is a precursor to mRNA that is processed through a series of steps, including splicing, before appearing as mature spliced mRNA.
  • nucleic acid construct means a nucleic acid molecule, either single- or double-stranded, which is isolated from a naturally occurring gene or is modified to contain segments of nucleic acids in a manner that would not otherwise exist in nature or which is synthetic.
  • nucleic acid construct is synonymous with the term “expression cassette” when the nucleic acid construct contains the control sequences required for expression of a coding sequence of the invention.
  • control sequences means all nucleic acid sequences necessary for the expression of a polynucleotide encoding a variant of the invention.
  • Each control sequence may be native (i.e. from the same gene) or foreign (i.e. from a different gene) to the polynucleotide encoding the variant or native or foreign to each other.
  • control sequences include, but are not limited to, a leader, polyadenylation sequence, propeptide sequence, promoter, signal peptide sequence, and transcription terminator.
  • the control sequences include a promoter, and transcriptional and translational stop signals.
  • the control sequences may be provided with linkers for the purpose of introducing specific restriction sites facilitating ligation of the control sequences with the coding region of the polynucleotide encoding a variant.
  • operably linked means a configuration in which a control seq uence is placed at an appropriate position relative to the coding sequence of a polynucleotide such that the control sequence directs the expression of the coding sequence.
  • expression includes any step involved in the production of the variant including, but not limited to, transcription, post-transcriptional modification, translation, post-translational modification, and secretion.
  • Expression vector means a linear or circular DNA molecule that comprises a polynucleotide encoding a variant and is operably linked to control sequences that provide for its expression.
  • host cell means any cell type that is susceptible to transformation, transfection, transduction, or the like with a nucleic acid construct or expression vector comprising a polynucleotide of the present invention.
  • host cell encompasses any progeny of a parent cell that is not identical to the parent cell due to mutations that occur during replication.
  • Plasma half-life Plasma half-life is ideally determined in vivo in suitable individuals.
  • a longer plasma half-life with respect to a variant albumin of the invention means that the variant has longer plasma half-life than the corresponding albumin having the same sequences except for the alteration(s) described herein, e.g. at two or more positions corresponding to 492, 550, 573, 574 and 580 of HSA (SEQ ID NO: 2).
  • a reference is an albumin, fusion, conjugate, composition, associate or nanoparticle to which an albumin variant, fusion , conjugate, composition , associate or nanoparticle is compared.
  • the reference may comprise or consist of full length albumin (such as HSA or a natural allele thereof) or a fragment thereof.
  • a reference may also be referred to as a 'corresponding' albumin, fusion, conjugate, composition, associate or nanoparticle to which an albumin variant, fusion, conjugate, composition, associate or nanoparticle is compared.
  • a reference may comprise or consist of HSA (SEQ I D NO: 2) or a fragment, fusion, conjugate, associate, nanoparticle or microparticle thereof.
  • the reference is identical to the polypeptide, fusion polypeptide, conjugate, composition, associate, nanoparticle or microparticle according to the invention ("being studied") with the exception of the albumin moiety.
  • the albumin moiety of the reference comprises or consists of an albumin (e.g. HSA, SEQ ID NO: 2) or a fragment thereof.
  • the amino acid sequence of the albumin moiety of the reference may be longer than, shorter than or, preferably, the same ( ⁇ 1 to 15 amino acids) length as the amino sequence of the albumin moiety of the polypeptide, fusion polypeptide, conjugate, composition , associate, nanoparticle or microparticle according to the invention ("being studied").
  • Equivalent amino acid positions are defined in relation to full-length mature human serum albumin (i.e. without leader sequence, SEQ I D NO: 2). However, the skilled person understands that the invention also relates to variants of non-human albumins (e.g. those disclosed herein) and/or fragments of a human or non-human albumin. Equivalent positions can be identified in fragments of human serum albumin, in animal albumins and in fragments, fusions and other derivative or variants thereof by comparing amino acid sequences using pairwise (e.g. ClustalW) or multiple (e.g. MUSCLE) alignments. For example, Fig.
  • Table 1 Example of identification of equivalent positions in HSA, animal albumins and albumin fragments
  • Fig. 1 was generated by MUSCLE using the default parameters including output in C l u sta lW 1 .81 fo rm at .
  • Th e raw ou tp u t d ata was s h ad ed u s i n g B oxS h ad e 3.2 1 (http://www.ch.embnet.org/software/BOX form.html) using Output Format: RTF_new; Font Size: 10; Consensus Line: no consensus line; Fraction of sequences (that must agree for shading): 0.5; Input sequence format: ALN.
  • amino acid positions defined in human serum albumin also apply to equivalent positions in fragments, derivatives or variants and fusions of human serum albumin, animals from other species and fragments and fusions thereof.
  • Such equivalent positions may have (i) a different residue number in its native protein and/or (ii) a different native amino acid in its native protein.
  • Fig. 2 shows that equivalent positions can be identified in fragments (e.g. domains) of an albumin with reference to SEQ ID NO: 2 (HSA).
  • the amino acid sequence of another albumin is aligned with the mature polypeptide disclosed in SEQ I D NO: 2, and based on the alignment, the amino acid position number corresponding to any amino acid residue in the mature polypeptide disclosed in SEQ I D NO: 2 is determined using the Needleman-Wunsch algorithm (Needleman and Wunsch, 1970, J. Mol. Biol. 48: 443-453) as implemented in the Needle program of the EMBOSS package (EMBOSS: The European Molecular Biology Open Software Suite, Rice et al., 2000, Trends Genet. 16: 276-277), preferably version 5.0.0 or later.
  • the parameters used are gap open penalty of 1 0, gap extension penalty of 0.5, and the EBLOSUM62 (EMBOSS version of BLOSUM62) substitution matrix.
  • Identification of the corresponding amino acid residue in another albumin can be determined or confirmed by an alignment of multiple polypeptide sequences using several computer programs including, but not limited to, MUSCLE (multiple sequence comparison by log-expectation; version 3.5 or later; Edgar, 2004, Nucleic Acids Research 32: 1792-1797), MAFFT (version 6.857 or later; Katoh and Kuma, 2002, Nucleic Acids Research 30: 3059-3066; Katoh et al., 2005, Nucleic Acids Research 33: 51 1 -518; Katoh and Toh, 2007, Bioinformatics 23: 372-374; Katoh et al., 2009, Methods in Molecular Biology 537:_39-64; Katoh and Toh, 2010, Bioinformatics 26:_1899-1900), and EMBOSS EMMA employing ClustalW (1 .83 or later; Thompson et al., 1994, Nucleic Acids Research 22: 4673-4680), using their respective default parameters.
  • MUSCLE
  • SEQ ID NO: 2 such that traditional sequence-based comparison fails to detect their relationship (Lindahl and Elofsson, 2000, J. Mol. Biol. 295: 613-615), other pairwise sequence comparison algorithms can be used. Greater sensitivity in sequence-based searching can be attained using search programs that utilize probabilistic representations of polypeptide families (profiles) to search databases. For example, the PSI-BLAST program generates profiles through an iterative database search process and is capable of detecting remote homologs (Atschul et al., 1997, Nucleic Acids Res. 25: 3389-3402). Even greater sensitivity can be achieved if the family or superfamily for the polypeptide has one or more representatives in the protein structure databases.
  • GenTH READER Programs such as GenTH READER (Jones, 1999, J. Mol. Biol. 287: 797-815; McGuffin and Jones, 2003, Bioinformatics 19: 874-881 ) utilize information from a variety of sources (PSI-BLAST, secondary structure prediction, structural alignment profiles, and solvation potentials) as input to a neural network that predicts the structural fold for a query sequence.
  • sources PSI-BLAST, secondary structure prediction, structural alignment profiles, and solvation potentials
  • Gough et al., 2000, J. Mol. Biol. 313: 903-919 can be used to align a sequence of unknown structure with the superfamily models present in the SCOP database. These alignments can in turn be used to generate homology models for the polypeptide, and such models can be assessed for accuracy using a variety of tools developed for that purpose.
  • proteins of known structure For proteins of known structure, several tools and resources are available for retrieving and generating structural alignments. For example the SCOP superfamilies of proteins have been structurally aligned, and those alignments are accessible and downloadable.
  • Two or more protein structures can be aligned using a variety of algorithms such as the distance alignment matrix (Holm and Sander, 1998, Proteins 33: 88-96) or combinatorial extension (Shindyalov and Bourne, 1998, Protein Engineering 11 : 739-747), and implementation of these algorithms can additionally be utilized to query structure databases with a structure of interest in order to discover possible structural homologs (e.g., Holm and Park, 2000, Bioinformatics 16: 566-567).
  • albumin variants of the present invention the nomenclature described below is adapted for ease of reference.
  • the accepted lUPAC single letter or three letter amino acid abbreviation is employed.
  • the term 'point mutation' and/or 'alteration' includes deletions, insertions and substitutions.
  • an insertion may be to the N-side ('upstream', 'X-1 ') or C-side ('downstream', 'X+1 ') of the amino acid occupying a position ('the named (or original) amino acid', 'X').
  • the inserted amino acid residue(s) are numbered by the addition of lower case letters to the position number of the amino acid residue preceding the inserted amino acid residue(s).
  • the sequence would thus be:
  • variants comprising multiple alterations are separated by addition marks ("+"), e.g., "Arg170Tyr+Gly195Glu” or “R170Y+G195E” representing a substitution of arginine and glycine at positions 170 and 195 tyrosine and glutamic acid, respectively.
  • the present invention relates to albumin variants, comprising an alteration at two or more positions selected among the group consisting of positions 492, 550, 573, 574 and 580 of the mature polypeptide of S EQ I D N O : 2, or at equivalent positions in other albumins or fragments thereof.
  • a first aspect of the invention provides polypeptides which are variant albumins or fragments thereof, or fusion polypeptides comprising the variant albumin or fragments thereof, of a parent albumin, comprising alterations at two or more positions corresponding to positions selected among the group consisting of positions 492, 550, 573, 574 and 580 of the mature polypeptide of SEQ ID NO: 2. It is preferred that the two or more alterations comprise alterations at positions corresponding to the following positions in SEQ ID NO: 2:
  • the polypeptide may comprise, three or more, four or more or five or more alterations as described in paragraphs (a), (b), (c), (d), (e), (f), (g), (h), (i) and ( ).
  • a preferred alteration is a substitution
  • parent albumin and/or the variant albumin comprises or consists of:
  • polypeptide encoded by a polynucleotide that hybridizes under low stringency conditions with (i) the mature polypeptide coding sequence of SEQ ID NO: 1 , or (ii) the full-length complement of (i);
  • the variants of albumin or fragments thereof or fusion polypeptides comprising albumin or fragments thereof comprise alterations, such as substitutions, deletions or insertions at two or more of positions selected among the group consisting of positions 492, 550, 573, 574 and 580 of the mature polypeptide of SEQ I D NO: 2 or in equivalent positions of other albumins or variants or fragments thereof.
  • a stop codon may be introduced in addition to the alterations described herein and if introduced is at position 574 or further downstream (e.g. in SEQ ID NO: 2 it is introduced at from position 574 to 585).
  • the variant albumin, a fragment thereof, or albumin part of a fusion polypeptide comprising variant albumin or a fragment thereof according to the invention has generally a sequence identity to the sequence of HSA shown in SEQ ID NO: 2 of at least 60%, preferably at least 70%, preferably at least 80%, preferably at least 85%, preferably at least 90 %, more preferred at least 95%, more preferred at least 96%, more preferred at least 97%, more preferred at least 98% and most preferred at least 99%.
  • the variant has less than 100% identity to SEQ ID NO: 2.
  • the variant albumin, a fragment thereof, or albumin part of a fusion polypeptide comprising variant albumin or a fragment thereof according to the invention has generally a sequence identity to the sequence of the parent albumin of at least 60%, preferably at least 70%, preferably at least 80%, preferably at least 85%, preferably at least 90 %, more preferred at least 95%, more preferred at least 96%, more preferred at least 97%, more preferred at least 98% and most preferred at least 99%.
  • the variant has less than 100% identity to the sequence of the parent albumin.
  • the number of alterations in the variants of the invention is 1 to 20, e.g., 1 to 10 and 1 to 5, such as 1 , 2, 3, 4, 5, 6, 7, 8, 9 or 10 alterations relative to SEQ ID NO: 2 or relative to the sequence of the parent albumin.
  • the alteration is a substitution, such as from the native amino acid to A, C, D, E, F, G, H, I, K, L, M, N, P, Q, R, S, T, V, W, Y, more preferred to G, D, F, H, M or R, even more preferred to G, or D and most preferred to G.
  • the native amino acid at position 492 is E, therefore a substitution to E is not preferred.
  • the alteration is a substitution, such as from the native amino acid to A, C, D, E, F, G, H, I, K, L, M, N, P, Q, R, S, T, V, W, Y, more preferred to K, L, M, E or R, even more preferred to K, L or M and most preferred to K.
  • the native amino acid at position 550 is D, therefore a substitution to D is not preferred.
  • the alteration is a substitution, such as from the native amino acid to A, C, D, E, F, G, H, I, K, L, M, N, P, Q, R, S, T, V, W, Y, more preferred to P, Y, W, H, F, T, I or V, even more preferred to P, Y or W and most preferred to P.
  • the native amino acid at position 573 is K, therefore a substitution to K is not preferred.
  • the alteration is a substitution, such as from the native amino acid to A, C, D, E, F, G, H, I, K, L, M, N, P, Q, R, S, T, V, W, Y, more preferred to H, G, D, F, N, S or Y, even more preferred to D, F, G or H and most preferred to H.
  • the native amino acid at position 574 is K, therefore a substitution to K is not preferred.
  • the alteration is a substitution, such as from the native amino acid to A, C, D, E, F, G, H, I, K, L, M, N, P, Q, R, S, T, V, W, Y, more preferred to K or R, most preferred to K.
  • the native amino acid at position 580 is Q, therefore a substitution to Q is not preferred.
  • a variant albumin may comprise alterations at positions corresponding to positions 492+550 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492D+550K (e.g. SEQ I D NO: 231 ), or 492G+550K (e.g. SEQ ID NO: 240).
  • a variant albumin may comprise alterations at positions corresponding to positions 492+573 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof).
  • Such alterations may comprise 492F+573P (e.g. SEQ ID NO: 109), 492G+573P (e.g. SEQ ID NO: 1 10), 492H+573P (e.g. SEQ ID NO: 1 1 1 ) or 492R+573P (e.g. SEQ I D NO: 1 13) or more preferably 492D+573P (e.g. SEQ ID NO: 108).
  • the variant does not consist of SEQ I D NO: 2 with only alterations 492G+573A, 492G+573A, 492G+N503K+573A, 492G+N503H+573A, 492G+573P, 492G+N503K+573 P o r 492G+N503H+573P.
  • a variant albumin may comprise alterations at positions corresponding to positions 492+574 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492D+574H (e.g. SEQ I D NO: 232), 492G+574H (e.g. SEQ ID NO: 241 ) or 492D+574H (e.g. SEQ ID NO: 232).
  • 492D+574H e.g. SEQ I D NO: 232
  • 492G+574H e.g. SEQ ID NO: 241
  • 492D+574H e.g. SEQ ID NO: 232
  • a variant albumin may comprise alterations at positions corresponding to positions 492+580 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof).
  • a variant albumin may comprise alterations at positions corresponding to positions 550+573 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 550K+573P (e.g. SEQ ID NO: 1 17).
  • a variant albumin may comprise alterations at positions corresponding to positions 550+574 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 550K+574H (e.g. SEQ ID NO: 130), 550M+574H
  • 550M+574H e.g. SEQ ID NO: 249
  • 550L+574H e.g. S EQ I D NO: 245
  • a variant albumin may comprise alterations at positions corresponding to positions 550+580 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 550K+580K (e.g. SEQ ID NO: 131 ), 550M+580K (e.g. SEQ ID NO: 251 ) or 550L+580K (e.g. SEQ ID NO: 247).
  • 550K+580K e.g. SEQ ID NO: 131
  • 550M+580K e.g. SEQ ID NO: 251
  • 550L+580K e.g. SEQ ID NO: 247.
  • a variant albumin may comprise alterations at positions corresponding to positions 573+574 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof).
  • Such alterations may comprise 574D+573P (e.g. SEQ I D NO: 121 ), 574F+573P (e.g. SEQ ID NO: 122), 574G+573P (e.g. SEQ ID NO: 123), 574H+573P (e.g . S EQ I D NO: 124), 574N+573P (e.g. SEQ ID NO: 125) or 574S+573P (e . g . S E Q I D N O : 126). It is preferred that the variant does not consist of SEQ I D NO: 2 with only alterations K573P+K574N+A577T+A578R+S579C+Q580K+A581 D+G584A.
  • a variant albumin may comprise alterations at positions corresponding to positions 573+580 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 580K+573P (e . g . S E Q I D N O : 1 2 8 ) o r 580R+573P (e.g. SEQ ID NO: 129). However, it is preferred that the variant does not consist of SEQ ID NO: 2 with only alterations K573P+A577E+A578S+Q580K+A582T.
  • a variant albumin may comprise alterations at positions corresponding to positions 574+580 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof).
  • a variant albumin may comprise alterations at positions corresponding to positions 492+550+573 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492G+550K+573P (e.g. SEQ ID NO: 254) or 492D+550K+573P (e.g. SEQ ID NO: 253).
  • a variant albumin may comprise alterations at positions corresponding to positions
  • SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof).
  • Such alterations may comprise 492G+550K+574H (e.g. SEQ ID NO: 255).
  • a variant albumin may comprise alterations at positions corresponding to positions 492+550+580 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492D+550K+580K (e.g. SEQ ID NO: 258) or 492G+550K+580K (e.g. SEQ ID NO: 259).
  • a variant albumin may comprise alterations at positions corresponding to positions 492+573+574 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492D+573P+574H (e . g . S E Q I D N O : 233).
  • a variant albumin may comprise alterations at positions corresponding to positions 492+573+580 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492D+573P+580K (e.g. SEQ ID NO: 234) or 492G+573P+580K (e.g. SEQ ID NO: 242).
  • a variant albumin may comprise alterations at positions corresponding to positions
  • 492+574+580 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492D+574H+580K (SEQ I D NO: 262) or 492G+574H+580K (e.g. SEQ ID NO: 263).
  • a variant albumin may comprise alterations at positions corresponding to positions 50+573+574 of SEQ ID NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 550K+574H+573P( e . g . S E Q I D N O : 1 3 1 ) , 550L+573P+574H (e.g. SEQ ID NO: 246) or 550M+573P+574H (e.g. SEQ ID NO: 250).
  • a variant albumin may comprise alterations at positions corresponding to positions 550+573+580 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 550L+573P+580K (e . g . S E Q I D N O : 248) or 550M+573P+580K (e.g. SEQ ID NO: 252).
  • a variant albumin may comprise alterations at positions corresponding to positions
  • a variant albumin may comprise alterations at positions corresponding to positions 573+574+580 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 574H+580K+573P (e . g . S E Q I D N O : 135).
  • a variant albumin may comprise alterations at positions corresponding to positions 492+550+573+574 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492G+550K+573P+574H (e.g. SEQ ID NO: 257) or 492D+550K+573P+574H (e.g. SEQ ID NO: 256).
  • a variant albumin may comprise alterations at positions corresponding to positions 492+550+573+580 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492D+550K+573P+580K (e.g . S EQ I D NO: 260) or 492G+550K+573P+580K (e.g. SEQ ID NO: 261 ).
  • a variant albumin may comprise alterations at positions corresponding to positions
  • a variant albumin may comprise alterations at positions corresponding to positions 492+573+574+580 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof).
  • Such alterations may comprise 492D+573P+574H+580K (e.g . S EQ I D NO: 1 14) or 492G+573P+574H+580K (e.g. SEQ I D NO: 1 15), 492F+573P+574G+580K (e.g. SEQ ID NO: 238), 492G+573P+574G+580K ( e . g .
  • a variant albumin may comprise alterations at positions corresponding to positions 550+573+574+580 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 550K+573P+574H+580K (e.g . S EQ I D NO: 265).
  • a variant albumin may comprise alterations at positions corresponding to positions 492+550+573+574+580 of SEQ ID NO: 2 (or equivalent position of other albumins or variants or fragments thereof).
  • Such alterations may comprise 492D+550K+573P+574H+580K (e.g. SEQ ID NO: 266) or 492G+550K+573P+574H+580K (e.g. SEQ I D NO: 267).
  • Such alterations may comprise 492D+550K+573P+574H (e.g. SEQ ID NO: 256).
  • Particularly preferred variants include:
  • SEQ ID NO: 2 such as (i) E492G and Q580K, or (ii) E492D and Q580K (or equivalent positions of other albumins or variants or fragment thereof);
  • a variant albumin comprising alterations at positions corresponding to positions 492 and 574 in SEQ I D NO: 2, such as (i) E492G and K574H, (ii) E492D and K574H , (iii) E492D and K574K, or (iv) E492G and K574K (or equivalent positions of other albumins or variants or fragment thereof);
  • a variant albumin comprising alterations at positions corresponding to positions 492 and 550 in SEQ I D NO: 2, such as (i) E492G and D550K, or (ii) E492D and D550K (or equivalent positions of other albumins or variants or fragment thereof);
  • a variant albumin comprising alterations at positions corresponding to positions 550 and
  • SEQ ID NO: 2 such as (i) D550K and K573P, (ii) D550L and K573P or (iii) D550M and K573P (or equivalent positions of other albumins or variants or fragment thereof);
  • a variant albumin comprising alterations at positions corresponding to positions 550 and
  • SEQ I D NO: 2 such as (i) D550K and K574H, or (ii) D550L and K574H (or equivalent positions of other albumins or variants or fragment thereof);
  • a variant albumin comprising alterations at positions corresponding to positions 550 and 580 in SEQ I D NO: 2, such as (i) D550M and Q580K, (ii) D550L and Q580K or (iii) D550K and Q580K (or equivalent positions of other albumins or variants or fragment thereof);
  • va ri ant albumin with alterations (e.g. comprising alterations) at positions corresponding to positions 580 and 573 in SEQ I D NO: 2, such as Q580K and K573P (or equivalent positions of other albumins or variants or fragment thereof) and preferably one or more (several) other alterations such at a position selected from 492, 550 and 574. If there is a K at position 580 and a P at position 573 it is preferred that the polypeptide comprises an additional alteration at a position selected from the group consisting of 492, 550, and 574;
  • a variant albu m i n with alterations e.g. comprising alterations
  • alterations e.g. comprising alterations
  • a variant albumin comprising alterations at positions corresponding to positions 492 and 573 in SEQ ID NO: 2, such as (i) E492D and K573P (or equivalent positions of other albumins or variants or fragment thereof) or (ii) E492G and K573P or (iii) E492G and K573A (or equivalent positions of other albumins or variants or fragment thereof), and preferably one or more (several) other alterations such at a position selected from 550, 574 and 580. If there is a variant albumin comprising alterations at positions corresponding to positions 492 and 573 in SEQ ID NO: 2, such as (i) E492D and K573P (or equivalent positions of other albumins or variants or fragment thereof) or (ii) E492G and K573P or (iii) E492G and K573A (or equivalent positions of other albumins or variants or fragment thereof), and preferably one or more (several) other alterations such at
  • polypeptide comprises an additional alteration at a position selected from the group consisting of 550, 574 and 580 (or equivalent positions of other albumins or variants or fragment thereof);
  • a variant albumin comprising alterations at positions corresponding to positions 573 and 574 in SEQ ID NO: 2, such as K573P and K574H (or equivalent positions of other albumins or variants or fragment thereof, and preferably one or more (several) alterations such as at a position selected from 492, 550 and 580. If there is a P at position 573 and an N at position 574 it is preferred that the polypeptide comprises an additional alteration at a position selected from the group consisting of 492, 550, and 580 (or equivalent positions of other albumins or variants or fragment thereof);
  • a variant albumin comprising alterations at positions corresponding to positions 574 and 580 in SEQ ID NO: 2, such as 574H and 580K (or equivalent positions of other albumins or variants or fragment thereof, and preferably one or more (several) alterations such as at a position selected from 492, 550 and 573. If there is a N at position 574 and a K at position 580 it is preferred that the polypeptide comprises an additional alteration at a position selected from the group consisting of 492, 550, and 573 (or equivalent positions of other albumins or variants or fragment thereof);
  • a variant albumin comprising alterations at positions corresponding to positions 492, 550 and 573 in SEQ ID NO: 2, such as E492G, D550K and K573P e.g. SEQ ID NO: 254 or such as E492D, D550K and K573P e.g. SEQ ID NO: 253;
  • variant albumin comprising alterations at positions corresponding to positions 550, 573 and 580 in SEQ ID NO: 2, such as D550K, K573P and Q580K e.g. SEQ ID NO: 133;
  • variant albumin comprising alterations at positions corresponding to positions 550, 573 and 574 in SEQ ID NO: 2, such as D550L, K573P and K574H e.g. SEQ ID NO:246;
  • a variant albumin comprising alterations at positions corresponding to positions 550, 573 and 574 in SEQ ID NO: 2, such as D550K, K573P and K574H e.g. SEQ ID NO: 131 ;
  • variant albumin comprising alterations at positions corresponding to positions 492, 573 and 580 in SEQ ID NO: 2, such as E492G, K573P and Q580K e.g. SEQ ID NO: 242;
  • variant albumin comprising alterations at positions corresponding to positions 492, 573 and 580 in SEQ ID NO: 2, such as E492D, K573P and Q580K e.g. SEQ ID NO: 234;
  • variant albumin comprising alterations at positions corresponding to positions 492, 550, 573 and 574 in SEQ I D NO: 2, such as E492D, D550K, K573P and K574H e.g. SEQ ID NO: 256;
  • variant albumin comprising alterations at positions corresponding to positions 492,
  • SEQ I D NO: 257 such as E492G, D550K, K573P and K574H e.g. SEQ ID NO: 257;
  • variant albumin comprising alterations at positions corresponding to positions 573, 574 and 580 in SEQ ID NO: 2, such as K573P, K574H and Q580K e.g. SEQ ID NO: 135;
  • a variant albumin with alterations e.g. comprising alterations
  • a variant albumin with alterations e.g. comprising alterations
  • positions corresponding to positions 492, 573, 574 and 580 in SEQ ID NO: 2 such as E492G, K573P, K574H and Q580K (or equivalent positions of other albumins or variants or fragment thereof), e.g. SEQ ID NO: 1 15.
  • variant albumin does not consist of SEQ ID NO: 2 with only the following alterations: K573P+K574N+A577T+A578R+S579C+Q580K+A581 D+G584A;
  • E492G+N503H+K573A E492G+K573P; E492G+N503K+K573P; E492G+N503H+K573P.
  • the variant albumin does not consist of SEQ I D NO: 2 with amino aci d s 573 to 585 replaced with (a ) KKLVAASQAALG L , (b) P KFVAASQAALA, (c)
  • PNLVTRCKDALA (d) PKLVESSKATLG or (e) PKLVASTQAALA.
  • the variant albu min , a fragment thereof or fusion polypeptide comprising the variant albumin or fragment thereof has altered binding affinity to FcRn and/or an altered plasma half-life compared with the corresponding parent or reference albumin, fragment thereof, or fusion polypeptide comprising the variant albumin or fragment thereof and/or an altered binding affinity to FcRn.
  • parent or reference albumin is HSA (SEQ ID NO: 1)
  • variant albumin a fragment thereof or fusion polypeptide comprising the variant albumin or fragment thereof has altered binding affinity to FcRn and/or an altered plasma half- life compared with the HSA, the corresponding fragment or fusion polypeptide comprising HSA or fragment thereof and/or an altered binding affinity to FcRn.
  • FcRn than the affinity of WT HSA to FcRn.
  • transgenic mouse having the natural mouse FcRn replaced with human FcRn has a higher serum albumin level than normal mouse (J Exp Med.
  • One way to determine whether the affinity of a variant albumin to FcRn is higher or lower than the parent or reference albumin is to use the Surface Plasmon Resonance assay (SPR) as described below.
  • SPR Surface Plasmon Resonance assay
  • the skilled person will understand that other methods might be useful to determine whether the affinity of a variant albumin to FcRn is higher or lower than the affinity of the parent or reference albumin to FcRn, e.g., determination and comparison of the binding constants KD.
  • the binding affinity (KD) between a first molecule (e.g. ligand) and a second molecule e.g.
  • variant albumins having a KD that is lower than the KD for natural HSA is considered to have a higher plasma half-life than HSA and variant albumins having a KD that is higher than the KD for natural HSA is considered to have a lower plasma half-life than HSA.
  • the variants of albumin or fragments thereof, or fusion polypeptides comprising variant albumin or a fragment thereof according to the invention have a plasma half-life that is longer than the plasma half-life of the parent or reference albumin fragment thereof or fusion polypeptide comprising the parent or reference albumin or a fragment thereof and/or an stronger binding affinity to FcRn.
  • variants of albumin or fragments thereof, or fusion polypeptides comprising variant albumin or fragments thereof according to the invention have a plasma half-life that is shorter than the plasma half-life of the parent or reference albumin fragment thereof or fusion polypeptide comprising the parent or reference albumin or a fragment thereof and/or an weaker binding affinity to FcRn.
  • the variant albumin or fragments thereof, or fusion polypeptides comprising variant albumin or fragments thereof according to the invention may contain additional substitutions, deletions or insertions in other positions of the molecules.
  • additional substitutions, deletions or insertions may be useful in order to alter other properties of the molecules such as but not limited to altered glycosylation ; i ntrod uction of reactive grou ps of the su rface such a thiol groups, removing/generating a carbamoylation site; etc.
  • Residues that might be altered in order to provide reactive residues on the surface and which advantageously could be applied to the invention has been disclosed in WO2010/092135 (incorporated herein by reference). Particular preferred residues include the positions corresponding to positions in SEQ ID NO: 2.
  • cysteine residue may be added to the N or C terminal of albumin.
  • the term 'reactive thiol' means and/or includes a thiol group provided by a Cys which is not disulphide bonded to a Cysteine and/or which is sterically available for binding to a partner such as a conjugation partner.
  • a second aspect of the invention relates to fusion polypeptides. Therefore, the variants of albumin or fragments thereof according to the invention may be fused with a non-albumin polypeptide fusion partner.
  • the fusion partner may in principle be any polypeptide but generally it is preferred that the fusion partner is a polypeptide having therapeutic, prophylactic (including vaccine), diagnostic, imaging or other beneficial properties. Such properties may be referred to as 'pharmaceutically beneficial properties' .
  • Fusion polypeptides comprising albumin or fragments thereof are known in the art. It has been found that such fusion polypeptides comprising albumin or a fragment thereof and a fusion partner polypeptide have a longer plasma half-life compared to the unfused fusion partner polypeptide alone.
  • 'Alter' includes both increasing the plasma half-life or decreasing the plasma half-life. Increasing the plasma half-life is preferred. The invention allows tailoring of half-life to a term desired.
  • One or more (several) therapeutic, prophylactic (including vaccine), diagnostic, imaging or other beneficial polypeptides may be fused to the N-terminus, the C-terminus of albumin, inserted into a loop in the albumin structure or any combination thereof. It may or it may not comprise linker sequences separating the various components of the fusion polypeptide.
  • WO 2001 /79271 A (particularly page 9 and/or Table 1 ), WO 2003/59934 (particularly Table 1 ), WO03/060071 (particularly Table 1 ) and WO01/079480 (particularly Table 1 ) (each incorporated herein by reference in their entirety) also contain examples of therapeutic, prophylactic (including vaccine), diagnostic, imaging or other beneficial polypeptides that may be fused to albumin or fragments thereof, and these examples apply also to the invention.
  • a third aspect of the invention relates to isolated polynucleotides that encode any of the variants or fusion polypeptides of the invention .
  • the polynucleotide may be an isolated polynucleotide.
  • the polynucleotide may be comprised in a vector (such as a plasmid) and/or in a host cell.
  • a fourth aspect of the invention relates to n ucleic acid constructs comprising a polynucleotide encoding a variant or fusion polypeptide of the invention operably linked to one or more (several) control sequences that direct the expression of the coding sequence in a suitable host cell under conditions compatible with the control sequences.
  • a polynucleotide may be manipulated in a variety of ways to provide for expression of a variant. Manipulation of the polynucleotide prior to its insertion into a vector may be desirable or necessary depending on the expression vector. The techniques for modifying polynucleotides utilizing recombinant DNA methods are well known in the art.
  • the control sequence may be a promoter sequence, which is recognized by a host cell for expression of the polynucleotide.
  • the promoter sequence contains transcriptional control sequences that mediate the expression of the variant.
  • the promoter may be any nucleic acid sequence that shows transcriptional activity in the host cell including mutant, truncated, and hybrid promoters, and may be obtained from genes encoding extracellular or intracellular polypeptides either homologous or heterologous to the host cell.
  • useful promoters are obtained from the genes for Saccharomyces cerevisiae enolase (ENO-1 ), Saccharomyces cerevisiae protease A (PRA1 ), Saccharomyces cerevisiae protease B (PRB1 ), Saccharomyces cerevisiae translation elongation factor (TEF1 ),
  • ENO-1 Saccharomyces cerevisiae enolase
  • PRA1 Saccharomyces cerevisiae protease A
  • PRB1 Saccharomyces cerevisiae protease B
  • TEF1 Saccharomyces cerevisiae translation elongation factor
  • Saccharomyces cerevisiae translation elongation factor TEF2
  • Saccharomyces cerevisiae galactokinase GAL1
  • ADH1 3-phosphate dehydrogenase
  • ADH2/GAP Saccharomyces cerevisiae triose phosphate isomerase
  • TPI Saccharomyces cerevisiae metallothionein
  • CU P1 Saccharomyces cerevisiae 3-phosphoglycerate kinase.
  • Other useful promoters for yeast host cells are described by Romanos et al., 1992, Yeast 8: 423-488.
  • useful promoters are obtained from cauliflower mosaic virus 35S RNA gene (CaMV35S), maize alcohol dehydrogenase (Adh1 ) and alpha Amy3.
  • useful promoters are obtained from Cytomegalovirus (CMV) and CAG hybrid promoter (hybrid of CMV early enhancer element and chicken beta-actin promoter), Simian vacuolating virus 40 (SV40).
  • CMV Cytomegalovirus
  • CAG hybrid promoter hybrid of CMV early enhancer element and chicken beta-actin promoter
  • SV40 Simian vacuolating virus 40
  • the control sequence may also be a suitable transcription terminator sequence, which is recognized by a host cell to terminate transcription.
  • the terminator sequence is operably linked to the 3'-terminus of the polynucleotide encoding the variant. Any terminator that is functional in the host cell may be used.
  • P referred term i n ators for yeast h ost cel ls are obtai n ed from th e gen es for Saccharomyces cerevisiae enolase, Saccharomyces cerevisiae cytochrome C (CYC 1 ), Saccharomyces cerevisiae alcohol dehydrogenase (ADH 1 ) and Saccharomyces cerevisiae glyceraldehyde-3-phosphate dehydrogenase.
  • Other useful terminators for yeast host cells are described by Romanos et al., 1992, supra. The skilled person knows useful terminators for use in rice and mammalian cells, such as CHO or H EK.
  • preferred terminators are obtained from Agrobacterium tumefaciens nopaline synthase (Nos) and cauliflower mosaic virus 35S RNA gene (CaMV35S).
  • the control sequence may also be a suitable leader sequence, a nontranslated region of an mRNA that is important for translation by the host cell.
  • the leader sequence is operably linked to the 5'-terminus of the polynucleotide encoding the variant. Any leader sequence that is functional in the host cell may be used.
  • Suitable leaders for yeast host cells are obtained from the genes for Saccharomyces cerevisiae enolase (E N O-1 ), Saccharomyces cerevisiae 3-phosphoglycerate kinase, Saccharomyces cerevisiae alpha-facto r, a n d Saccharomyces cerevisiae alcohol dehydrogenase/glyceraldehyde-3-phosphate dehydrogenase (ADH2/GAP).
  • the control sequence may also be a polyadenylation sequence, a sequence operably linked to the 3'-terminus of the variant-encoding sequence and, when transcribed, is recognized by the host cell as a signal to add polyadenosine residues to transcribed mRNA. Any polyadenylation sequence that is functional in the host cell may be used.
  • the control sequence may also be a signal peptide coding region that encodes a signal peptide linked to the N-terminus of a variant and directs the variant into the cell's secretory pathway.
  • the 5'-end of the coding sequence of the polynucleotide may inherently contain a signal peptide coding region naturally linked in translation reading frame with the segment of the coding region that encodes the variant.
  • the 5'-end of the coding sequence may contain a signal peptide coding region that is foreign to the coding sequence.
  • the foreign signal peptide coding region may be required where the coding sequence does not naturally contain a signal peptide coding region.
  • the foreign signal peptide coding region may simply replace the natural signal peptide coding region in order to enhance secretion of the variant.
  • any signal peptide coding region that directs the expressed variant into the secretory pathway of a host cell may be used.
  • the propeptide region is positioned next to the N-terminus of the variant and the signal peptide region is positioned next to the N-terminus of the propeptide region.
  • a fifth aspect of the invention relates to a method for preparing or obtaining a variant albumin or fragment thereof, or fusion polypeptides comprising the variant albumin or fragments thereof, or associates of variant albumin or fragment thereof comprising:
  • the resultant variant albumin or fragment thereof may have altered FcRn-binding affinity compared to the FcRn-binding affinity of a reference such as a parent albumin or fragment which does not comprise the alterations. More preferably, the resultant variant albumin or fragment thereof has a stronger FcRn-binding affinity.
  • the invention includes a method for preparing a polypeptide which is a variant of albumin, fragment thereof or fusion polypeptide comprising said variant albumin or fragment thereof having a binding affinity to FcRn which is altered compared to the binding affinity of a reference albumin, fragment or fusion thereof to FcRn, comprising:
  • step (b) modifying the sequence of step (a), to encode a polypeptide which is a variant albumin, fragment thereof or fusion polypeptide comprising said variant albumin or fragment thereof comprising alterations at two or more positions corresponding to positions selected from selected among two or more of the group consisting of positions 492, 550, 573, 574 and 580 in SEQ ID NO: 2;
  • step (c) optionally, introducing the modified sequence of step (b) in a suitable host cell;
  • polypeptide has an altered binding affinity to FcRn and/or an altered plasma half-life compared with the half-life of a parent albumin, reference albumin, fragment thereof or fusion polypeptide comprising said parent albumin, reference albumin or fragment or fusion thereof.
  • parent albumin and/or the variant albumin comprises or consists of:
  • variants can be prepared by those skilled persons using any mutagenesis procedure known in the art, such as site-directed mutagenesis, synthetic gene construction, semi-synthetic gene construction, random mutagenesis, shuffling, etc.
  • Site-directed mutagenesis is a technique in which one or more (several) mutations (alterations) are created at one or more (several) defined sites in a polynucleotide encoding the parent.
  • Site-directed mutagenesis can be accomplished in vitro by PCR involving the use of oligonucleotide primers containing the desired mutation. Site-directed mutagenesis can also be performed in vitro by cassette mutagenesis involving the cleavage by a restriction enzyme at a site in the plasmid comprising a polynucleotide encoding the parent and subsequent ligation of an oligonucleotide containing the mutation in the polynucleotide. Usually the restriction enzyme that digests at the plasmid and the oligonucleotide is the same, permitting ligation of the plasmid and insert to one another. See, e.g., Scherer and Davis, 1979, Proc. Natl. Acad. Sci. USA 76:
  • Site-directed mutagenesis can also be accomplished in vivo by methods known in the art. See, e.g., U .S. Patent Application Publication NO: 2004/0171 154; Storici et al., 2001 , Nature Biotechnol. 19: 773-776; Kren et al., 1998, Nat. Med. 4: 285-290; and Calissano and Macino, 1996, Fungal Genet. Newslett. 43: 15-16.
  • Any site-directed mutagenesis procedure can be used in the invention.
  • Synthetic gene construction entails in vitro synthesis of a designed polynucleotide molecule to encode a polypeptide of interest. Gene synthesis can be performed utilizing a number of techniques, such as the multiplex microchip-based technology described by Tian et al. (2004, Nature 432: 1 050-1054) and similar technologies wherein oligonucleotides are synthesized and assembled upon photo-programmable microfluidic chips.
  • Single or multiple amino acid substitutions, deletions, and/or insertions can be made and tested using known methods of mutagenesis, recombination, and/or shuffling, followed by a relevant screening procedure, such as those disclosed by Reidhaar-Olson and Sauer, 1988, Science 241 : 53-57; Bowie and Sauer, 1989, Proc. Natl. Acad. Sci. USA 86: 2152-2156; WO 95/17413; or WO 95/22625.
  • Other methods that can be used include error-prone PCR, phage display (e.g., Lowman et al., 1991 , Biochemistry 30: 10832-10837; U.S. Patent NO: 5,223,409; WO 92/06204) and region-directed mutagenesis (Derbyshire et al., 1986, Gene 46: 145; Ner ei a/., 1988, DNA 7: 127).
  • Mutagenesis/shuffling methods can be combined with high-throughput, automated screening methods to detect activity of cloned, mutagenized polypeptides expressed by host cells (Ness et al., 1999, Nature Biotechnology 17: 893-896). Mutagenized DNA molecules that encode active polypeptides can be recovered from the host cells and rapidly sequenced using standard methods in the art. These methods allow the rapid determination of the importance of individual amino acid residues in a polypeptide.
  • Semi-synthetic gene construction is accomplished by combining aspects of synthetic gene construction, and/or site-directed mutagenesis, and/or random mutagenesis, and/or shuffling.
  • Semi-synthetic construction is typified by a process utilizing polynucleotide fragments that are synthesized, in combination with PCR techniques. Defined regions of genes may thus be synthesized de novo, while other regions may be amplified using site-specific mutagenic primers, while yet other regions may be subjected to error-prone PCR or non-error prone PCR amplification. Polynucleotide sub sequences may then be shuffled.
  • a sixth aspect of the invention relates to methods of preparation of a variant according to the invention.
  • the variants of the invention can be prepared using techniques well known to the skilled person. One convenient way is by cloning nucleic acid encoding the parent albumin or a fragment thereof or fusion polypeptide comprising albumin or a fragment thereof, modifying said nucleic acid to introduce the desired substitution(s) at two or more positions corresponding to positions selected from 492, 550, 573, 574 and 580 of the mature polypeptide of SEQ ID NO: 2 (or equivalent positions in other albumins or fragments thereof), preferably as described for the first or fifth aspects of the invention, preparing a suitable genetic construct where the modified nucleic acid is placed in operative connection with suitable regulatory genetic elements, such as promoter, terminator, activation sites, ribosome binding sites etc., introducing the genetic construct into a suitable host organism , culturing the transformed host organism under conditions leading to expression of the variant and recovering the variant. All these techniques are known in the art and it is
  • the variant polypeptide of the invention may also be connected to a signal sequence in order to have the variant polypeptide secreted into the growth medium during culturing of the transformed host organism. It is generally advantageous to have the variant polypeptide secreted into the growth medium in order to ease recovery and purification.
  • Albumins have been successfully expressed as recombinant proteins in a range of hosts including fungi (including but not limited to Aspergillus (WO06066595), Kluyveromyces (Fleer 1991 , Bio/technology 9, 968-975), Pichia (Kobayashi 1998 Therapeutic Apheresis 2, 257-262) and Saccharomyces (Sleep 1990, Bio/technology 8, 42-46)), bacteria (Pandjaitab 2000, J. Allergy Clin. Immunol.
  • fungi including but not limited to Aspergillus (WO06066595), Kluyveromyces (Fleer 1991 , Bio/technology 9, 968-975), Pichia (Kobayashi 1998 Therapeutic Apheresis 2, 257-262) and Saccharomyces (Sleep 1990, Bio/technology 8, 42-46)
  • bacteria Pandjaitab 2000, J. Allergy Clin. Immunol.
  • the variant polypeptide of the invention is preferably produced recombinantly in a suitable host cell.
  • a suitable host cell any host cell capable of producing a polypeptide in su itable amounts may be used and it is within the skills of the average practitioner to select a suitable host cell according to the invention.
  • a preferred host organism is yeast, preferably selected among Saccharomycacae, more preferred Saccharomyces cerevisiae.
  • variant polypeptides of the invention may be recovered and purified from the growth medium using a combination of known separation techniques such as filtration, centrifugation, chromatography, and affinity separation techniques etc. It is within the skills of the average practitioner to purify the variants of the invention using a particular combination of such known separation steps.
  • purification techniques that may be applied to the variants of the invention can be mentioned the teaching of WO00/44772.
  • the variant polypeptides of the invention may be used for delivering a therapeutically beneficial compound (including prophylactically beneficial compound such as a vaccine) to an animal or a human individual in need thereof.
  • a therapeutically beneficial compound including prophylactically beneficial compound such as a vaccine
  • therapeutically beneficial compounds include, but are not limited, to labels and readily detectable compounds for use in diagnostics, such as various imaging techniques; pharmaceutical active compounds such as drugs, or specifically binding moieties such as antibodies.
  • the variants of the invention may even be connected to two or more different therapeutically beneficial compounds, e.g. , an antibody and a drug, which gives the combined molecule the ability to bind specifically to a desired target and thereby provide a high concentration of the connected drug at that particular target.
  • a seventh aspect of the invention relates to conjugates (conjugations). Therefore, the variants of albumin or fragments thereof or fusion polypeptides according to the invention may be conjugated to a second molecule ('conjugation partner') using techniques known within the art.
  • the conjugation partner may be a therapeutic, prophylactic (including vaccine), diagnostic, imaging or other beneficial moiety.
  • Said conjugation partner may be a polypeptide or a non- polypeptide chemical.
  • the conjugation partner may be a polypeptide, chemical (e.g. chemically synthesised drug) or a nucleic acid (e.g. DNA, RNA, siRNA).
  • Said second molecule may comprise a diagnostic or imaging moiety, and in this embodiment the conjugate may be useful as a diagnostic tool such as in imaging; or the second molecule may be a therapeutic or prophylactic (e.g. vaccine) compound and in this embodiment the conjugate may be used for therapeutic or prophylactic (e.g. vaccination) purposes where the conjugate will have the therapeutic or prophylactic properties of the therapeutic or prophylactic compound as well as the desirable plasma half-life provided by the albumin part of the conjugate.
  • Conjugates of albumin and a therapeutic molecule are known in the art and it has been verified that such conjugates have long plasma half-life compared with the non- conjugated, free therapeutic molecule as such.
  • 'Alter' includes both increasing the plasma half-life and decreasing the plasma half-life binding affinity to FcRn and/or increasing the binding affinity and decreasing the binding affinity to FcRn.
  • Increasing the plasma half-life and/or binding affinity to FcRn is preferred.
  • the conjugates may conveniently be linked via a free thiol group present on the surface of HSA (amino acid residue 34 of mature HSA) using well known chemistry.
  • the variant albumin or fragment thereof is conjugated to a beneficial therapeutic or prophylactic (including vaccine) compound and the conjugate is used for treatment of a condition in a patient in need thereof, which condition is responsive to the particular selected therapeutic compound.
  • a beneficial therapeutic or prophylactic (including vaccine) compound and the conjugate is used for treatment of a condition in a patient in need thereof, which condition is responsive to the particular selected therapeutic compound.
  • Techniques for conjugating such a therapeutically useful compou nd to the variant albumin or fragment thereof are known in the art.
  • WO 2009/019314 (incorporated herein by reference in its entirety) discloses examples of techniques suitable for conjugating a therapeutically compound to a polypeptide which techniques can also be applied to the invention.
  • WO 2009/019314 discloses examples of compounds and moieties that may be conjugated to substituted transferrin and these examples may also be applied to the invention. The teaching of WO 2009/019314 is included herein by reference.
  • HSA contains in its natural form one free thiol group (at Cys34) that conveniently may be used for conjugation.
  • the variant albumin or fragment thereof may comprise further modifications provided to generate additional free thiol groups on the surface. This has the benefit that the payload of the variant albumin or fragment thereof is increased so that more than one molecule of the therapeutic (e.g.
  • prophylactic) compound can be conjugated to each molecule of variant albumin or fragment thereof, or two or more (several) different therapeutic compounds may be conjugated to each molecule of variant albumin or fragment thereof, e.g., a compound having targeting properties such as an antibody specific for example a tumour; and a cytotoxic drug conjugated to the variant albumin or fragment thereof thereby creating a highly specific drug against a tumour.
  • a compound having targeting properties such as an antibody specific for example a tumour
  • a cytotoxic drug conjugated to the variant albumin or fragment thereof thereby creating a highly specific drug against a tumour.
  • the conjugation partner may alternatively be conjugated to a fusion polypeptide (described herein), resulting in a molecule comprising a fusion partner fused to the albumin as well as a conjugation partner conjugated to the same albumin or even to the fusion partner.
  • a fusion polypeptide described herein
  • An eighth aspect of the invention relates to associates. Therefore, the variants of albumin or fragments thereof or fusion polypeptides may further be used in form of "associates".
  • associate is intended to mean a compound comprising a variant of albumin or a fragment thereof and another compound bound or associated to the variant albumin or fragment thereof by non-covalent binding.
  • an associate consisting of variant albumin and a lipid associated to albumin by a hydrophobic interaction.
  • Such associates are known in the art and they may be prepared using well known techniques.
  • an associate comprising variant albumin and a taxane, a taxol or taxol derivative (e.g. paclitaxel).
  • associates comprise a therapeutic, prophylactic (including vaccine), diagnostic, imaging or other beneficial moiety.
  • the half-life of an albumin associate according to the invention may be longer or shorter than the half-life of the 'other compound' alone.
  • the half-life of an albumin associate according to the invention may be longer or shorter than the half-life of the analogous / equivalent albumin associate comprising or consisting of a reference albumin such as native HSA (instead of an albumin variant or derivative according to the invention) and the 'other compound'.
  • the binding affinity to FcRn of an albumin associate according to the invention may be stronger or weaker than the binding affinity to FcRn of the analogous / equivalent albumin associate comprising or consisting of a reference albumin such as native HSA (instead of an albumin variant or derivative according to the invention) and the 'other compound'.
  • a ninth aspect of the invention relates to compositions. Therefore the invention is also directed to the use of a variant of albumin or a fragment thereof or fusion polypeptides comprising variant albumin or fragments thereof, or a conjugate comprising a variant of albumin or a fragment thereof, or an associate comprising a variant of albumin or a fragment thereof for the manufacture of a pharmaceutical composition, wherein the variant of albumin or a fragment thereof or fusion polypeptides comprising variant albumin or fragments thereof, or a conjugate comprising a variant of albumin or a fragment thereof, or an associate comprising a variant of albumin or a fragment thereof has an altered binding affinity to FcRn and/or an altered plasma half-life compared with HSA or the corresponding fragment thereof or fusion polypeptide comprising HSA or fragment thereof or conjugate comprising HSA.
  • the corresponding fragment of HSA is intended to mean a fragment of HSA that aligns with and has same number of amino acids as the fragment of the variant albumin with which it is compared.
  • the corresponding fusion polypeptide comprising HSA or conjugate comprising HSA is intended to mean molecules having same size and amino acid sequence as the fusion polypeptide of conjugate comprising variant albumin, with which it is compared.
  • composition may comprise a pharmaceutically acceptable carrier or excipient such as water, polysorbate 80 or those specified in the US Pharmacopoeia for human albumin.
  • a tenth aspect of the invention relates to a nanoparticle comprising a variant, fusion, conjugate, associate, nanoparticle, composition or polynucleotide as disclosed herein.
  • the average diameter of a nano-particle is from 5 to 1000 nm, more preferably 5, 10, 20, 30, 40, 50, 80, 100, 130, 150, 200, 300, 400, 500, 600, 700, 800, 900, or 999 to 5, 10, 20, 30, 40, 50, 80, 1 00, 1 30, 1 50, 200, 300, 400, 500, 600, 700, 800, 900, or 1 000 nm.
  • An advantage of a microparticle less than 200 nm diameter, and more particularly less than 130 nm, is that is amenable to sterilisation by filtration through a 0.2 ⁇ (micron) filter.
  • the average diameter of a micro-particle is from 1 000 nm (1 pm (micron)) to 1 00 pm (micron), more preferably from 1 , 2, 5, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100 to 1 , 2, 5, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100 m (micron).
  • An eleventh aspect of the invention relates to use of a variant albumin, fragment, fusion or conjugate thereof or nanoparticle or associate thereof. Use may be, for example, in a method of treatment, prophylaxis, diagnosis or imaging.
  • the variant albumin or fragments thereof or fusion polypeptides comprising variant albumin or fragments thereof according to the invention have the benefit that their binding affinity to FcRn and/or plasma half-life is altered compared to the parent or reference albumin or fragments thereof or fusion polypeptides comprising parent or reference albumin or fragments thereof.
  • albumin, variant, fragment, fusion, conjugate or associate or composition thereof having a longer plasma half-life than the reference molecule or composition since this would have the benefit that the administration of the albumin, variant, fragment, fusion, conjugate or associate or composition thereof would be needed less frequently or at a reduced dose (and consequently with fewer side effects) compared to the situation where the reference molecule or composition was used.
  • the albumin moiety may comprise one more alterations as disclosed herein.
  • an albumin, variant, fragment, fusion, conjugate or associate or composition thereof having a shorter plasma half-life than the reference molecule or composition since this would have the benefit that the administration of the albumin, variant, fragment, fusion, conjugate or associate or composition thereof can be carried out at a higher dose compared to the situation where the reference molecule or composition was used with the benefit that the administered compound clears from the recipient more quickly than if the reference molecule or composition was used.
  • the albumin moiety may comprise one more alterations as disclosed herein.
  • a conjugate, associate or fusion polypeptide used for imaging purposes in animals or human beings where the imaging moiety has an very short half-life and a conjugate or a fusion polypeptide comprising HSA has a plasma half-life that is far longer than needed for the imaging purposes it would be advantageous to use a variant albumin or fragment thereof of the invention having a shorter plasma half-life than the parent or reference albumin or fragment thereof, to provide conjugates of fusion polypeptides having a plasma half- life that is sufficiently long for the imaging purpose but sufficiently short to be cleared form the body of the particular patient on which it is applied.
  • an associate or fusion polypeptide comprising a therapeutic compound effective to treat or alleviate a particular condition in a patient in need for such a treatment it would be advantageous to use the variant albumin or fragment thereof having a longer plasma half-life than the parent or reference albumin or fragment thereof, to provide associates or conjugates or fusion polypeptides having longer plasma half-lives which would have the benefit that the admin istration of the associate or conjugate or fusion polypeptide of the invention would be needed less frequently or at reduced dose with less side effects compared to the situation where the parent or reference albumin or associates thereof or fragment thereof was used .
  • the invention provides a method of treating a proliferative disease in an individual, comprising administering the individual an effective amount of an associate according to the invention in which the associate comprises a taxane, a taxol or taxol derivative (e.g. paclitaxel).
  • the associate comprises a taxane, a taxol or taxol derivative (e.g. paclitaxel).
  • compositions comprising the variant albumin, associates thereof or fragment thereof, variant albumin fragment or associates thereof or fusion polypeptide comprising variant albumin or fragment thereof according to the invention.
  • the compositions are preferably pharmaceutical compositions.
  • the composition may be prepared using techniques known in the area such as disclosed in recognized handbooks within the pharmaceutical field. Since the albumin, variant, fragment, fusion, conjugate or associate thereof has a binding affinity to FcRn and/or plasma half-life which is modulated (i.e.
  • the composition also has a binding affinity to FcRn and/or modulated plasma half-life relative to an equivalent composition comprising the reference molecule in place of the albumin, variant, fragment, fusion, conjugate or associate thereof as described herein.
  • the composition may be a vaccine.
  • the polypeptide according to the invention may be an active pharmaceutical or an excipient.
  • the composition is provided in unit dosage form.
  • the albumin, variant, fragment, fusion, conjugate or associate thereof has a plasma half-life that is longer than the plasma half-life of the reference molecule e.g. the same composition except that the albu min component (e.g. albumin, variant, fragment, fusion, conjugate or associate) is wild-type albumin (e.g. HSA) or a variant, fragment, fusion, conjugate or associate.
  • the albu min component e.g. albumin, variant, fragment, fusion, conjugate or associate
  • HSA wild-type albumin
  • compositions comprise a variant albumin or a fragment thereof according to the invention and a compound comprising a pharmaceutically beneficial moiety and an albumin binding domain (ABD).
  • ABD means a site, moiety or domain capable of binding to circulating albumin in vivo and thereby conferring transport in the circulation of the ABD and any compound or moiety bound to said ABD.
  • ABD's are known in the art and have been shown to bind very tight to albumin so a compound comprising an ABD bound to albumin will to a certain extent behave as a single molecule.
  • the inventors have realized by using the variant albumin or fragment thereof according to the invention together with a compound comprising a pharmaceutically beneficial moiety and an
  • ABD makes it possible to alter the binding affinity to FcRn and/or plasma half-life of the compound comprising a pharmaceutically beneficial moiety and an ABD compared to the situation where said compound were injected as such in a patient having need thereof or administered in a formulation comprising natural albumin or a fragment thereof.
  • variant albumin or fragments thereof, conjugates comprising variant albumin or a fragment thereof or fusion polypeptide comprising variant albumin or a fragment thereof, or an associate comprising variant albumin or a fragment thereof according to the invention may also be incorporated into nano- or microparticles using techniques well known within the art.
  • a preferred method for preparing nano- or microparticles that may be applied to the variant albumins or fragments thereof according to the invention is disclosed in WO 2004/071536 or WO2008/007146 or Oner & Groves (Pharmaceutical Research, Vol 10(9), 1993, pages 1387 to 1388) which are incorporated herein by reference.
  • a twelfth aspect of the invention provides a method for altering the FcRn-binding affinity or half-life of a molecule comprising:
  • Examples of 'molecule' include those useful in therapy, prophylaxis (including those used in vaccines either as an active pharmaceutical ingredient or as an excipient), imaging and diagnosis, such as those described herein.
  • the variant of albumin or a fragment thereof or fusion polypeptides comprising variant albumin or fragments thereof, fragment thereof, conjugate, nanoparticle, associate or composition may have a plasma half-life that is either longer or shorter, preferably longer, than the plasma half-life than a corresponding albumin or a fragment thereof or fusion polypeptides comprising albumin or fragments thereof, fragment thereof, conjugate, nanoparticle, associate or composition or a binding to FcRn that is stronger or weaker, preferably weaker.
  • the variant of albumin or a fragment thereof or fusion polypeptides comprising variant albumin or fragments thereof, fragment thereof, conjugate, nanoparticle, associate or composition has a plasma half-life that is longer than the plasma half-life of HSA or the corresponding albumin or a fragment thereof or fusion polypeptides comprising albumin or fragments thereof, fragment thereof, conjugate, nanoparticle, associate or composition.
  • this may be expressed as the variant of albumin or a fragment thereof or fusion polypeptides comprising variant albumin or fragments thereof, fragment thereof, conjugate, nanoparticle, associate or composition having a KD to FcRn (e.g. shFcRn) that is lower than the corresponding KD for HSA to FcRn or the corresponding fragment thereof or fusion polypeptide comprising HSA or fragment thereof.
  • a KD to FcRn e.g. shFcRn
  • the KD for the variant of albumin or a fragment thereof or fusion polypeptides comprising variant albumin or fragments thereof, fragment thereof, conjugate, nanoparticle, associate or composition is less than 0.9X KD for HSA to FcRn, more preferred less than 0.5X KD for HSA to FcRn, more preferred less than 0.1 X KD for HSA to FcRn, even more preferred less than 0.05X KD for HSA to FcRn, even more preferred less than 0.02X KD for HSA to FcRn and most preferred less than 0.01 X KD for HSA to FcRn (where X means 'multiplied by').
  • the KD of the variant of albumin or a fragment thereof or fusion polypeptides comprising variant albumin or fragments thereof, fragment thereof, conjugate, nanoparticle, associate or composition may be between the KD of WT albumin (e.g. SEQ ID No. 2) for FcRn and the KD of HSA K573P (SEQ ID No. 3) for FcRn.
  • WT albumin e.g. SEQ ID No. 2
  • HSA K573P SEQ ID No. 3
  • Such KDs represent binding affinities that are higher than the binding affinity between HSA and FcRn. A higher binding affinity indicates a longer half-life, for example plasma half-life.
  • the variant of albumin or a fragment thereof or fusion polypeptides comprising variant albumin or fragments thereof, fragment thereof, conjugate, nanoparticle, associate or composition has a plasma half-life that is shorter than the plasma half-life of HSA or the corresponding fragment thereof or fusion polypeptide comprising HSA or fragment thereof.
  • This may be expressed as the variant of albumin or a fragment thereof or fusion polypeptides comprising variant albumin or fragments thereof, fragment thereof, conjugate, nanoparticle, associate or com position having a KD to FcRn that is higher than the corresponding KD for HSA to FcRn or the corresponding of albumin or a fragment thereof or fusion polypeptides comprising albumin or fragments thereof, fragment thereof, conjugate, nanoparticle, associate or composition.
  • the KD for the variant of albumin or a fragment thereof or fusion polypeptides comprising variant albumin or fragments thereof, fragment thereof, or a conjugate comprising a variant of albumin or a fragment thereof is more than 2X KD for HSA to FcRn, more preferred more than 5X KD for HSA to FcRn, more preferred more than 10X KD for HSA to FcRn, even more preferred more than 25X KD for HSA to FcRn, most preferred more than 50X KD, more than 60X, more than 70X KD, more than 80X, more than 90X or more than 100X KD for HSA to FcRn.
  • the variant of albumin or a fragment thereof or fusion polypeptides comprising variant albumin or fragments thereof, fragment thereof, conjugate, nanoparticle, associate or composition may be a null binder to FcRn.
  • the variant of albumin or a fragment thereof or fusion polypeptides comprising variant albumin or fragments thereof, fragment thereof, or a conjugate or nanoparticle or associate or composition comprising a variant of albumin or a fragment thereof is preferably the variant of albumin or a fragment thereof or fusion polypeptides comprising variant albumin or fragments thereof, fragment thereof, or a conjugate or nanoparticle or associate or composition comprising a variant of albumin or a fragment thereof according to the invention.
  • a lower binding affinity indicates a shorter half-life, for example plasma half-life.
  • One advantage of the invention is that it allows the half-life of albumin, a variant of albumin or a fragment thereof or fusion polypeptides comprising variant albumin or fragments thereof, fragment thereof, conjugate, nanoparticle, associate or composition to be tailored in order to achieve a binding affinity or half-life which meets the needs of the user.
  • FcRn human FcRn, preferably soluble human FcRn, optionally coupled to a tag such as
  • GST or H is, most preferably His such as 6 histidines at the C-terminus of the beta-2- microglobulin (SEQ ID NO: 31 ).
  • Coupling chemistry amine coupling chemistry (e.g. as described in the protocol provided by the manufacturer of the instrument).
  • the coupling may be performed by injecting 20 ⁇ g ml of the protein in
  • HBS-EP buffer 0.15 M NaCI, 0.005% Tween 20 at pH 5.5
  • Regeneration of the surfaces may be done using injections of HBS-EP buffer (0.01 M HEPES,
  • test molecule e.g. HSA or variant
  • the preferred method for determining KD is provided in Example 2.
  • the invention discloses that two or more positions selected among the group consisting of positions 492, 550, 573, 574 and 580 in SEQ ID NO: 2 (and therefore equivalent positions in albumins and fragments from human serum and albumin and non-human serum albumins) may be altered in order to modulate (increase of decrease) the binding affinity and/or half-life e.g. plasma half-life of an albu min , fragment, fusion , conjugate, associate, nanoparticle or composition.
  • An alteration may be a substitution, insertion or deletion. Substitution is preferred.
  • a substitution or insertion may or may not comprise introduction of a conserved amino acid, i.e. conserved in relation to the amino acid at the position of interest.
  • conserved amino acids are shown by the groups of Fig. 3: aliphatic, aromatic, hydrophobic, charged, polar, positive, tiny and small.
  • the alteration is a substitution, such as from the native amino acid to A, C, D, E, F, G, H, I , K, L, M, N, P, Q, R, S, T, V, W, Y, more preferred to G, D, F, H , M or R, even more preferred to G or D and most preferred to G.
  • a substitution to E is not preferred.
  • SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof), it is preferred that the alteration is a substitution, such as from the native amino acid to A, C, D, E, F, G, H, I, K, L, M, N, P, Q, R, S, T, V, W, Y, more preferred to K, L, M, E or R, even more preferred to K, L or M and most preferred to K.
  • the native amino acid at position 550 is D, therefore a substitution to D is not preferred.
  • the alteration is a substitution, such as from the native amino acid to A, C, D, E, F, G, H, I, K, L, M, N, P, Q, R, S, T, V, W, Y, more preferred to P, Y, W, H, F, T, I or V, even more preferred to P, Y or W and most preferred to P.
  • the native amino acid at position 573 is K, therefore a substitution to K is not preferred.
  • the alteration is a substitution, such as from the native amino acid to A, C, D, E, F, G, H, I, K, L, M, N, P, Q, R, S, T, V, W, Y, more preferred to D, F, G, H, N, S or Y, even more preferred to H, D, F or G and most preferred to H.
  • the native amino acid at position 574 is K, therefore a substitution to K is not preferred.
  • SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof), it is preferred that the alteration is a substitution, such as from the native amino acid to A, C, D, E, F, G, H, I, K, L, M, N, P, Q, R, S, T, V, W, Y, more preferred to K or R, most preferred to K.
  • the native amino acid at position 580 is Q, therefore a substitution to Q is not preferred.
  • a variant albumin may comprise alterations at positions corresponding to positions 492+550 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492D+550K (e.g. SEQ I D NO: 231 ), or 492G+550K (e.g. SEQ ID NO: 240).
  • a variant albumin may comprise alterations at positions corresponding to positions 492+573 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof).
  • Such alterations may comprise 492F+573P (e.g. SEQ I D NO: 109), 492G+573P (e.g. SEQ ID NO: 1 10), 492H+573P (e.g. SEQ ID NO: 1 1 1 ) or 492R+573P (e.g. SEQ I D NO: 1 13) or more preferably 492D+573P (e.g. SEQ ID NO: 108).
  • the variant does not consist of SEQ ID NO: 2 with only alterations 492G+K573A, E492G+K573A, E492G+N503K+K573A, E492G+N503H + K573A, E492G+K573P, E492G+N503K+K573P or E492G+N503H+K573P.
  • a variant albumin may comprise alterations at positions corresponding to positions 492+574 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492D+574H (e.g. SEQ I D NO: 232), 492G+574H (e.g. SEQ ID NO: 241 ) or 492D+574H (e.g. SEQ ID NO: 232).
  • 492D+574H e.g. SEQ I D NO: 232
  • 492G+574H e.g. SEQ ID NO: 241
  • 492D+574H e.g. SEQ ID NO: 232
  • a variant albumin may comprise alterations at positions corresponding to positions 492+580 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof).
  • a variant albumin may comprise alterations at positions corresponding to positions 550+573 of SEQ ID NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise D550K+K573P (e.g. SEQ ID NO: 1 17).
  • a variant albumin may comprise alterations at positions corresponding to positions 550+574 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 550K+574H (e.g. SEQ ID NO: 130), 550M+574H
  • 550M+574H e.g. SEQ ID NO: 249
  • 550L+574H e.g. S EQ I D NO: 245
  • a variant albumin may comprise alterations at positions corresponding to positions 550+580 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 550K+580K (e.g. SEQ ID NO: 131 ), 550M+580K (e.g. SEQ ID NO: 251 ) or 550L+580K (e.g. SEQ ID NO: 247).
  • 550K+580K e.g. SEQ ID NO: 131
  • 550M+580K e.g. SEQ ID NO: 251
  • 550L+580K e.g. SEQ ID NO: 247.
  • a variant albumin may comprise alterations at positions corresponding to positions 573+574 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof).
  • Such alterations may comprise 574D+573P (e.g. SEQ ID NO: 121 ), 574F+573P (e.g. SEQ ID NO: 122), 574G+573P (e.g. SEQ ID NO: 123), 574H+573P (e.g . S EQ I D NO: 124), 574N+573P (e.g. SEQ ID NO: 125) or 574S+573P (e . g . S E Q I D N O : 126). It is preferred that the variant does not consist of SEQ I D NO: 2 with only alterations K573P+K574N+A577T+A578R+S579C+Q580K+A581 D+G584A.
  • a variant albumin may comprise alterations at positions corresponding to positions 573+580 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 580K+573P (e . g . S E Q I D N O : 128) or 580R+573P (e.g. SEQ ID NO: 129). However, it is preferred that the variant does not consist of SEQ ID NO: 2 with only alterations K573P+A577E+A578S+Q580K+A582T.
  • a variant albumin may comprise alterations at positions corresponding to positions 574+580 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof).
  • a variant albumin may comprise alterations at positions corresponding to positions 492+550+573 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492G+550K+573P (e.g. SEQ ID NO: 254) or 492D+550K+573P (e.g. SEQ ID NO: 253).
  • a variant albumin may comprise alterations at positions corresponding to positions 492+550+574 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise E492G+D550K+K574H (e.g. SEQ ID NO: 255).
  • a variant albumin may comprise alterations at positions corresponding to positions
  • 492+550+580 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492D+550K+580K (e.g. SEQ ID NO: 258) or 492G+550K+580K (e.g. SEQ ID NO: 259).
  • a variant albumin may comprise alterations at positions corresponding to positions 492+573+574 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492D+573P+574H (e . g . S E Q I D N O : 233).
  • a variant albumin may comprise alterations at positions corresponding to positions 492+573+580 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492D+573P+580K (e.g. SEQ ID NO: 234) or 492G+573P+580K (e.g. SEQ ID NO: 242).
  • a variant albumin may comprise alterations at positions corresponding to positions 492+574+580 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492D+574H+580K (SEQ I D NO: 262) or 492G+574H+580K (e.g. SEQ ID NO: 263).
  • a variant albumin may comprise alterations at positions corresponding to positions 550+573+574 of S EQ I D NO : 2 (or equivalent position of other albumins or variants or fragments thereof).
  • Such alterations may comprise 550K+574H+573P (e.g. SEQ ID NO: 131 ), 550L+573P+574H (e.g. SEQ ID NO: 246) or 550M+573P+574H (e.g. SEQ ID NO: 250).
  • a variant albumin may comprise alterations at positions corresponding to positions 550+573+580 of SEQ ID NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 550L+573P+580K (e.g. SEQ ID NO: 248) or 550M+573P+580K (e.g. SEQ ID NO: 252).
  • a variant albumin may comprise alterations at positions corresponding to positions 550+574+580 of SEQ ID NO: 2 (or equivalent position of other albumins or variants or fragments thereof).
  • a variant albumin may comprise alterations at positions corresponding to positions
  • 573+574+580 of SEQ ID NO: 2 (or equivalent position of other albumins or variants or fragments thereof).
  • Such alterations may comprise 574H+580K+573P (e.g. SEQ ID NO: 135).
  • a variant albumin may comprise alterations at positions corresponding to positions 492+550+573+574 of SEQ ID NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492G+550K+573P+574H (e.g. SEQ ID NO: 257) or492D+550K+573P+574H (e.g. SEQ ID NO: 256).
  • a variant albumin may comprise alterations at positions corresponding to positions 492+550+573+580 of SEQ ID NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492D+550K+573P+580K (e.g. SEQ ID NO: 260)or492G+550K+573P+580K(e.g. SEQ ID NO: 261).
  • a variant albumin may comprise alterations at positions corresponding to positions 492+550+574+580 of SEQ ID NO: 2 (or equivalent position of other albumins or variants or fragments thereof).
  • a variant albumin may comprise alterations at positions corresponding to positions
  • 492+573+574+580 of SEQ ID NO: 2 (or equivalent position of other albumins or variants or fragments thereof).
  • Such alterations may comprise 492D+573P+574H+580K (e.g. SEQ ID NO: 114)or492G+573P+574H+580K (e.g. SEQ ID NO: 115), 492F+573P+574G+580K (e.g. SEQ ID NO: 238), 492G+573P+574G+580K (e.g. SEQ I D NO : 234), 492D+573P+574G+580K (e.g.
  • SEQ ID NO: 235 492F+573P+574H+580R (e.g. SEQ ID NO: 239), 492D+573P+574H+580K (e.g. SEQ ID NO: 264), 492G+573P+574H+580R (e.g. SEQ ID NO: 244), 492D+573P+574H+580R (e.g. SEQ ID NO: 236) or 492F+573P+574H+580K (e.g. SEQ ID NO: 237).
  • a variant albumin may comprise alterations at positions corresponding to positions 550+573+574+580 of SEQ ID NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 550K+573P+574H+580K (e.g. SEQ ID NO: 265).
  • a variant albumin may comprise alterations at positions corresponding to positions 492+550+573+574+580 of SEQ ID NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492D+550K+573P+574H+580K (e.g.
  • SEQ I D NO: 266 or 492G+550K+573P+574H+580K (e.g. SEQ I D NO: 267).
  • Such alterations may comprise 492D+550K+573P+574H (e.g. SEQ ID NO: 256).
  • Particularly preferred variants include:
  • variant albumin comprising alterations at positions corresponding to positions 492 and 580 in SEQ ID NO: 2, such as (i) E492G and Q580K, or (ii) E492D and Q580K (or equivalent positions of other albumins or variants or fragment thereof);
  • a variant albumin comprising alterations at positions corresponding to positions 492 and 574 in SEQ I D NO: 2, such as (i) E492G and K574H, (ii) E492D and K574H , (iii) E492D and K574K, or (iv) E492G and K574K (or equivalent positions of other albumins or variants or fragment thereof);
  • variant albumin comprising alterations at positions corresponding to positions 492 and
  • E492G and D550K such as (i) E492G and D550K, or (ii) E492D and D550K (or equivalent positions of other albumins or variants or fragment thereof);
  • a variant albumin comprising alterations at positions corresponding to positions 550 and
  • SEQ ID NO: 2 such as (i) D550K and K573P, (ii) D550L and K573P or (iii) D550M and K573P (or equivalent positions of other albumins or variants or fragment thereof);
  • a variant albumin comprising alterations at positions corresponding to positions 550 and
  • SEQ I D NO: 2 such as (i) D550K and K574H, or (ii) D550L and K574H (or equivalent positions of other albumins or variants or fragment thereof);
  • a variant albumin comprising alterations at positions corresponding to positions 550 and 580 in SEQ I D NO: 2, such as (i) D550M and Q580K, (ii) D550L and Q580K or (iii) D550K and Q580K (or equivalent positions of other albumins or variants or fragment thereof);
  • a variant albu m i n with alterations at positions corresponding to positions 580 and 573 in SEQ I D NO: 2, such as Q580K and K573P (or equivalent positions of other albumins or variants or fragment thereof) and preferably one or more (several) other alterations such at a position selected from 492, 550 and 574. If there is a K at position 580 and a P at position 573 it is preferred that the polypeptide comprises an additional alteration at a position selected from the group consisting of 492, 550, and 574.
  • a variant albumin with alterations e.g. comprising alterations
  • alterations e.g. comprising alterations
  • SEQ I D NO: 2 such as a variant albumin comprising alterations at positions corresponding to positions 492 and 573 in SEQ I D NO: 2, such as (i) E492D and K573P or (ii) E492G and K573P or (iii) E492G and K573A (or equivalent positions of other albumins or variants or fragment thereof), and preferably one or more (several) other alterations such at a position selected from 550, 574 and 580.
  • the polypeptide comprises an additional alteration at a position selected from the group consisting of 550,574 and 580 (or equivalent positions of other albumins or variants or fragment thereof);
  • a variant albumin with alterations e.g. comprising alterations
  • the polypeptide comprises an additional alteration at a position selected from the group consisting of 492, 550, and 580 (or equivalent positions of other albumins or variants or fragment thereof);
  • a variant albumin comprising alterations at positions corresponding to positions 574 and 580 in SEQ ID NO: 2, such as 574H and 580K (or equivalent positions of other albumins or variants or fragment thereof. If there is a N at position 574 and a K at position 580 it is preferred that the polypeptide comprises an additional alteration at a position selected from the group consisting of 492, 550, and 573 (or equivalent positions of other albumins or variants or fragment thereof);
  • a variant albumin comprising alterations at positions corresponding to positions 492, 550 and 573 in SEQ ID NO: 2, such as E492G, D550K and K573P e.g. SEQ ID NO: 254 or such as E492D, D550K and K573P e.g. SEQ ID NO: 253;
  • variant albumin comprising alterations at positions corresponding to positions 550, 573 and 580 in SEQ ID NO: 2, such as D550K, K573P and Q580K e.g. SEQ ID NO: 133;
  • a variant albumin comprising alterations at positions corresponding to positions 550, 573 and 580 in SEQ ID NO: 2, such as D550L, K573P and Q580K e.g. SEQ ID NO: 248 or such as D550M, K573P and Q580K e.g. SEQ ID NO:252;
  • variant albumin comprising alterations at positions corresponding to positions 550, 573 and 574 in SEQ ID NO: 2, such as D550L, K573P and K574H e.g. SEQ ID NO:246;
  • a variant albumin comprising alterations at positions corresponding to positions 550, 573 and 574 in SEQ ID NO: 2, such as D550K, K573P and K574H e.g. SEQ ID NO: 131 ;
  • variant albumin comprising alterations at positions corresponding to positions 492, 573 and 580 in SEQ ID NO: 2, such as E492G, K573P and Q580K e.g. SEQ ID NO: 242;
  • variant albumin comprising alterations at positions corresponding to positions 492, 573 and 580 in SEQ ID NO: 2, such as E492D, K573P and Q580K e.g. SEQ ID NO: 234;
  • variant albumin comprising alterations at positions corresponding to positions 492, 550, 573 and 574 in SEQ I D NO: 2, such as E492D, D550K, K573P and K574H e.g. SEQ ID NO: 256;
  • variant albumin comprising alterations at positions corresponding to positions 492, 550, 573 and 574 in SEQ I D NO: 2, such as E492G, D550K, K573P and K574H e.g. SEQ I D NO: 257; a variant albumin comprising alterations at positions corresponding to positions 573, 574 and 580 in SEQ ID NO: 2, such as K573P, K574H and Q580K e.g. SEQ ID NO: 135;
  • a variant albumin comprising alterations at positions corresponding to positions 492, 573, 574 and 580 in SEQ I D NO: 2, such as E492D, K573P, K574H and Q580K (or equivalent positions of other albumins or variants or fragment thereof), e.g. SEQ ID NO: 1 14;
  • a variant albumin comprising alterations at positions corresponding to positions 492, 573, 574 and 580 in SEQ ID NO: 2, such as E492G, K573P, K574H and Q580K (or equivalent positions of other albumins or variants or fragment thereof), e.g. SEQ ID NO: 1 15.
  • alteration at position 492 is conserved relative to D or E. It is preferred that the alteration at position 574 is conserved relative to H. It is preferred that the alteration at position 580 is conserved relative to K.
  • the polypeptide retains substantially the same tertiary structure (or, for a fragment, the relevant part of the structure) as a reference or parent albumin such as HSA.
  • a reference or parent albumin such as HSA.
  • the skilled person understand the term 'substantially the same tertiary structure' bearing in mind that some degree of variation in tertiary structure is expected as all proteins have some degree of structural flexibility. This applies particularly to polypeptides having a higher binding affinity to FcRn than the parent or reference albumin (e.g. HSA) has to FcRn.
  • One or more (several) of the His residues may or may not be maintained relative to the parent albumin.
  • one or more (several) of the following His residues may be maintained: 3, 9, 39, 67, 105, 128, 146, 242, 247, 288, 338, 367, 440, 464, 510, 535.
  • One or more (several), preferably all, of the His residues in domain I are maintained ⁇ i.e. 3, 9, 39, 67, 105, 128, 146.).
  • One or more (several), preferably all, of the His residues in domain I are maintained ⁇ i.e. 242, 247, 288, 338, 367).
  • One or more (several), preferably all, of the His residues in domain I II are maintained ⁇ i.e. 440, 464, 510, 535).
  • One or more (several) or all three of His 464, 510, 535 may be maintained.
  • At least 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16 or 17 of the disulphide bonds of the albumin are maintained in the polypeptide.
  • all disulphide bonds usually present in that albumin are maintained.
  • all disulphide bonds usually present in that fragment are maintained.
  • Cys34 (or equivalent in non-human albumins) is maintained.
  • fusion partner polypeptides and/or conjugates may comprise one or more (several) of: 4-1 BB ligand, 5-helix, A human C-C chemokine, A human
  • L105 chemokine A human L105 chemokine designated huL105_3.
  • Complement Com ponent C 1 q C Adenoid-expressed chemokine (ADEC), aFGF; FGF-1 , AGF, AGF Protein, albumin, an etoposide, angiostatin, Anthrax vaccine, Antibodies specific for collapsin, antistasin, Anti-TGF beta family antibodies, antithrombin I I I , APM-1 ; ACRP-30; Famoxin, apo-lipoprotein species, Arylsulfatase B, b57 Protein, BCMA, Beta-thromboglobulin protein (beta-TG), bFGF; FGF2, Blood coagulation factors, BMP Processing Enzyme Fur
  • I L-3 mutant proteins I L-3 variants, IL-3 variants, IL-4, I L-4 mutein, IL-4 mutein Y124G, IL-4 mutein Y124X, IL-4 muteins, II-5 receptor, I L-6, I I-6 receptor, I L-7 receptor clone, I L-8 receptor, I L-9 mature protein variant (Met1 17 version), immunoglobulins or immunoglobulin-based molecules or fragment of either (e.g.
  • SMIP Small Modular ImmunoPharmaceutical
  • dAb Fab' fragments, F(ab')2, scAb, scFv or scFv fragment
  • IP- 10 Interferon gamma-inducible protein
  • interferons such as interferon alpha species and sub-species, interferon beta species and sub-species, interferon gamma species and sub-species
  • interferons such as interferon alpha species and sub-species, interferon beta species and sub-species, interferon gamma species and sub-species
  • interferons such as interferon alpha species and sub-species, interferon beta species and sub-species, interferon gamma species and su b-species
  • Interleukin 6, Interleukin 8 (IL-8) receptor Interleukin 8 receptor B, lnterleukin-1 alpha
  • anticalin(s), adnectin(s), fibrinogen fragment(s), nanobodies such as camelid nanobodies, infestin, and/or any of the molecules mentioned in WO01/79271 (particularly page 9 and/or Table 1 ), WO 2003/59934 (particularly Table 1 ), WO03/060071 (particularly Table 1 ) or WO01/079480 (particularly Table 1 ) (each incorporated herein by reference in their entirety).
  • conjugates may comprise one or more (several) of chemotherapy drugs such as: 13-cis-Retinoic Acid, 2-CdA, 2-Chlorodeoxyadenosine, 5-Azacitidine, 5-Fluorouracil, 5- FU , 6-Mercaptopurine, 6-MP, 6-TG , 6-Thioguanine, A, Abraxane, Accutane ® , Actinomycin-D, Adriamycin ® , Adrucil ® , Agrylin ® , Ala-Cort ® , Aldesleukin, Alemtuzumab, ALIMTA, Alitretinoin, Alkaban-AQ ® , Alkeran ® , Al l-transretinoic Acid, Alpha Interferon, Altretamine, Amethopterin, Amifostine, Aminoglutethimide, Anagrelide, Anandron ® , Anastrozole, Arabinosylcytosine, Ara-C
  • Paclitaxel or Paclitaxel Protein-bound Pamidronate, Panitumumab, Panretin ® , Paraplatin ® , Pediapred ® , P E G I nterferon , Pegaspa rgase , Pegfi lg rasti m , P E G-INTRONTM, PEG-L- asparaginase, PEMETREXED, Pentostatin, Phenylalanine Mustard, Platinol ® , Platinol-AQ ® , Prednisolone, Prednisone, Prelone ® , Procarbazine, PROCRIT ® , Proleukin ® , Prolifeprospan 20 with Carmustine Implant, Purinethol ® , R, Raloxifene, Revlimid ® , Rheumatrex ® , Rituxan ® , Rituximab, Roferon-A ® (Interferon Alfa-2a), Rub
  • fusion partners, conjugation partners and/or molecules for inclusion in a nanoparticle, associate or composition according to the invention include: acromegaly drugs e.g. somatuline, lanreotide, octreotide, Sandostatin; antithrombotics e.g. bivalirudin, Angiomax, dalteparin, Fragmin, enoxaparin, Lovenox, Drotrecogin alfa (e.g. Activated), Xigris, heparin; assisted reproductive therapy compounds e.g. choriogonadotropin, Ovidrel, follitropin, alpha/beta; enzymes e.g.
  • acromegaly drugs e.g. somatuline, lanreotide, octreotide, Sandostatin
  • antithrombotics e.g. bivalirudin, Angiomax, dalteparin, Fragmin, enoxaparin
  • hyaluronidase Hylenex
  • diabetes drugs e.g. exenatide, Byetta, glucagon, insulin, liraglutide, albiglutide, GLP-1 agonists, exendin or an exendin analog
  • compounds useful in diagnosis e.g. protirelin, Thyrel TRH Thypinone, secretin (e.g. synthetic human), Chirhostim, thyrotropin (e.g. alpha), Thyrogen' erythropoiesis drugs e.g. Darbepoetin alfa, Aranesp, Epoetin alfa, Epogen, Eprex, drugs for the treatment of genetic defects e.g.
  • pegademase drugs for the treatment of growth failure e.g. Adagen, mecasermin, rinfabate, drugs for the treatment of cystic fibrosis e.g. Dornase alfa, Pulmozyme, drugs for the treatment of metaoblic disorders e.g. Agalsidase beta, Fabrazyme, alglucosidase alpha, Myozyme, Laronidase, Aldurazyme, drugs for the treatment of genital wart intralesional e.g. Interferon alfa- n3, Alferon N , drugs for the treatment of granulomatous disease e.g.
  • Interferon gamma-1 b Actimmune; drugs for the treatment of growth failure e.g. pegvisomant, Somavert, somatropin, Genotropin, Nutropin, Humatrope,Serostim, Protropin; drugs for the treatment of heart failure e.g. nesiritide, Natrecor; drugs for the treatment of hemophilia e.g. a coagulation factor e.g. Factor VI I I , H el ixate FS , Kogenate FS , Factor IX, BeneFIX, Factor Vi l a , Novoseven, desmopressin, Stimate, DDAVP; hemopoetic drugs e.g.
  • G-CSF Filgrastim
  • Neupogen Oprelvekin
  • Neumega Pegfilgrastim
  • Neulasta Sargramostim
  • Leukine drugs for the treatment of hepatitis C e.g. Interferon alfa-2a, Roferon A, Interferon alfa-2b, Intron A, Interferon alfacon-1 , Infergen, Peginterferon alfa-2a, Pegasys, Peginterferon alfa-2b, PEG-lntron
  • drugs for the treatment of H IV e.g. enfuvirtide, Fuzeon
  • Fabs e.g.
  • Fab antithrombin
  • Abciximab ReoPro
  • monoclonal antibodies e.g. Daclizumab, Zenapax
  • antiviral monoclonal antibodies e.g. Palivizumab, Synagis
  • monoclonal antibodies for the treatment of asthma e.g. Omalizumab, Xolair
  • monoclonal antibodies for use in diagnostic imaging e.g. Arcitumomab, CEA-Scan, Capromab Pendetide, ProstaScint, Satumomab Pendetide, OncoScint CR/OV
  • Fabs for use in diagnostic imaging e.g.
  • Nofetumomab, Verluma iimmuno-supressant monoclonal antibodies e.g. Basiliximab, Simulect, Muromonab-CD3, Orthoclone OKT3; monoclonal antibodies for the treatment of malignancy e.g. Alemtuzumab, Campath, Ibritumomab tiuxetan, Zevalin, Rituximab, Rituxan, Trastuzumab, Herceptin ; monoclonal antibodies for the treatment of rheumatoid arthritis (RA) e.g.
  • RA rheumatoid arthritis
  • Macugen drugs for the treatment of malignancy e.g. Aldesleukin, Proleukin, lnterleukin-2,
  • Asparaginase Elspar, Rasburicase, Elitek, Denileukin diftitox, Ontak, Pegaspargase, Oncaspar, goserelin, leuprolide; drugs for the treatment of multiple sclerosis (MS) e.g. Glatiramer acetate (e.g. copolymer-1 ), Copaxone, Interferon beta-1 a, Avonex, Interferon beta-1 a, Rebif, Interferon beta-1 b, Betaseron; drugs for the treatment of mucositis e.g.
  • MS multiple sclerosis
  • MS multiple sclerosis
  • Glatiramer acetate e.g. copolymer-1
  • Copaxone Interferon beta-1 a
  • Avonex Interferon beta-1 a
  • Rebif Interferon beta-1 b
  • Betaseron drugs for the treatment of mucositis e.g.
  • palifermin, Kepivance drug for the treatment of dystonia e.g., neurotoxin, Botulinum Toxin Type A, BOTOX, BOTOX Cosmetic, Botulinum Toxin Type B, MYOBLOC; drugs for the treatment of osteoporosis e.g. teriparatide,Forteo; drugs for the treatment of psoriasis e.g. Alefacept, Amevive; drugs for the treatment of RA e.g. abatacept, Orencia, Anakinra, Kineret, Etanercept, Enbrel; thrombolytics e.g.
  • Alteplase Activase, rtPA, Anistreplase, Eminase, Reteplase, Retavase, Streptokinase, Streptase, Tenecteplase, TNKase, Urokinase, Abbokinase, Kinlytic; drugs for the treatment of osteoporosis e.g. calcitonin (e.g. salmon), Miacalcin, Fortical, drugs for the treatment of skin ulcers e.g. Becaplermin, Regranex, Collagenase, Santyl.
  • drugs for the treatment of osteoporosis e.g. calcitonin (e.g. salmon), Miacalcin, Fortical, drugs for the treatment of skin ulcers e.g. Becaplermin, Regranex, Collagenase, Santyl.
  • Such polypeptides and chemical compounds may be referred to as diagnostic moieties, therapeutic moieties, prophylactic moieties or beneficial moieties.
  • the fusion partner and/or conjugation partner is not an albumin, variant or fragment thereof.
  • One or more (several) therapeutic or prophylactic polypeptides may be fused to the N- terminus, the C-terminus of albumin, inserted into a loop in the albumin structure or any combination thereof. It may or it may not comprise linker sequences separating the various components of the fusion polypeptide.
  • polypeptide according to embodiment 1 wherein the polypeptide comprises alterations at two or more positions selected from positions corresponding to positions (a) 492 and 580; (b) 492 and 574; (c) 492 and 550; (d) 550 and 573; (e) 550 and 574; (f) 550 and 580 in SEQ ID NO: 2.
  • polypeptide according to embodiment 1 or 2 comprising alterations at positions corresponding to positions 492 and 580 of SEQ I D NO: 1 , further comprising one or more (several) alterations at positions corresponding to positions selected from the group consisting of 550, 573 and 574 of SEQ ID NO: 2.
  • polypeptide according to embodiment 1 , 2 or 3 comprising alterations at positions corresponding to positions 492 and 574 of SEQ I D NO: 1 , further comprising one or more (several) alterations at positions corresponding to positions selected from the group consisting 550, 573 and 580 of SEQ ID NO:
  • polypeptide according to any preceding embodiment comprising alterations at positions corresponding to positions 492 and 550 of SEQ ID NO: 1 , further comprising one or more (several) alterations at positions corresponding to positions selected from the group consisting 573 , 574 and 580of SEQ ID NO: 2.
  • polypeptide according to any preceding embodiment comprising alterations at positions corresponding to positions 550 and 573 of SEQ ID NO: 1 , further comprising one or more (several) alterations at positions corresponding to positions selected from the group consisting 492, 574, and 580 of SEQ ID NO: 2.
  • polypeptide according to any preceding embodiment comprising alterations at positions corresponding to positions 550 and 574 of SEQ ID NO: 1 , further comprising one or more (several) alterations at positions corresponding to positions selected from the group consisting 492, 573 and 580 of SEQ ID NO: 2.
  • polypeptide according to any preceding embodiment comprising alterations at positions corresponding to positions 550 and 580 of SEQ I D NO: 1 , further comprising an alteration at a position corresponding to position 492, 573 and 574 of SEQ ID NO: 2.
  • polypeptide comprises an additional alteration at a position selected from the group consisting of 550, 574 and 580 (or equivalent positions of other albumins or variants or fragment thereof).
  • polypeptide comprises an additional alteration at a position selected from the group consisting of 492, 550, and 580 (or equivalent positions of other albumins or variants or fragment thereof);
  • polypeptide comprises an additional alteration at a position selected from the group consisting of 492, 550, and 573 (or equivalent positions of other albumins or variants or fragment thereof).
  • polypeptide according to any preceding embodiment, wherein the polypeptide comprises alterations at three or more positions selected from positions corresponding to positions 492, 550, 573, 574 and 580 in SEQ ID NO: 2.
  • polypeptide of embodiment 14 wherein the substitution at the position corresponding to position 492 is to G, D, F, H, M or R.
  • polypeptide of any of embodiments 13 to 24 wherein the alteration at the position corresponding to position 574 is a substitution to H, G, D, F, N, S or Y.
  • 580K + 573P e.g. SEQ ID NO: 128)
  • 580R + 573P e.g. SEQ ID NO: 129
  • 574D + 573P e.g.
  • SEQ ID NO: 121 SEQ ID NO: 121
  • 574F + 573P e.g. SEQ ID NO: 122
  • 574G + 573P e.g. SEQ ID NO: 123
  • 574H + 573P e.g. SEQ I D NO: 124
  • 574N + 573P e.g. SEQ ID NO: 125
  • 574S + 573P e.g.
  • SEQ I D NO: 126 550K + 580K (e.g. SEQ I D NO: 132); 550K + 574H (e.g. SEQ ID NO: 130); 550K + 573P (e.g. SEQ I D NO: 1 17); 492D + 573P (e.g. SEQ ID NO: 108); 492F + 573P (e.g.
  • SEQ ID NO: 109 SEQ ID NO: 109
  • 492H + 573P e.g. SEQ ID NO: 1 1 1
  • 492R + 573P e.g. SEQ I D NO: 1 12
  • 574H + 580K e.g. SEQ ID NO: 134
  • 550L + 574H e.g. SEQ ID NO: 245
  • 550L + 580K e.g.
  • SEQ ID NO: 247 ; 550M + 580K (e.g. SEQ ID NO: 251 ); 492D + 550K (e.g. SEQ ID NO: 231 ); 550M + 574H (e.g. SEQ ID NO: 249); 492D + 574H (e.g. SEQ ID NO: 232); 492G + 550K (e.g. SEQ I D NO: 240); 550M + 574H (e.g. SEQ I D NO: 249); or 492G + 574H (e.g. SEQ I D NO: 241 ).
  • 550M + 580K e.g. SEQ ID NO: 251
  • 492D + 550K e.g. SEQ ID NO: 231
  • 550M + 574H e.g. SEQ ID NO: 249
  • 492D + 574H e.g. SEQ ID NO: 232
  • 492G + 550K e.g. S
  • polypeptide according to any preceding embodiment wherein the polypeptide comprises three or more alterations at positions selected from the group consisting of positions corresponding to the following positions of SEQ ID NO: 2: 492, 550, 573, 574 and 580.
  • SEQ I D NO: 242 550M + 573P + 574H (e.g. SEQ I D NO: 250); 492G + 550K + 573P (e.g. SEQ I D NO: 254); 550L + 573P + 574H (e.g. SEQ I D NO: 246); 492D + 573P + 580K (e.g. SEQ I D NO: 234); 492D + 573P + 574H (e.g. SEQ ID NO: 233); 492G + 574H + 580K (e.g. SEQ ID NO: 263); 492G + 550K + 580K (e.g.
  • SEQ ID NO: 259 SEQ ID NO: 259; 492D + 550K + 580K (e.g. SEQ I D NO: 258); 492D + 574H + 580K (e.g. SEQ ID NO: 262); or 492G + 550K + 574H (e.g. SEQ ID NO: 255).
  • polypeptide according to any preceding embodiment wherein the polypeptide comprises four or more alterations at positions selected from the group consisting of positions corresponding to the following positions of SEQ ID NO: 2: 492, 550, 573, 574 and 580.
  • SEQ I D NO: 239 492D + 573P + 574G + 580K (e.g. SEQ ID NO: 235); or 492F + 573P + 574G + 580K (e.g. SEQ ID NO: 238);
  • polypeptide according to any preceding embodiment wherein the polypeptide comprises five or more alterations at positions selected from the group consisting of positions corresponding to the following positions of SEQ ID NO: 2: 492, 550, 573, 574 and 580.
  • polypeptide according to embodiment wherein the polypeptide comprises alterations at positions corresponding to the following positions of SEQ I D NO: 2: 492G + 550K + 573P + 574H + 580K (e.g. SEQ ID NO: 267) or 492D + 550K + 573P + 574H + 580K (e.g. SEQ ID NO: 266).
  • polypeptide according to any preceding embodiment comprising alterations corresponding to the following positions in SEQ ID NO: 2: 492G+573P+574H+580K (e.g. SEQ ID NO: 1 15); 492G+550K+573P+574H (e.g. SEQ ID NO: 257); 492D+550K+573P+574H (e.g. SEQ ID NO: 256); 492G+550K+573P (e.g. SEQ ID NO: 254); 492D+550K+573P (e.g. SEQ ID NO: 253); 550M+573P+580K (e.g.
  • SEQ ID NO: 252 SEQ ID NO: 252; 550L+573P+580K (e.g. SEQ ID NO: 248); 550L+573P+574 H (e.g . SEQ I D NO : 246); 492G+573 P+580 K (e.g. SEQ I D NO: 242); 492D+573P+580 K (e.g . S EQ I D N O: 234); 573 P+574H +580 K (e.g . SEQ I D NO : 1 35); 550K+573P+580K (e.g. SEQ I D NO: 1 33); 550K+573P+574H (e.g. SEQ I D NO: 1 31 ); or 492D+573P+574H+580K (SEQ ID NO: 1 14).
  • polypeptide of any of embodiments 1 to 38 wherein the reference albumin is HSA (SEQ ID No: 2) or a fragment thereof, or a fusion polypeptide comprising HSA or a fragment thereof, most preferably SEQ ID NO: 2.
  • polypeptide according to any of embodiments 1 to 39 having a stronger binding affinity to FcRn and/or longer plasma half-life than a parent albumin, reference albumin, fragment thereof or fusion polypeptide comprising said parent albumin, reference albumin or fragment or fusion thereof.
  • polypeptide according any of embodiments 1 to 40 wherein the sequence identity of the polypeptide to SEQ ID NO: 2 is more than 80%, preferably more than 90%, more preferred more than 95%, more preferred more than 96%, even more preferred more than 97%, more preferred more than 98% and most preferred more than 99%.
  • a fusion polypeptide comprising a polypeptide according to any of embodiments 1 to 41 and a fusion partner polypeptide selected from a therapeutic, prophylactic, diagnostic, imaging or other beneficial moiety.
  • step (b) Modifying the sequence of step (a), to encode a polypeptide which is a variant albumin, fragment thereof or fusion polypeptide comprising said variant albumin or fragment thereof comprising alterations at two or more positions corresponding to positions selected from 492, 550, 573, 574 and 580 in SEQ ID NO: 2, preferably as described in any of embodiments 1 to 13;
  • step (c) Introducing the modified sequence of step (b) in a suitable host cell;
  • polypeptide has an altered binding affinity to FcRn and/or an altered plasma half-life compared with the half-life of a parent albumin, reference albumin, fragment thereof or fusion polypeptide comprising said parent albumin, reference albumin or fragment or fusion thereof.
  • substitution at the position corresponding to position 492 is a substitution to G, D, F, H, M or R, preferably G or D
  • substitution at the position corresponding to position 573 is a substitution to P, Y, W, H, F, T, I or V, preferably P, Y, or W, most preferably P.
  • substitution at the position corresponding to position 574 is a substitution to H, D, F, G, N, S or Y, preferably H, D, F,or G, most preferably H.
  • SEQ ID NO: 125 574S + 573P (e.g. SEQ ID NO: 126); 550K + 580K (e.g. SEQ I D NO: 132); 550K + 574H (e.g. SEQ I D NO: 130); 550K + 573P (e.g. SEQ ID NO: 1 17); 492D + 573P (e.g. SEQ ID NO: 108); 492F + 573P (e.g. SEQ ID NO: 109); 492H + 573P (e.g. SEQ I D NO: 1 1 1 ); 492R + 573P (e.g. SEQ ID NO: 1 12); 574H + 580K (e.g.
  • SEQ ID NO: 134 550L + 574H (e.g. SEQ ID NO: 245); 550L + 580K (e.g. SEQ ID NO: 247); 550M + 580K (e.g. SEQ ID NO: 251 ); 492D + 550K (e.g. SEQ ID NO: 231 ); 550M + 574H (e.g. SEQ ID NO: 249); 492D + 574H (e.g. SEQ ID NO: 232); 492G + 550K (e.g. SEQ ID NO: 240); 550M + 574H (e.g. SEQ ID NO: 249); or 492G + 574H (e.g. SEQ ID NO: 241 ).
  • 550L + 574H e.g. SEQ ID NO: 245
  • 550L + 580K e.g. SEQ ID NO: 247
  • 550M + 580K e.g. SEQ ID NO: 251
  • SEQ ID NO: 242 550M + 573P + 574H (e.g. SEQ ID NO: 250); 492G + 550K + 573P (e.g. SEQ ID NO: 254); 550L + 573P + 574H (e.g. SEQ ID NO: 246); 492D + 573P + 580K (e.g. SEQ ID NO: 234); 492D + 573P + 574H (e.g. SEQ ID NO: 233); 492G + 574H + 580K (e.g. SEQ ID NO: 263); 492G + 550K + 580K (e.g. SEQ ID NO: 259); 492D + 550K + 580K (e.g. SEQ ID NO: 258); 492D + 574H + 580K (e.g. SEQ ID NO: 262); or 492G + 550K + 574H (e.g. SEQ ID NO: 255).
  • SEQ ID NO: 239 492D + 573P + 574G + 580K (e.g. SEQ ID NO: 235); or 492F + 573P + 574G + 580K (e.g. SEQ ID NO: 238);
  • polypeptide according to any of embodiments 43 to 57 comprising alterations at positions corresponding to the following positions of SEQ ID NO: 2: 492G + 550K + 573P + 574H + 580K (e.g. SEQ ID NO: 267) or 492D + 550K + 573P + 574H + 580K (e.g. SEQ ID NO: 266).
  • 492D+550K+573P+574H e.g. SEQ ID NO: 256
  • 492G+550K+573P e.g. SEQ ID NO: 254
  • 492G+573P+580K e.g. SEQ ID NO: 242
  • 492D+573P+580K e.g. SEQ ID NO: 234
  • 573P+574H+580K (e.g. SEQ ID NO: 135); 550K+573P+580K (e.g. SEQ ID NO: 133);
  • 550K+573P+574H (e.g. SEQ ID NO: 131); or 492D+573P+574H+580K (SEQ ID NO: 114).
  • 60. The method any of embodiments 43 to 59 wherein the reference albumin is HSA (SEQ ID No: 2) or a fragment thereof, or a fusion polypeptide comprising HSA or a fragment thereof, most preferably SEQ ID NO: 2.
  • a conjugate comprising a polypeptide according to any of embodiments 1 to 42 or obtainable by a method according to any of embodiments 43 to 61 and a conjugation partner.
  • An associate comprising a polypeptide according to any of embodiments 1 to 42 or obtainable by a method according to any of embodiments 43 to 61 and a therapeutic, prophylactic, diagnostic, imaging or other beneficial moiety.
  • a composition comprising a polypeptide according to any of embodiments 1 to 42 or obtainable by a method according to any of embodiments 43 to 61 , a conjugate according to embodiment 62 or 63, an associate according to embodiment 64 or a nanoparticle or microparticle according embodiment 65, wherein the binding affinity of the polypeptide, fusion polypeptide, conjugate, associate or nanoparticle or microparticle to FcRn is stronger than the binding affinity of a composition comprising the corresponding parent albumin , reference albumin , fragment thereof or fusion polypeptide, conjugate, associate or nanoparticle or microparticle comprising said parent albumin, reference albumin or fragment or fusion thereof to FcRn.
  • a composition according to embodiment 66 or 67, wherein the binding coefficient of the variant of the polypeptide, fusion polypeptide, conjugate, associate or nanoparticle or microparticle to FcRn is less than 0.9X KD of HSA to FcRn, more preferred less than 0.5X KD of HSA to FcRn, more preferred less than 0.1 X KD of HSA to FcRn, even more preferred less than 0.05X KD of HSA to FcRn, even more preferred less than 0.02X KD of HSA to FcRn and most preferred less than 0.01X KD of HSA to FcRn.
  • composition according to any of embodiments 66 to 68 comprising a polypeptide according to any of embodiments 1 to 42 or obtainable by a method according to any of embodiments 43 to 61 , a conjugate according to embodiment 62 or 63, an associate according to embodiment 64 o r a n anoparticle or microparticle according embodiment 65, further comprising a compound comprising an antibody binding domain (ABD) and a therapeutic, prophylactic, diagnostic, imaging or other beneficial moiety.
  • ABS antibody binding domain
  • a method for altering the binding affinity to FcRn or half-life preferably in plasma, of a molecule comprising:
  • a polypeptide, fusion polypeptide, conjugate, associate, nanoparticle or microparticle or composition thereof according to any of embodiments 1 to 42 or 62 to 70 or obtainable by the method of embodiments 43 to 61 wherein the polypeptide, fusion polypeptide, conjugate, associate, nanoparticle or microparticle or composition comprises one or more (several) moiety selected from those described herein.
  • a vector comprising a nucleic acid according to embodiment 77.
  • a host cell comprising a nucleic acid according to embodiment 77 or a vector according to embodiment 78.
  • a host cell according to embodiment 79 wherein the host cell is a eukaryote, preferably a yeast (such as Saccharomyces cerevisiae) or a mammalian cell (such as CHO or HEK) or a plant cell (such as rice).
  • a yeast such as Saccharomyces cerevisiae
  • a mammalian cell such as CHO or HEK
  • a plant cell such as rice
  • a method of prophylaxis, treatment or diagnosis comprising administering a polypeptide, fusion polypeptide, conjugate, composition, associate, nanoparticle or microparticle or polynucleotide according to any of embodiments 1 to 42 or 62 to 70 or obtainable by the method of any of embodiments 43 to 61 to a subject.
  • HSA variants were expressed using standard molecular biology techniques, such as described in Sambrook, J. and D.W. Russell, 2001 (Molecular Cloning: a laboratory manual, 3 rd ed. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y).
  • Method 1 Permutation libraries at positions E492, D550, K574H and Q580K were produced by PCR amplification of pDB4081 , encoding wild type HSA (described below), using a mutagenic forward primer and non-mutagenic reverse primer as shown in Table 2. The PCR conditions are shown in Tables 3 and 4. 2 ⁇ of reaction product was assessed by agarose gel electrophoresis and the remainder treated with 5 ⁇ of 10x buffer 4 (New England Biolabs - 50 mM potassium acetate, 20mM Tris-acetate, 10mM magnesium acetate, 1 mM DTT pH 7.9 at 25 ⁇ C) and 1 ⁇ Dpn ⁇ (NEB) for 1 hour at 37° C.
  • 10x buffer 4 New England Biolabs - 50 mM potassium acetate, 20mM Tris-acetate, 10mM magnesium acetate, 1 mM DTT pH 7.9 at 25 ⁇ C
  • NEB 1 ⁇ Dpn ⁇
  • the reactions were purified by addition of 50 ⁇ water, prior to application of a vacuum for 20 minutes. A further 50 ⁇ of water was added and a vacuum applied until the well was dry. Plasmid DNA was recovered by addition of 30 ⁇ of water and incubation for 1 minute. 2 ⁇ of this purified product was transformed into 15 ⁇ TOP10 E. coli cells by standard protocols. Clones were selected and grown overnight in LB supplemented with 100 ⁇ g ml ampicillin and purified using a Qiagen QIAprep 96 miniprep kit according to manufacturer's instructions. Variants were verified by sequencing.
  • pDB4081 was made by the ligation of a synthetic DNA fragment, Bsa ⁇ /Sph ⁇ digested, which had been generated by gene assembly (DNA2.0 Inc, USA), containing 3' region of the PRB1 promoter, modified fusion leader sequence, nucleotide sequence encoding HSA and 5' region of the modified AD ⁇ terminator) into H/ndl I l/Sp/7 l-d i gested pDB4005.
  • pDB4005 is described in WO 201 1/051489 (incorporated herein by reference). Table 2: Plasmid and amino acid substitution and relevant primers
  • HSA Q580C 90 HSA-59 (194) REV Q580 (215) pDB4822
  • HSA Q580N 100 HSA-69 (204) REV Q580 (215) pDB4832
  • Method 2 Variants described in Table 5 and 6 were produced as described in WO2012/150319, Example 6, Method 2 'Production of combination Variants with K573P' (incorporated herein by reference), with the following modifications.
  • a fragment encoding the K573P mutation was removed from pDB4673 via the Sa/I and Bsu36 ⁇ restriction sites and inserted into similarly digested parent plasmid, as indicated in Table 5.
  • pDB4673 HSA K573P
  • Method 3 Variants described in Table 7 were produced as described in WO2012/150319, Example 6, Method 1 (incorporated herein by reference), using the plasmids indicated in the tables as templates for the PCR reactions. The conditions described in Tables 8 and 9 were used to produce pDB4997-5004, pDB 5042-46, and pDB5030-1 . Those described in Tables 10 and 1 1 were used to produce pDB5027-29 and pDB5032. The mutated PCR products were digested using Sa/I and Bsu36 ⁇ restriction enzymes and inserted into similarly digested pDB4081 , encoding wild type HSA (as described above).
  • Preparation of expression plasmids and transformation of S. cerevisiae was performed as described in WO2012/150319 (incorporated herein by reference), employing the 24 (pDB5027-9, pDB5032 and pDB5091 -92), or 48 hour stocking method (all remaining variants).
  • the host strain for pDB4990-5, pDB4997-5004, pDB5042-5046, pDB5027-32 and pDB5091 -2 was S. cerevisiae DYB7 (Payne et al (2008) Applied and Environmental Microbiology Vol 74(24): 7759-7766) with four copies of PDI integrated into the genome.
  • the host strain for the remaining plasmids was S. cerevisiae BXP10cir° as described in WO2012/150319 (incorporated herein by reference). Protein isolation and purification from shake flask was performed as described in WO201 1/051489.
  • Chip surface was left to stabilize with a constant flow (5 ⁇ _ ⁇ ) of running buffer - Di-basic/Mono-basic phosphate buffer pH5.5 ((67 mM phosphate buffer, 0.15 M NaCI, 0.005% Tween 20) at pH 5.5)) at 25 °C (i.e. ambient temperature) overnight.
  • the chip surface was conditioned by injecting 5-12 x 45 ⁇ _ Di-basic/Mono-basic phosphate buffer at 30 ⁇ _ ⁇ followed by HBS_EP (0.01 M H EPES, 0.15 M NaCI , 3mM EDTA, 0.005% surfactant P20) at pH 7.4 (GE Healthcare)) regeneration steps (12s) in between each injection.
  • HSA-E492G+K573P (run 2) 1 10 1 1.3 2.04 0.18
  • Variants HSA-E492D+K573P and HSA-E492G+K573P were selected for further analysis based on their apparent slow 'off rates' (i.e. dissociation constants (Kd)).
  • Kd dissociation constants
  • HSA-K574G+K573P (run 1 ) 123 1 1.5 5.5 0.48
  • HSA-K574H+K573P showed increased binding compared to WT albumin (Table 16).
  • Introduction of the K574H mutation into HSA-Q580K+K573P (Table 16) to produce HSA-K574H+Q580K+K573P resulted in no improvement in binding affinity (compared to WT albumin) over HSA-Q580K+K573P.
  • introduction of this mutation into HSA-D550K+K573P, producing HSA-D550K+K574H+K573P has resulted in an increase in affinity compared to WT albumin and HSA-D550K+K573P.
  • Table 18 show that variants containing four substitutions at positions 492 573P, 574 and 580 show a marked increase in binding affinity as compared to variants containing three mutations, such as HSA-K574H+Q580K+K573P (Table 17), variants containing two mutations, such as K574H+K573P and Q580K+K573P (Table 16) and the single K573P variant or WT albumin.
  • each of the PCR-generated plasmids was prepared for in vivo recombination by the addition of 0.5 ⁇ of each of Dpn ⁇ , Nsi ⁇ and Pvu ⁇ restriction enzymes (New England Biolabs), followed by incubation at 37°C for one hour.
  • 3 ⁇ of the prepared plasmid, 3 ⁇ of Acc65 ⁇ /Bam ⁇ - digested pDB3936 and 1 ⁇ of salmon sperm DNA were used to transform S. cerevisiae according to the protocol described in Example 1 .
  • the host strain used was a S. cerevisiae strain derived from DYB7 ura3 (Payne et a/ 2008, Appl. Environ. Microbiol.
  • Single colonies were patched onto BMMD+CSM-leu plates (as described in WO 2012/150319, incorporated herein by reference) to enable assessment of expression prior to production of yeast stocks.
  • Stocks were produced by the 48-hour method described in Example 1. 200 ⁇ of each yeast strain was used to inoculate 10 ml BMMS in 50 ml shake flask, followed by incubation at 30°C, 200 rpm for four days. Culture supernatant was harvested by centrifugation at 3000 rpm for 5 minutes.
  • Albumin variants were purified from 10ml_ shake flasks using a single chromatographic step with an albumin affinity matrix (AlbuPureTM - ProMetic Biosciences, Inc.). Micro-scale affinity chromatography was performed on an automated platform (Perkin Elmer, Janus) with 200 ⁇ _ custom packed Atoll columns with 8 run in parallel in a 96-well format using the same procedure as described in WO 201 1/051489 (incorporated herein by reference), with volumes scaled down appropriately.
  • shFcRn-GST/FLAG refers to GST- tag (glutathione-transferase) and a FLAG-tag (DYKDDDDK) on the C-terminal of the alpha chain of FcRn .
  • GST/FLAG-tagged shFcRn was purified using IgG affinity chromatography.
  • sh FcRn was captured on a GSTrap column and eluted with reduced glutathione.
  • sh FcRn was d ialyzed into PBS into pH 7.4 and further pu rified using IgG SepharoseTM 6 Fast Flow (GE Healthcare).
  • the shFcRn was captured on the resin in 50mM Na- acetate, 1 50m M N aCI pH 5.5 a nd el uted with P BS p H 7.4.
  • the el uted shFcRn was concentrated to 2-5mg/ml_ and stored at -20°C until use.
  • Immobilised level of sh FcRn- GST/FLAG was at a response level more than 1 nm, and achieved using a FcRn concentration of 2-10 ⁇ g/mL in sodium acetate, followed by ethanolamine quenching of the amine coupling reaction.
  • the sensors were either used directly or soaked in 15% (w/v) sucrose and dried until use.
  • Plasmid preparation and yeast transformations were performed as described in example 1 , using strain BXP10 cir°. 24 hour yeast stocks were produced as described in example 1 .
  • the variants listed in Table 26 and 27 were analyzed by SPR using a Biacore 3000 instrument as described in Example 2 with the exception that, prior to use, the His-tagged shFcRn was purified using IgG affinity chromatography. More specifically, the His-tagged shFcRn was captured on a Ni-HiTrap column and eluted with imidazole. The shFcRn was captured on the resin in 50mM Na-acetate, 150mM NaCI pH 5.5 and eluted with PBS pH 7.4. The eluted shFcRn was concentrated to 2-5mg/ml_ and stored at -20°C until use. using shFcRn-HIS
  • Table 26 and 27 show an improvement in affinity over WT HSA in variants containing a combination of substitutions. Generally, variants containing three or four substitutions show improved binding characteristics over those containing two substitutions. Generally, inclusion of Q580K contributes substantially to an improved binding affinity.

Abstract

The present invention relates to variants of a parent albumin, the variants having altered plasma half-life compared with the parent albumin. The present invention also relates to polynucleotides encoding the variants; nucleic acid constructs, vectors, and host cells comprising the polynucleotides; and methods of using the variants.

Description

ALBUMIN VARIANTS
Reference to a Sequence Listing
This application contains a Sequence Listing in computer readable form, which is incorporated herein by reference.
Background of the Invention
Field of the Invention
The invention relates to variants of albumin or fragments thereof or fusion polypeptides comprising variant albumin or fragments thereof having a change in binding affinity to FcRn and/or a change in half-life compared to the albumin, fragment thereof or fusion polypeptide comprising albumin or a fragment thereof. The invention allows tailoring of binding affinity and/or half-life of an albumin to the requirements and desires of a user or application. Description of the Related Art
Albumin is a protein naturally found in the blood plasma of mammals where it is the most abundant protein. It has important roles in maintaining the desired osmotic pressure of the blood and also in transport of various substances in the blood stream. Albumins have been characterized from many species including human, pig, mouse, rat, rabbit and goat and they share a high degree of sequence and structural homology.
Albumin binds in vivo to its receptor, the neonatal Fc receptor (FcRn) "BrambeH" and this interaction is known to be important for the plasma half-life of albumin. FcRn is a membrane bound protein, expressed in many cell and tissue types. FcRn has been found to salvage albumin from intracellular degradation (Roopenian D. C. and Akilesh, S. (2007), Nat. Rev. Immunol 7, 715-725.). FcRn is a bifunctional molecule that contributes to maintaining a high level of IgGs and albumin in serum in mammals such as human beings.
Whilst the FcRn-immunoglobulin (IgG) interaction has been characterized in the prior art, the FcRn-albumin interaction is less well characterized. The major FcRn binding site is localized within Dil l (381 -585), (Andersen et al (2010), Clinical Biochemistry 43,367-372). A number of key amino acids have been shown to be important in binding, notably histidines H464, H510 a n d H 53 6 and Lys500 (Andersen et al (2010), Nat. Commun. 3:610. DOI:10.1038/ncomms1607). The crystal structure of a human serum albumin (HSA) variant (V418M+T420A+E505G+V547A) with strong affinity to FcRn at acidic pH and in addition with increased binding at neutral pH has been reported allowing more detailed understanding of the interacting interfaces. In addition, the authors were able to alter the affinity to FcRn through amino acid substitution and show that this could translate into increased circulatory half-lives in mice and monkey, most notably for HSA E505G+V547A (Schmidt et al (2012), Cell Structure. 21 , Issue 1 1 , (doi:10.1016/j.str.2013.08.022)). Data indicates that IgG and albumin bind non-cooperatively to distinct sites on FcRn (Andersen et al. (2006), Eur. J. Immunol 36, 3044-3051 ; Chaudhury et al. (2006), Biochemistry 45, 4983-4990).
It is known that mouse FcRn binds IgG from mice and humans whereas human FcRn appears to be more discriminating (Ober et al. (2001 ) Int. Immunol 13, 1551 -1559). Andersen et al. (2010) Journal of Biological Chemistry 285(7):4826-36, describes the affinity of human and mouse FcRn for each mouse and human albumin (all possible combinations). No binding of albumin from either species was observed at physiological pH to either receptor. At acidic pH, a 100-fold difference in binding affinity was observed. In all cases, binding of albumin and IgG from either species to both receptors were additive.
Human serum albumin (HSA) has been well characterized as a polypeptide of 585 amino acids, the sequence of which can be found in Peters, T., Jr. (1996) All about Albumin: Biochemistry, Genetics and Medical, Applications pp10, Academic Press, Inc., Orlando (ISBN 0- 12-5521 10-3). It has a characteristic binding to its receptor FcRn, where it binds at pH 6.0 but not at pH 7.4.
The plasma half-life of HSA has been found to be approximately 19 days. A natural variant having lower plasma half-life has been identified (Peach, R. J . and Brennan, S. 0., (1991 ) Biochim Biophys Acta.1097:49-54) having the substitution D494N . This substitution generated an N-glycosylation site in this variant, which is not present in the wild-type albumin. It is not known whether the glycosylation or the amino acid change is responsible for the change in plasma half- life.
Albumin has a long plasma half-life and because of this property it has been suggested for use i n d rug del ivery. Al bu m i n has been conjugated to pharmaceutically beneficial compounds (WO2000/69902), and it was found that the conjugate maintained the long plasma half-life of albumin. The resulting plasma half-life of the conjugate was generally considerably longer than the plasma half-life of the beneficial therapeutic compound alone.
Further, albumin has been genetically fused to therapeutically beneficial peptides (WO 2001/79271 A and WO2003/59934) with the typical result that the fusion has the activity of the therapeutically beneficial peptide and a considerably longer plasma half-life than the plasma half-life of the therapeutically beneficial peptides alone.
Otagiri et al (2009), Biol. Pharm. Bull. 32(4), 527-534, discloses more than 70 albumin variants, of these 25 of these are found to be mutated in domain III. A natural variant lacking the last 175 amino acids at the carboxy termini has been shown to have reduced half-life (Andersen et al (2010), Clinical Biochemistry 43, 367-372). Iwao et al (2007) studied the half-life of naturally occurring human albumin variants using a mouse model, and found that K541 E and K560E had reduced half-life, E501 K and E570K had increased half-life and K573E had almost no effect on half-life (Iwao, et. al. (2007) B.B.A. Proteins and Proteomics 1774, 1582-1590).
Galliano et al (1993) Biochim. Biophys. Acta 1225, 27-32 discloses a natural variant E505K. Minchiotti ei a/ (1990) discloses a natural variant K536E. Minchiotti ei a/ (1987) Biochim. Biophys. Acta 916, 41 1 -418, discloses a natural variant K574N. Takahashi et al (1987) Proc. Natl. Acad. Sci. USA 84, 4413-4417, discloses a natural variant D550G. Carlson et al (1992). Proc. Nat. Acad. Sci. USA 89, 8225- 8229, discloses a natural variant D550A.
WO201 1/051489 and WO2012/150319 disclose a number of point mutations in albumin which modulate the binding of albumin to FcRn. WO2010/092135 discloses a number of point mutations in albumin which increase the number of thiols available for conjugation in the albumin, the disclosure is silent about the affect of the mutations on the binding of the albumin to FcRn. WO201 1/103076 discloses albumin variants, each containing a substitution in Domain III of HSA. WO2012/1 12188 discloses albumin variants containing substitutions in Domain III of HSA.
Albumin has the ability to bind a number of ligands and these become associated (associates) with albumin. This property has been utilized to extend the plasma half-life of drugs having the ability to non-covalently bind to albumin. This can also be achieved by binding a pharmaceutical beneficial compound, which has little or no albumin binding properties, to a moiety having albumin binding properties, see review article and reference therein, Kratz (2008) Journal of Controlled Release 132, 171 -183.
Albumin is used in preparations of pharmaceutically beneficial compounds, in which such a preparation may be for example, but not limited to, a nanoparticle or microparticle of albumin. In these examples the delivery of a pharmaceutically beneficial compound or mixture of compounds may benefit from alteration in the albumin's affinity to its receptor where the beneficial compound has been shown to associate with albumin for the means of delivery. It is not clear what determines the plasma half-life of the formed associates (for example but not limited to Levemir®, Kurtzhals P et al. Biochem. J. 1995; 312:725-731 ), conjugates or fusion polypeptides but it appears to be a result of the combination of the albumin and the selected pharmaceutically beneficial compound/polypeptide. It would be desirable to be able to control the plasma half-life of given albumin conjugates, associates or albumin fusion polypeptides so that a longer or shorter plasma half-life can be achieved than given by the components of the association, conjugation or fusion, in order to be able to design a particular drug according to the particulars of the indication intended to be treated.
Albumin is known to accumulate and be catabolised in tumours, it has also been shown to accumulate in inflamed joints of rheumatoid arthritis sufferers. See review article and reference therein, Kratz (2008) Journal of Controlled Release 132, 171 -183. It is envisaged that HSA variants with increased affinity for FcRn would be advantageous for the delivery of pharmaceutically beneficial compounds.
It may even be desirable to have variants of albumin that have little or no binding to FcRn in order to provide shorter half-lives or controlled serum pharmacokinetics as described by Kenanova et al (2009) J. Nucl. Med.; 50 (Supplement 2): 1582). Kenanova et al (2010, Protein Engineering, Design & Selection 23(10): 789-798; WO2010/1 18169) discloses a docking model comprising a structural model of domain III of HSA (solved at pH 7 to 8) and a structural model of FcRn (solved at pH 6.4). Kenanova et al discloses that positions 464, 505, 510, 531 and 535 in domain III potentially interact with FcRn. The histidines at positions 464, 510 and 535 were identified as being of particular interest by Chaudhury et al., (2006, op. cit.) and these were shown to have a significant reduction in affinity and shorter half-life in mouse by Kenanova (2010, op. cit). However, the studies of Kenanova et al are limited to domain I I I of HSA and therefore do not consider HSA in its native intact configuration. Furthermore, the identified positions result in a decrease in affinity for the FcRn receptor.
The present invention provides further variants having altered binding affinity to the FcRn receptor. The albumin moiety or moieties may therefore be used to tailor the binding affinity to FcRn and/or half-life of fusion polypeptides, conjugates, associates, nanoparticles and compositions comprising the albumin moiety.
Summary of the Invention
The present invention relates to albumin variants comprising an alteration at positions corresponding to positions selected among two or more of the group consisting of positions 492, 550, 573, 574 and 580 of the mature polypeptide of SEQ ID NO: 2 or equivalent positions of other albumins or fragments thereof. Position 492 is located in the connector loop between subdomain Dllla and subdomain Dlllb. Positions 550 , 573 , 574 and 580 are located in subdomain Dlllb. Subdomain 1Mb is located proximal to the connector loop between subdomain Dllla and subdomain Dlllb.
The present invention also relates to isolated polynucleotides encoding the variants; nucleic acid constructs, vectors, and host cells comprising the polynucleotides; and methods of producing the variants.
The invention also relates to conjugates or associates comprising the variant albumin or fragment thereof according to the invention and a beneficial therapeutic moiety or to a fusion polypeptide comprising a variant albumin or fragment thereof of the invention and a fusion partner polypeptide.
The invention further relates to compositions comprising the variant albumin, fragment thereof, fusion polypeptide comprising variant albumin or fragment thereof or conjugates comprising the variant albumin or fragment thereof, according to the invention or associates comprising the variant albumi n or fragment thereof, accordi ng to the invention . The compositions are preferably pharmaceutical compositions.
The invention further relates to a pharmaceutical composition comprising a variant albumin, fragment thereof, fusion polypeptide comprising variant albumin or fragment thereof or conjugates comprising the variant albumin or fragment thereof, or associates comprising the variant albumin or fragment thereof.
The invention also relates to the use of the variants, fragments, fusion polypeptides, conjugates, associates, nanoparticles and microparticles.
The invention also relates to a method for preparing a variant albumin, fragment thereof, fusion polypeptide comprising variant albumin or fragment thereof or conjugates comprising the variant albumin or fragment thereof, or associates comprising the variant albumin or fragment thereof.
Brief Description of the Figures
Figure 1 : Multiple alignment of amino acid sequences of (i) full length mature HSA (Hu_1_2_3), (ii) an albumin variant comprising domain I and domain III of HSA (Hu_1_3), (iii) an albumin variant comprising domain II and domain I II of HSA (Hu_2_3), (iv) full-length Macaca mulatta albumin (Mac_mul), (v) full-length Rattus norvegicus albumin (Rat) and (vi) full-length Mus musculus albumin (Mouse). Positions 500, 550 and 573 (relative to full length HSA) are indicated by arrows. In Figure 1 , Domains I, II and III are referred to as 1 , 2 and 3 (respectively).
Figure 2: Multiple alignment of amino acid sequences of mature albumin from human, sheep, mouse, rabbit and goat and immature albumins from chimpanzee ("Chimp"), macaque, hamster, guinea pig, rat, cow, horse, donkey, dog, chicken, and pig. The Start and End amino acids of domains 1 , 2 and 3 (as defined by Dockal et al (The Journal of Biological Chemistry, 1999, Vol. 274(41 ): 29303-29310)) are indicated with respect to mature human albumin.
Figure 3: Conserved groups of amino acids based on their properties.
Figure 4: Representation of shFcRn-HSA docking model. (A-B) Two orientations of the complex are shown. Albumin is shown by a space-filling diagram, FcRn is shown as a ribbon diagram. The core binding interface of HSA is highlighted in pink (in grey-scale this is seen as the darkest (almost black) region; Dl (CBI)), while the area distally localized from the interface is shown as DM (orange) and DI N is split into sub-domains Dl I la (in colour, this is cyan) and Dlllb (in colour, this is blue).
Definitions
Variant: The term "variant" means a polypeptide derived from a parent albumin by one or more (several) alteration(s), i.e., a substitution, insertion, and/or deletion, at one or more (several) positions. A substitution means a replacement of an amino acid occupying a position with a different amino acid; a deletion means removal of an amino acid occupying a position; and an insertion means adding 1 or more, such as 1 , 2, 3, 4, 5, 6, 7, 8, 9 or 10, preferably 1 -3 amino acids immediately adjacent an amino acid occupying a position. In relation to substitutions, 'immediately adjacent' may be to the N-side ('upstream') or C-side ('downstream') of the amino acid occupying a position ('the named amino acid'). Therefore, for an amino acid named/numbered 'X', the insertion may be at position 'X+1 ' ('downstream') or at position 'X-1 ' ('upstream').
Mutant: The term "mutant" means a polynucleotide encoding a variant.
Wild-Type Albumin: The term "wild-type" (WT) albumin means albumin having the same amino acid sequence as naturally found in an animal or in a human being.
Parent Albumin: The term "parent" or "parent albumin" means an albumin to which an alteration is made by the hand of man to produce the albumin variants of the invention. The parent may be a naturally occurring (wild-type) polypeptide or an allele thereof, or even a variant thereof.
Albumin: Albumins are proteins and constitute the most abundant protein in plasma in mammals and albumins from a long n umber of mammals have been characterized by biochemical methods and/or by sequence information. Several albumins, e.g., human serum albumin (HSA), have also been characterized crystallographically and the structure determined (HSA: He XM, Carter DC (July 1 992). "Atomic structure and chemistry of human serum albumin". Nature 358 (6383): 209-15; horse albumin: Ho, J.X. et al. (2001 ). X-ray and primary structure of horse serum albumin (Equus caballus) at 0.27-nm resolution. Eur J Biochem. 215(1 ):205-12).
The term "albumin" means a protein having the same and/or very similar th ree dimensional (tertiary) structure as HSA or HSA domains and has similar properties to HSA or to the relevant domains. Similar three dimensional structures are for example the structures of the albumins from the species mentioned herein. Some of the major properties of albumin are i) its ability to regulate plasma volume (oncotic activity), ii) a long plasma half-life of around 19 days ± 5 days, iii) binding to FcRn, iv) ligand-binding, e.g. binding of endogenous molecules such as acidic, lipophilic compounds including bilirubin, fatty acids, hemin and thyroxine (see also table
1 of Kragh-Hansen et al, 2002, Biol. Pharm. Bull. 25, 695, hereby incorporated by reference), v) binding of small organic compounds with acidic or electronegative features e.g. drugs such as warfarin, diazepam, ibuprofen and paclitaxel (see also table 1 of Kragh-Hansen et al, 2002, Biol. Pharm. Bull. 25, 695, hereby incorporated by reference). Not all of these properties need to be fulfilled in order to characterize a protein or fragment as an albumin. If a fragment, for example, does not comprise a domain responsible for binding of certain ligands or organic compounds the variant of such a fragment will not be expected to have these properties either.
Albumins have generally a long plasma half-life of approximately 20 days or longer, e.g., HSA has a plasma half-life of 19 days. It is known that the long plasma half-life of HSA is mediated via interaction with its receptor FcRn, however, an understanding or knowledge of the exact mechanism behind the long half-life of HSA is not essential for the invention.
As examples of albumin proteins , more specifically albumin proteins which may be used as parent 'backbones' as a starting point for making the albumin variants according to the invention, can be mentioned human serum albumin (e.g. AAA98797 or P02768-1 , SEQ I D NO:
2 (mature), SEQ I D NO: 4 (immature)), primate serum albumin, (such as chimpanzee serum albumin (e.g. predicted sequence XP_517233.2 SEQ I D NO: 5), gorilla serum albumin or macaque serum albumin (e.g. NP_001 182578, SEQ ID NO: 6), rodent serum albumin (such as hamster serum albumin (e.g. A6YF56, SEQ I D NO : 7), gu inea pig serum albumi n (e.g. Q6WDN9-1 , SEQ I D NO: 8), mouse serum albumin (e.g. AAH49971 or P07724-1 Version 3, SEQ I D NO: 9) and rat serum albumin (e.g. AAH85359 or P02770-1 Version 2, SEQ I D NO: 10))), bovine serum albumin (e.g. cow serum albumin P02769-1 , SEQ I D NO: 1 1 ), equine serum albumin such as horse serum albumin (e.g. P35747-1 , SEQ ID NO: 12) or donkey serum albumin (e.g. Q5XLE4-1 , SEQ ID NO: 13), rabbit serum albumin (e.g. P49065-1 Version 2, SEQ ID NO: 14), goat serum albumin (e.g. ACF10391 , SEQ ID NO: 15), sheep serum albumin (e.g. P14639-1 , SEQ I D NO: 1 6), dog serum albumin (e.g. P49822-1 , SEQ I D NO: 1 7), chicken serum albumin (e.g. P19121 -1 Version 2, SEQ ID NO: 18) and pig serum albumin (e.g. P08835- 1 Version 2, SEQ ID NO: 19) or a polypeptide having at least 70, 75, 80, 85, 90, 91 , 92, 93, 94, 95, 96, 97, 98 or at least 99% amino acid identity to such an albumin. The parent or reference albumin may be an artificial variant such as HSA. K573P (SEQ ID NO: 3) or a chimeric albumin such as the N-terminal of HSA and the C-terminal of macaca albumin (SEQ I D NO: 20), N- terminal of HSA and the C-terminal of mouse albumin (SEQ ID NO: 21 ), N-terminal of HSA and the C-terminal of rabbit albumin (SEQ I D NO: 22), N-terminal of HSA and the C-terminal of sheep albumin (SEQ ID NO: 23).
Other examples of albumin, which are also included in the scope of this application, include ovalbumin (e.g. P01012.pro: chicken ovalbumin; 073860. pro: turkey ovalbumin).
HSA as disclosed in SEQ I D NO: 2 or any naturally occurring allele thereof, is the preferred parent albumin according to the invention. HSA is a protein consisting of 585 amino acid residues and has a molecular weight of 67 kDa. In its natural form it is not glycosylated. The skilled person will appreciate that natural alleles may exist having essentially the same properties as HSA but having one or more (several) amino acid changes compared to SEQ ID NO: 2, and the inventors also contemplate the use of such natural alleles as parent albumin according to the invention.
The parent albumin , a fragment thereof, or albumin part of a fusion polypeptide comprising albumin or a fragment thereof according to the invention preferably has a sequence identity to the sequence of HSA shown in SEQ I D NO: 2 of at least 60%, preferably at least 70%, preferably at least 80%, preferably at least 85%, preferably at least 86%, preferably at least 87%, preferably at least 88%, preferably at least 89%, preferably at least 90%, preferably at least 91 %, preferably at least 92%, preferably at least 93%, preferably at least 94%, preferably at least 95%, more preferred at least 96%, more preferred at least 97%, more preferred at least 98% and most preferred at least 99%. It is preferred that the parent albumin maintains at least one of the major properties of albumin or a similar tertiary structure as an albumin, such as HSA. The sequence identity may be over the full-length of SEQ I D NO: 2 or over a molecule consisting or comprising of a fragment such as one or more (several) domains of SEQ ID NO: 2 such as a molecule consisting of or comprising domain I II (e.g. SEQ ID NO: 27), a molecule consisting of or comprising domain II and domain I II (e.g. SEQ I D NO: 25), a molecule consisting of or comprising domain I and domain III (e.g. SEQ ID NO: 24), a molecule consisting of or comprising two copies of domain III (e.g. SEQ ID NO: 26), a molecule consisting of or comprising three copies of domain III (e.g. SEQ I D NO: 28) or a molecule consisting of or comprising domain I and two copies of domain III (e.g. SEQ ID NO: 29).
The parent preferably comprises or consists of the amino acid sequence of SEQ I D NO: 4 (immature sequence of HSA) or SEQ ID NO: 2 (mature sequence of HSA).
In another embodiment, the parent is an allelic variant of the mature polypeptide of SEQ ID NO: 2.
The parent albumin may be encoded by a polynucleotide that hybridizes under very low stringency conditions, low stringency conditions, medium stringency conditions, medium-high stringency conditions, high stringency conditions, or very high stringency conditions with (i) the mature polypeptide coding sequence of SEQ I D NO: 1 or (ii) the full-length complementary strand of (i) (J. Sambrook, E.F. Fritsch, and T. Maniatis, 1989, Molecular Cloning, A Laboratory Manual, 2d edition, Cold Spring Harbor, New York).
The polynucleotide of SEQ I D NO: 1 or a subsequence thereof, as well as the amino acid sequence of SEQ I D NO: 2 or a fragment thereof, may be used to design nucleic acid probes to identify and clone DNA encoding a parent from strains of different genera or species according to methods well known in the art. I n particular, such probes can be used for hybridization with the genomic or cDNA of the genus or species of interest, following standard Southern blotting procedures, in order to identify and isolate the corresponding gene therein. Such probes can be considerably shorter than the entire sequence, but should be at least 14, e.g. , at least 25, at least 35, or at least 70 nucleotides in length. Preferably, the nucleic acid probe is at least 1 00 nucleotides in length , e.g., at least 200 nucleotides, at least 300 nucleotides, at least 400 nucleotides, at least 500 nucleotides, at least 600 nucleotides, at least 700 nucleotides, at least 800 nucleotides, or at least 900 nucleotides in length. Both DNA and RNA probes can be used. The probes are typically labelled for detecting the corresponding gene (for example, with 32P, 3H, 35S, biotin, or avidin). Such probes are encompassed by the invention.
A genomic DNA or cDNA library prepared from such other organisms may be screened for DNA that hybridizes with the probes described above and encodes a parent. Genomic or other DNA from such other organisms may be separated by agarose or polyacrylamide gel electrophoresis, or other separation techniques. DNA from the libraries or the separated DNA may be transferred to and immobilized on nitrocellulose or other suitable carrier material. In order to identify a clone or DNA that is homologous with SEQ I D NO: 1 or a subsequence thereof, the carrier material is used in a Southern blot.
For purposes of the invention, hybridization indicates that the polynucleotide hybridizes to a labelled nucleotide probe corresponding to the polynucleotide shown in SEQ I D NO: 1 , its complementary strand, or a subsequence thereof, under low to very high stringency conditions. Molecules to which the probe hybridizes can be detected using, for example, X-ray film or any other detection means known in the art.
The nucleic acid probe may comprise or consist of the mature polypeptide coding sequence of SEQ ID NO: 1 , i.e. nucleotides 1 to 1785 of SEQ ID NO: 1 .The nucleic acid probe may comprise or consist of a polynucleotide that encodes the polypeptide of SEQ ID NO: 2 or a fragment thereof.
For long probes of at least 100 nucleotides in length, very low to very high stringency conditions are defined as pre-hybridization and hybridization at 42°C in 5X SSPE, 0.3% SDS, 200 micrograms/ml sheared and denatured salmon sperm DNA, and either 25% formamide for very low and low stringencies, 35% formamide for medium and medium-high stringencies, or 50% formamide for high and very high stringencies, following standard Southern blotting procedures for 12 to 24 hours optimally. The carrier material is finally washed three times each for 15 minutes using 2X SSC, 0.2% SDS at 45°C (very low stringency), 50°C (low stringency), 55°C (medium stringency), 60°C (medium-high stringency), 65°C (high stringency), or 70°C (very high stringency).
For short probes that are about 1 5 nucleotides to about 70 nucleotides in length , stringency conditions are defined as pre-hybridization and hybridization at about 5°C to about 10°C below the calculated Tm using the calculation according to Bolton and McCarthy (1962, Proc. Natl. Acad. Sci. USA 48: 1390) in 0.9 M NaCI, 0.09 M Tris-HCI pH 7.6, 6 mM EDTA, 0.5% NP-40, 1 X Denhardt's solution , 1 m M sodium pyrophosphate, 1 mM sodium monobasic phosphate, 0.1 mM ATP, and 0.2 mg of yeast RNA per ml following standard Southern blotting procedures for 12 to 24 hours optimally. The carrier material is finally washed once in 6X SCC plus 0.1 % SDS for 15 minutes and twice each for 15 minutes using 6X SSC at 5°C to 10°C below the calculated Tm.
The parent may be encoded by a polynucleotide with a sequence identity to the mature polypeptide coding sequence of SEQ ID NO: 1 of at least 60%, e.g., at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%, which encodes a polypeptide which is able to function as an albumin . I n an embodiment, the parent is encoded by a polynucleotide comprising or consisting of SEQ ID NO: 1 .
Albumin moiety: The albumin part of a fusion polypeptide, conjugate, associate, nanoparticle or composition comprising the albumin variant or fragment thereof according to the invention, may be referred to as an 'albumin moiety' or 'albumin component'. A polypeptide according to the invention may comprise or consist of an albumin moiety.
FcRn and shFcRn: The term "FcRn" means the human neonatal Fc receptor (FcRn). shFcRn is a soluble recombinant form of FcRn. hFcRn is a heterodimer of SEQ I D NO: 30 (truncated heavy chain of the major histocompatibility complex class l-like Fc receptor (FCGRT)) and SEQ ID NO: 31 (beta-2-microglobulin). Together, SEQ I D NO: 30 and 31 form hFcRn.
Isolated variant: The term "isolated variant" means a variant in a form or environment which does not occur in nature. Non-limiting examples of isolated variants include (1 ) any non- naturally occurring variant, (2) any variant that is at least partially removed from one or more (several) or all of the naturally occurring constituents with which it is associated in nature; (3) any variant modified by the hand of man relative to the polypeptide from which it is derived (e.g. the polypeptide from which it is derived as found in nature); or (4) any variant modified by increasing the amount of the variant e relative to other components with which it is naturally associated (e.g., multiple copies of a gene encoding the substance; use of a stronger promoter than the promoter naturally associated with the gene encoding the substance). An isolated variant may be present in a fermentation broth sample.
Substantially pure variant: The term "substantially pure variant" means a preparation that contains at most 10%, at most 8%, at most 6%, at most 5%, at most 4%, at most 3%, at most 2%, at most 1 %, and at most 0.5% by weight of other polypeptide material with which it is natively or recombinantly associated. Preferably, the variant is at least 92% pure, e.g., at least 94% pure, at least 95% pure, at least 96% pure, at least 97% pure, at least 98% pure, at least 99%, at least 99.5% pure, and 100% pure by weight of the total polypeptide material present in the preparation. Purity may be determined by SDS-PAGE or GP-HPLC. The variants of the invention are preferably in a substantially pure form. This can be accomplished, for example, by preparing the variant by well-known recombinant methods and by purification methods.
Mature polypeptide: The term "mature polypeptide" means a polypeptide in its final form followi ng translation and any post-translational modifications , such as N-terminal processing, C-terminal truncation, glycosylation, phosphorylation, etc. The mature polypeptide may be amino acids 1 to 585 of SEQ ID NO: 2, e.g. with the inclusion of alterations according to the invention and/or any post-translational modifications.
Mature polypeptide coding sequence: The term "mature polypeptide cod ing sequence" means a polynucleotide that encodes a mature albumin polypeptide. The mature polypeptide coding sequence may be nucleotides 1 to 1 758 of SEQ I D NO: 1 e.g. with the alterations required to encode a variant according to the invention.
Sequence Identity:
The relatedness between two amino acid sequences or between two nucleotide sequences is described by the parameter "sequence identity".
For purposes of the present invention, the sequence identity between two amino acid sequences is determined using the Needleman-Wunsch algorithm (Needleman and Wunsch, 1970, J. Mol. Biol. 48: 443-453) as implemented in the Needle program of the EMBOSS package (EMBOSS: The European Molecular Biology Open Software Suite, Rice et al., 2000, Trends Genet. 16: 276-277), preferably version 5.0.0 or later. The parameters used are gap open penalty of 10, gap extension penalty of 0.5, and the EBLOSUM62 (EMBOSS version of BLOSUM62) substitution matrix. The output of Needle labeled "longest identity" (obtained using the -nobrief option) is used as the percent identity and is calculated as follows:
(Identical Residues x 100)/(Length of Alignment - Total Number of Gaps in Alignment)
For pu rposes of th e present i nvention , th e seq u en ce id entity between two deoxyribonucleotide sequences is determined using the Need leman-Wunsch algorithm (Needleman and Wunsch, 1970, supra) as implemented in the Needle program of the EMBOSS package (EMBOSS: The European Molecular Biology Open Software Suite, Rice et al., 2000, supra), preferably version 5.0.0 or later. The parameters used are gap open penalty of 10, gap extension penalty of 0.5, and the EDNAFULL (EMBOSS version of NCBI NUC4.4) substitution matrix. The output of Needle labeled "longest identity" (obtained using the -nobrief option) is used as the percent identity and is calculated as follows:
(Identical Deoxyribonucleotides x 100)/(Length of Alignment - Total N um ber of Gaps i n Alignment)
Fragment: The term "fragment" means a polypeptide having one or more (several) amino acids deleted from the amino and/or carboxyl terminus of an albumin and/or an internal region of albumin that has retained the ability to bind to FcRn. Fragments may consist of one uninterrupted sequence derived from HSA or it may comprise two or more sequences derived from HSA. The fragments according to the invention have a size of more than approximately 20 amino acid residues, preferably more than 30 amino acid residues, more preferred more than 40 amino acid residues, more preferred more than 50 amino acid residues, more preferred more than 75 amino acid residues, more preferred more than 1 00 amino acid residues, more preferred more than 200 amino acid residues, more preferred more than 300 amino acid residues, even more preferred more than 400 amino acid residues and most preferred more than 500 amino acid residues. A fragment may comprise or consist of one more domains of albumin such as Dl + DM , Dl + Di l l , DM + DI N , Di l l + Di l l , Dl + Di l l + Di l l, Di l l + Di l l +DI II , or fragments of such domains or combinations of domains.
Domains I , I I and I I I may be defined with reference to HSA (SEQ I D NO: 2). For example, HSA domain I may consist of or comprise amino acids 1 to 194 (± 1 to 15 amino acids) of SEQ I D NO: 2, HSA domain I I may consist of or comprise amino acids 192 (± 1 to 15 amino acids) to 387 (± 1 to 15 amino acids) of SEQ ID NO: 2 and domain I II may consist of or comprise amino acid residues 381 (± 1 to 15 amino acids) to 585 (± 1 to 15 amino acids) of SEQ ID NO: 2. "± 1 to 15 amino acids" means that the residue number may deviate by 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, or 15 amino acids to the C-terminus and/or to the N-terminus of the stated amino acid position. Examples of domains I, II and III are described by Dockal et al (The Journal of Biological Chemistry, 1999, Vol. 274(41 ): 29303-29310) and Kjeldsen et al (Protein Expression and Purification, 1998, Vol 13: 163-169) and are tabulated below.
Figure imgf000013_0001
The skilled person can identify domains I , I I and I I I in non-human albumins by amino acid sequence alignment with HSA, for example using the Needleman-Wunsch algorithm (Needleman and Wunsch, 1970, J. Mol. Biol. 48: 443-453) as implemented in the Needle program of the EMBOSS package (EMBOSS: The European Molecular Biology Open Software Suite, Rice et al., 2000, Trends Genet. 16: 276-277), preferably version 3.0.0 or later. The optional parameters used are gap open penalty of 10, gap extension penalty of 0.5, and the EBLOSUM62 (EMBOSS version of BLOSU M62) substitution matrix. Other suitable software includes M USCLE ((Multiple sequence comparison by log-expectation, Robert C. Edgar, Version 3.6, http://www.drive5.com/muscle; Edgar (2004) Nucleic Acids Research 32(5), 1792- 97 and Edgar (2004) BMC Bioinformatics, 5(1 ):1 13) which may be used with the default settings as described in the User Guide (Version 3.6, September 2005). Versions of MUSCLE later than 3.6 may also be used for any aspect of the invention). Examples of suitable alignments are provided in Figures 1 and 2.
It is preferred that domains have at least 70, 75, 80, 85, 90, 95, 96, 97, 98, 99, 99.5 % identity or 100% identity to Domain I, II or III of HSA (SEQ ID NO: 2).
Allelic variant: The term "allelic variant" means any of two or more alternative forms of a gene occupying the same chromosomal locus. Allelic variation arises naturally through mutation, and may result in polymorphism within populations. Gene mutations can be silent (no change in the encoded polypeptide) or may encode polypeptides having altered amino acid sequences. An allelic variant of a polypeptide is a polypeptide encoded by an allelic variant of a gene.
Coding sequence: The term "coding sequence" means a polynucleotide, which directly specifies the amino acid sequence of its translated polypeptide product. The boundaries of the coding sequence are generally determined by an open reading frame, which usually begins with the ATG start codon or alternative start codons such as GTG and TTG and ends with a stop codon such as TAA, TAG, and TGA. The coding sequence may be a DNA, cDNA, synthetic, or recombinant polynucleotide.
cDNA: The term "cDNA" means a DNA molecule that can be prepared by reverse transcription from a mature, spliced, mRNA molecule obtained from a eukaryotic cell. cDNA lacks intron sequences that may be present in the corresponding genomic DNA. The initial, primary RNA transcript is a precursor to mRNA that is processed through a series of steps, including splicing, before appearing as mature spliced mRNA.
Nucleic acid construct: The term "nucleic acid construct" means a nucleic acid molecule, either single- or double-stranded, which is isolated from a naturally occurring gene or is modified to contain segments of nucleic acids in a manner that would not otherwise exist in nature or which is synthetic. The term nucleic acid construct is synonymous with the term "expression cassette" when the nucleic acid construct contains the control sequences required for expression of a coding sequence of the invention.
Control sequences: The term "control sequences" means all nucleic acid sequences necessary for the expression of a polynucleotide encoding a variant of the invention. Each control sequence may be native (i.e. from the same gene) or foreign (i.e. from a different gene) to the polynucleotide encoding the variant or native or foreign to each other. Such control sequences include, but are not limited to, a leader, polyadenylation sequence, propeptide sequence, promoter, signal peptide sequence, and transcription terminator. At a minimum, the control sequences include a promoter, and transcriptional and translational stop signals. The control sequences may be provided with linkers for the purpose of introducing specific restriction sites facilitating ligation of the control sequences with the coding region of the polynucleotide encoding a variant.
Operably linked: The term "operably linked" means a configuration in which a control seq uence is placed at an appropriate position relative to the coding sequence of a polynucleotide such that the control sequence directs the expression of the coding sequence.
Expression: The term "expression" includes any step involved in the production of the variant including, but not limited to, transcription, post-transcriptional modification, translation, post-translational modification, and secretion.
Expression vector: The term "expression vector" means a linear or circular DNA molecule that comprises a polynucleotide encoding a variant and is operably linked to control sequences that provide for its expression.
Host cell: The term "host cell" means any cell type that is susceptible to transformation, transfection, transduction, or the like with a nucleic acid construct or expression vector comprising a polynucleotide of the present invention. The term "host cell" encompasses any progeny of a parent cell that is not identical to the parent cell due to mutations that occur during replication.
Plasma half-life: Plasma half-life is ideally determined in vivo in suitable individuals.
However, since it is time consuming and expensive and there inevitable are ethical concerns connected with doing experiments in animals or man it is desirable to use an in vitro assay for determining whether plasma half-life is extended or reduced. It is known that the binding of albumin to its receptor FcRn is important for plasma half-life and the correlation between receptor binding and plasma half-life is that a higher affinity of albumin to its receptor leads to longer plasma half-life. Thus for the invention a higher affinity of albumin to FcRn is considered indicative of an increased plasma half-life and a lower affinity of albumin to its receptor is considered indicative of a reduced plasma half-life.
In this application and claims the binding of albumin to its receptor FcRn is described using the term affinity and the expressions "stronger" or "weaker". Thus, it should be understood that a molecule having a higher affinity to FcRn than HSA is considered to bind stronger to FcRn than HSA and a molecule having a lower affinity to FcRn than HSA is considered to bind weaker to FcRn than HSA.
The terms "longer plasma half-life" or "shorter plasma half-life" and similar expressions are understood to be in relationship to the corresponding parent or reference or corresponding albumin molecule. Thus, a longer plasma half-life with respect to a variant albumin of the invention means that the variant has longer plasma half-life than the corresponding albumin having the same sequences except for the alteration(s) described herein, e.g. at two or more positions corresponding to 492, 550, 573, 574 and 580 of HSA (SEQ ID NO: 2).
Reference: a reference is an albumin, fusion, conjugate, composition, associate or nanoparticle to which an albumin variant, fusion , conjugate, composition , associate or nanoparticle is compared. The reference may comprise or consist of full length albumin (such as HSA or a natural allele thereof) or a fragment thereof. A reference may also be referred to as a 'corresponding' albumin, fusion, conjugate, composition, associate or nanoparticle to which an albumin variant, fusion, conjugate, composition, associate or nanoparticle is compared. A reference may comprise or consist of HSA (SEQ I D NO: 2) or a fragment, fusion, conjugate, associate, nanoparticle or microparticle thereof. Preferably, the reference is identical to the polypeptide, fusion polypeptide, conjugate, composition, associate, nanoparticle or microparticle according to the invention ("being studied") with the exception of the albumin moiety. Preferably the albumin moiety of the reference comprises or consists of an albumin (e.g. HSA, SEQ ID NO: 2) or a fragment thereof. The amino acid sequence of the albumin moiety of the reference may be longer than, shorter than or, preferably, the same (± 1 to 15 amino acids) length as the amino sequence of the albumin moiety of the polypeptide, fusion polypeptide, conjugate, composition , associate, nanoparticle or microparticle according to the invention ("being studied").
Equivalent amino acid positions: Throughout this specification amino acid positions are defined in relation to full-length mature human serum albumin (i.e. without leader sequence, SEQ I D NO: 2). However, the skilled person understands that the invention also relates to variants of non-human albumins (e.g. those disclosed herein) and/or fragments of a human or non-human albumin. Equivalent positions can be identified in fragments of human serum albumin, in animal albumins and in fragments, fusions and other derivative or variants thereof by comparing amino acid sequences using pairwise (e.g. ClustalW) or multiple (e.g. MUSCLE) alignments. For example, Fig. 1 shows that positions equivalent to 500, 550 and 573 in full length human serum albumin are easily identified in fragments of human serum albumin and in albumins of other species. Positions 500, 550 and 573 are indicated by arrows. Further details are provided in Table 1 below.
Table 1 : Example of identification of equivalent positions in HSA, animal albumins and albumin fragments
Figure imgf000016_0001
Fig. 1 was generated by MUSCLE using the default parameters including output in C l u sta lW 1 .81 fo rm at . Th e raw ou tp u t d ata was s h ad ed u s i n g B oxS h ad e 3.2 1 (http://www.ch.embnet.org/software/BOX form.html) using Output Format: RTF_new; Font Size: 10; Consensus Line: no consensus line; Fraction of sequences (that must agree for shading): 0.5; Input sequence format: ALN. Therefore, throughout this specification amino acid positions defined in human serum albumin also apply to equivalent positions in fragments, derivatives or variants and fusions of human serum albumin, animals from other species and fragments and fusions thereof. Such equivalent positions may have (i) a different residue number in its native protein and/or (ii) a different native amino acid in its native protein.
Likewise, Fig. 2 shows that equivalent positions can be identified in fragments (e.g. domains) of an albumin with reference to SEQ ID NO: 2 (HSA).
Conventions for Designation of Variants
For purposes of the present invention, the mature polypeptide disclosed in SEQ ID NO:
2 is used to determine the corresponding amino acid residue in another albumin. The amino acid sequence of another albumin is aligned with the mature polypeptide disclosed in SEQ I D NO: 2, and based on the alignment, the amino acid position number corresponding to any amino acid residue in the mature polypeptide disclosed in SEQ I D NO: 2 is determined using the Needleman-Wunsch algorithm (Needleman and Wunsch, 1970, J. Mol. Biol. 48: 443-453) as implemented in the Needle program of the EMBOSS package (EMBOSS: The European Molecular Biology Open Software Suite, Rice et al., 2000, Trends Genet. 16: 276-277), preferably version 5.0.0 or later. The parameters used are gap open penalty of 1 0, gap extension penalty of 0.5, and the EBLOSUM62 (EMBOSS version of BLOSUM62) substitution matrix.
Identification of the corresponding amino acid residue in another albumin can be determined or confirmed by an alignment of multiple polypeptide sequences using several computer programs including, but not limited to, MUSCLE (multiple sequence comparison by log-expectation; version 3.5 or later; Edgar, 2004, Nucleic Acids Research 32: 1792-1797), MAFFT (version 6.857 or later; Katoh and Kuma, 2002, Nucleic Acids Research 30: 3059-3066; Katoh et al., 2005, Nucleic Acids Research 33: 51 1 -518; Katoh and Toh, 2007, Bioinformatics 23: 372-374; Katoh et al., 2009, Methods in Molecular Biology 537:_39-64; Katoh and Toh, 2010, Bioinformatics 26:_1899-1900), and EMBOSS EMMA employing ClustalW (1 .83 or later; Thompson et al., 1994, Nucleic Acids Research 22: 4673-4680), using their respective default parameters.
When the other polypeptide (or protein) has diverged from the mature polypeptide of
SEQ ID NO: 2 such that traditional sequence-based comparison fails to detect their relationship (Lindahl and Elofsson, 2000, J. Mol. Biol. 295: 613-615), other pairwise sequence comparison algorithms can be used. Greater sensitivity in sequence-based searching can be attained using search programs that utilize probabilistic representations of polypeptide families (profiles) to search databases. For example, the PSI-BLAST program generates profiles through an iterative database search process and is capable of detecting remote homologs (Atschul et al., 1997, Nucleic Acids Res. 25: 3389-3402). Even greater sensitivity can be achieved if the family or superfamily for the polypeptide has one or more representatives in the protein structure databases. Programs such as GenTH READER (Jones, 1999, J. Mol. Biol. 287: 797-815; McGuffin and Jones, 2003, Bioinformatics 19: 874-881 ) utilize information from a variety of sources (PSI-BLAST, secondary structure prediction, structural alignment profiles, and solvation potentials) as input to a neural network that predicts the structural fold for a query sequence. Similarly, the method of Gough et al., 2000, J. Mol. Biol. 313: 903-919, can be used to align a sequence of unknown structure with the superfamily models present in the SCOP database. These alignments can in turn be used to generate homology models for the polypeptide, and such models can be assessed for accuracy using a variety of tools developed for that purpose.
For proteins of known structure, several tools and resources are available for retrieving and generating structural alignments. For example the SCOP superfamilies of proteins have been structurally aligned, and those alignments are accessible and downloadable. Two or more protein structures can be aligned using a variety of algorithms such as the distance alignment matrix (Holm and Sander, 1998, Proteins 33: 88-96) or combinatorial extension (Shindyalov and Bourne, 1998, Protein Engineering 11 : 739-747), and implementation of these algorithms can additionally be utilized to query structure databases with a structure of interest in order to discover possible structural homologs (e.g., Holm and Park, 2000, Bioinformatics 16: 566-567).
In describing the albumin variants of the present invention, the nomenclature described below is adapted for ease of reference. The accepted lUPAC single letter or three letter amino acid abbreviation is employed. The term 'point mutation' and/or 'alteration' includes deletions, insertions and substitutions.
Substitutions. For an amino acid substitution , the following nomenclature is used : Original amino acid, position, substituted amino acid. Accordingly, the substitution of threonine at position 226 with alanine is designated as "Thr226Ala" or "T226A". Multiple mutations (or alterations) are separated by addition marks ("+"), e.g., "Gly205Arg + Ser41 1 Phe" or "G205R + S41 1 F", representing substitutions at positions 205 and 41 1 of glycine (G) with arginine (R) and serine (S) with phenylalanine (F), respectively. The Figures also use ("/"), e.g., "E492T/N503D" this should be viewed as interchangeable with ("+").
Deletions. For an amino acid deletion, the following nomenclature is used: Original amino acid, position*. Accordingly, the deletion of glycine at position 195 is designated as "Gly195*" or "G195*". Multiple deletions are separated by addition marks ("+"), e.g., "Gly195* + Ser41 1 *" or "G195* + S41 1 *".
Insertions. As disclosed above, an insertion may be to the N-side ('upstream', 'X-1 ') or C-side ('downstream', 'X+1 ') of the amino acid occupying a position ('the named (or original) amino acid', 'X').
For an amino acid insertion to the C-side ('downstream', 'X+1 ') of the original amino acid
('X'), the following nomenclature is used: Original amino acid, position, original amino acid, inserted amino acid . Accordingly the insertion of lysine after glycine at position 1 95 is designated "Gly195Glyl_ys" or "G195GK". An insertion of multiple amino acids is designated [Original amino acid, position, original amino acid, inserted amino acid #1 , inserted amino acid #2; etc.]. For example, the insertion of lysine and alanine after glycine at position 1 95 is indicated as "Gly195Glyl_ysAla" or "G195GKA".
In such cases the inserted amino acid residue(s) are numbered by the addition of lower case letters to the position number of the amino acid residue preceding the inserted amino acid residue(s). In the above example, the sequence would thus be:
Figure imgf000018_0001
For an amino acid insertion to the N-side ('upstream', 'X-1 ') of the original amino acid (X), the following nomenclature is used: Original amino acid, position, inserted amino acid, original amino acid. Accordingly the insertion of lysine (K) before glycine (G) at position 195 is designated "Gly195LysGly" or "G195KG". An insertion of multiple amino acids is designated [Original amino acid, position, inserted amino acid #1 , inserted amino acid #2; etc., original amino acid]. For example, the insertion of lysine (K) and alanine (A) before glycine at position 195 is indicated as "Gly195LysAlaGly" or "G195KAG". In such cases the inserted amino acid residue(s) are numbered by the addition of lower case letters with prime to the position number of the amino acid residue following the inserted amino acid residue(s). In the above example, the sequence would thus be:
Figure imgf000019_0001
Multiple alterations. Variants comprising multiple alterations are separated by addition marks ("+"), e.g., "Arg170Tyr+Gly195Glu" or "R170Y+G195E" representing a substitution of arginine and glycine at positions 170 and 195 tyrosine and glutamic acid, respectively.
Different alterations. Where different alterations can be introduced at a position, the different alterations are separated by a comma, e.g., "Arg170Tyr,Glu" represents a substitution of arginine at position 170 with tyrosine or glutamic acid. Thus, "Tyr167Gly,Ala + Arg170Gly,Ala" designates the following variants:
"Tyr1 67 G ly+Arg 1 70 G ly" , "Tyr1 67 G ly+Arg 1 70Al a" , "Tyr1 67Al a +Arg 1 70 G ly" , a n d "Tyr167Ala+Arg170Ala".
Detailed Description of the Invention
The present invention relates to albumin variants, comprising an alteration at two or more positions selected among the group consisting of positions 492, 550, 573, 574 and 580 of the mature polypeptide of S EQ I D N O : 2, or at equivalent positions in other albumins or fragments thereof. Variants
A first aspect of the invention provides polypeptides which are variant albumins or fragments thereof, or fusion polypeptides comprising the variant albumin or fragments thereof, of a parent albumin, comprising alterations at two or more positions corresponding to positions selected among the group consisting of positions 492, 550, 573, 574 and 580 of the mature polypeptide of SEQ ID NO: 2. It is preferred that the two or more alterations comprise alterations at positions corresponding to the following positions in SEQ ID NO: 2:
(a) 492 and 580; and/or
(b) 492 and 574; and/or
(c) 492 and 550; and/or
(d) 550 and 573; and/or
(e) 550 and 574; and/or
(f) 550 and 580; and/or
and/or that
(g) the two or more alterations comprise:
• an alteration to generate at a position corresponding to position 492 of SEQ ID NO: 2 an amino acid from the group consisting of A, C, D, F, H, I, K, L, M, N, P, Q, R, S, T, V, W, Y, preferably D and an alteration to generate at a position corresponding to position 573 of SEQ I D NO: 2 to an amino acid from the group consisting of C, D, E, F, G, H, I, L, M, N, Q, R, S, T, V, W, Y, preferably Y, W or H or
• an alteration to generate at a position corresponding to position 492 of SEQ ID NO: 2 a G and an alteration to generate at a position corresponding to position 573 an A or P and an additional alteration at a position selected from the group consisting of 550,574 and 580; and/or
(h) the two or more alterations comprise:
• an alteration to generate at a position corresponding to position 573 of SEQ ID NO: 2 an amino acid from the group consisting of A, C, D, E, F, G, H, I, L, M, N, Q, R, S, T, V, W, Y, preferably Y, W or H and an alteration to generate at a position corresponding to position 574 of SEQ ID NO: 2 an amino acid from the group consisting of A, C, D, E, F, G, H, I , L, M, P, Q, R, S, T, V, W, Y, H , D, F, G, N , S or Y, more preferably H , D, F or G, most preferably H or
• an alteration to generate at a position corresponding to position 573 of SEQ I D NO: 2 a P and an alteration to generate at a position corresponding to position 574 of SEQ ID NO: 2 an N and an additional alteration at a position selected from the group consisting of 492, 550, and 580; and/or
(i) the two or more alterations comprise:
• an alteration to generate at a position corresponding to position 573 of SEQ ID NO: 2 an amino acid from the group consisting of A, C, D, E, F, G, H, I, L, M, N, Q, R, S, T, V, W, Y, preferably Y, W or H and an alteration to generate at a position corresponding to position 580 of SEQ ID NO: 2 an amino acid from the group consisting of C, D, E, F, G, H, I, L, M,
N, P, R, S, T, V, W, Y or
• an alteration to generate at a position corresponding to position 573 of SEQ I D NO: 2 a P and an alteration to generate at a position corresponding to position 580 of SEQ ID NO: 2 a K and an additional alteration at a position selected from the group consisting of 492, 550, and 574; and/or
(j) the two or more alterations comprise:
• an alteration to generate at a position corresponding to position 574 of SEQ ID NO: 2 an amino acid from the group consisting of A, C, D, E, F, G, H, I, L, M, P, Q, R, S, T, V, W, Y, H , D, F, G, N , S or Y, more preferably H and an alteration to generate at a position corresponding to position 580 of SEQ ID NO: 2 to an amino acid from the group consisting of A, C, D, E, F, G, H, I, L, M, N, P, R, S, T, V, W, Y or
• an alteration to generate at a position corresponding to position 574 of SEQ ID NO: 2 an N and an alteration to generate at a position corresponding to position 580 of SEQ ID NO: 2 a K and an additional alteration at a position selected from the group consisting of 492, 550, and 573
The polypeptide may comprise, three or more, four or more or five or more alterations as described in paragraphs (a), (b), (c), (d), (e), (f), (g), (h), (i) and ( ).
A preferred alteration is a substitution.
It is preferred that the parent albumin and/or the variant albumin comprises or consists of:
(a) a polypeptide having at least 60% sequence identity to the mature polypeptide of
SEQ ID NO: 2;
(b) a polypeptide encoded by a polynucleotide that hybridizes under low stringency conditions with (i) the mature polypeptide coding sequence of SEQ ID NO: 1 , or (ii) the full-length complement of (i);
(c) a polypeptide encoded by a polynucleotide having at least 60% identity to the mature polypeptide coding sequence of SEQ ID NO: 1 ; and/or
(d) a fragment of the mature polypeptide of SEQ ID NO: 2.
The variants of albumin or fragments thereof or fusion polypeptides comprising albumin or fragments thereof comprise alterations, such as substitutions, deletions or insertions at two or more of positions selected among the group consisting of positions 492, 550, 573, 574 and 580 of the mature polypeptide of SEQ I D NO: 2 or in equivalent positions of other albumins or variants or fragments thereof. A stop codon may be introduced in addition to the alterations described herein and if introduced is at position 574 or further downstream (e.g. in SEQ ID NO: 2 it is introduced at from position 574 to 585).
The variant albumin, a fragment thereof, or albumin part of a fusion polypeptide comprising variant albumin or a fragment thereof according to the invention has generally a sequence identity to the sequence of HSA shown in SEQ ID NO: 2 of at least 60%, preferably at least 70%, preferably at least 80%, preferably at least 85%, preferably at least 90 %, more preferred at least 95%, more preferred at least 96%, more preferred at least 97%, more preferred at least 98% and most preferred at least 99%. The variant has less than 100% identity to SEQ ID NO: 2.
The variant albumin, a fragment thereof, or albumin part of a fusion polypeptide comprising variant albumin or a fragment thereof according to the invention has generally a sequence identity to the sequence of the parent albumin of at least 60%, preferably at least 70%, preferably at least 80%, preferably at least 85%, preferably at least 90 %, more preferred at least 95%, more preferred at least 96%, more preferred at least 97%, more preferred at least 98% and most preferred at least 99%. The variant has less than 100% identity to the sequence of the parent albumin.
In one aspect, the number of alterations in the variants of the invention is 1 to 20, e.g., 1 to 10 and 1 to 5, such as 1 , 2, 3, 4, 5, 6, 7, 8, 9 or 10 alterations relative to SEQ ID NO: 2 or relative to the sequence of the parent albumin.
At position 492 of SEQ ID NO: 2 (or equivalent position of other albumins or variants or fragments thereof), it is preferred that the alteration is a substitution, such as from the native amino acid to A, C, D, E, F, G, H, I, K, L, M, N, P, Q, R, S, T, V, W, Y, more preferred to G, D, F, H, M or R, even more preferred to G, or D and most preferred to G. In SEQ ID NO: 2 the native amino acid at position 492 is E, therefore a substitution to E is not preferred.
At position 550 of SEQ ID NO: 2 (or equivalent position of other albumins or variants or fragments thereof), it is preferred that the alteration is a substitution, such as from the native amino acid to A, C, D, E, F, G, H, I, K, L, M, N, P, Q, R, S, T, V, W, Y, more preferred to K, L, M, E or R, even more preferred to K, L or M and most preferred to K. In SEQ ID NO: 2 the native amino acid at position 550 is D, therefore a substitution to D is not preferred.
At position 573 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof), it is preferred that the alteration is a substitution, such as from the native amino acid to A, C, D, E, F, G, H, I, K, L, M, N, P, Q, R, S, T, V, W, Y, more preferred to P, Y, W, H, F, T, I or V, even more preferred to P, Y or W and most preferred to P. In SEQ ID NO: 2 the native amino acid at position 573 is K, therefore a substitution to K is not preferred.
At position 574 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof), it is preferred that the alteration is a substitution, such as from the native amino acid to A, C, D, E, F, G, H, I, K, L, M, N, P, Q, R, S, T, V, W, Y, more preferred to H, G, D, F, N, S or Y, even more preferred to D, F, G or H and most preferred to H. In SEQ ID NO: 2 the native amino acid at position 574 is K, therefore a substitution to K is not preferred.
At position 580 of SEQ ID NO: 2 (or equivalent position of other albumins or variants or fragments thereof), it is preferred that the alteration is a substitution, such as from the native amino acid to A, C, D, E, F, G, H, I, K, L, M, N, P, Q, R, S, T, V, W, Y, more preferred to K or R, most preferred to K. In SEQ I D NO: 2 the native amino acid at position 580 is Q, therefore a substitution to Q is not preferred.
A variant albumin may comprise alterations at positions corresponding to positions 492+550 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492D+550K (e.g. SEQ I D NO: 231 ), or 492G+550K (e.g. SEQ ID NO: 240).
A variant albumin may comprise alterations at positions corresponding to positions 492+573 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492F+573P (e.g. SEQ ID NO: 109), 492G+573P (e.g. SEQ ID NO: 1 10), 492H+573P (e.g. SEQ ID NO: 1 1 1 ) or 492R+573P (e.g. SEQ I D NO: 1 13) or more preferably 492D+573P (e.g. SEQ ID NO: 108). However, it is preferred that the variant does not consist of SEQ I D NO: 2 with only alterations 492G+573A, 492G+573A, 492G+N503K+573A, 492G+N503H+573A, 492G+573P, 492G+N503K+573 P o r 492G+N503H+573P.
A variant albumin may comprise alterations at positions corresponding to positions 492+574 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492D+574H (e.g. SEQ I D NO: 232), 492G+574H (e.g. SEQ ID NO: 241 ) or 492D+574H (e.g. SEQ ID NO: 232).
A variant albumin may comprise alterations at positions corresponding to positions 492+580 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof).
A variant albumin may comprise alterations at positions corresponding to positions 550+573 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 550K+573P (e.g. SEQ ID NO: 1 17).
A variant albumin may comprise alterations at positions corresponding to positions 550+574 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 550K+574H (e.g. SEQ ID NO: 130), 550M+574H
(e.g. SEQ ID NO: 249), 550M+574H (e.g. SEQ ID NO: 249) or 550L+574H (e.g . S EQ I D NO: 245).
A variant albumin may comprise alterations at positions corresponding to positions 550+580 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 550K+580K (e.g. SEQ ID NO: 131 ), 550M+580K (e.g. SEQ ID NO: 251 ) or 550L+580K (e.g. SEQ ID NO: 247).
A variant albumin may comprise alterations at positions corresponding to positions 573+574 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 574D+573P (e.g. SEQ I D NO: 121 ), 574F+573P (e.g. SEQ ID NO: 122), 574G+573P (e.g. SEQ ID NO: 123), 574H+573P (e.g . S EQ I D NO: 124), 574N+573P (e.g. SEQ ID NO: 125) or 574S+573P (e . g . S E Q I D N O : 126). It is preferred that the variant does not consist of SEQ I D NO: 2 with only alterations K573P+K574N+A577T+A578R+S579C+Q580K+A581 D+G584A.
A variant albumin may comprise alterations at positions corresponding to positions 573+580 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 580K+573P ( e . g . S E Q I D N O : 1 2 8 ) o r 580R+573P (e.g. SEQ ID NO: 129). However, it is preferred that the variant does not consist of SEQ ID NO: 2 with only alterations K573P+A577E+A578S+Q580K+A582T.
A variant albumin may comprise alterations at positions corresponding to positions 574+580 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof).
A variant albumin may comprise alterations at positions corresponding to positions 492+550+573 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492G+550K+573P (e.g. SEQ ID NO: 254) or 492D+550K+573P (e.g. SEQ ID NO: 253).
A variant albumin may comprise alterations at positions corresponding to positions
492+550+574 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492G+550K+574H (e.g. SEQ ID NO: 255).
A variant albumin may comprise alterations at positions corresponding to positions 492+550+580 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492D+550K+580K (e.g. SEQ ID NO: 258) or 492G+550K+580K (e.g. SEQ ID NO: 259).
A variant albumin may comprise alterations at positions corresponding to positions 492+573+574 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492D+573P+574H (e . g . S E Q I D N O : 233).
A variant albumin may comprise alterations at positions corresponding to positions 492+573+580 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492D+573P+580K (e.g. SEQ ID NO: 234) or 492G+573P+580K (e.g. SEQ ID NO: 242).
A variant albumin may comprise alterations at positions corresponding to positions
492+574+580 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492D+574H+580K (SEQ I D NO: 262) or 492G+574H+580K (e.g. SEQ ID NO: 263).
A variant albumin may comprise alterations at positions corresponding to positions 50+573+574 of SEQ ID NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 550K+574H+573P( e . g . S E Q I D N O : 1 3 1 ) , 550L+573P+574H (e.g. SEQ ID NO: 246) or 550M+573P+574H (e.g. SEQ ID NO: 250). A variant albumin may comprise alterations at positions corresponding to positions 550+573+580 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 550L+573P+580K (e . g . S E Q I D N O : 248) or 550M+573P+580K (e.g. SEQ ID NO: 252).
A variant albumin may comprise alterations at positions corresponding to positions
550+574+580 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof).
A variant albumin may comprise alterations at positions corresponding to positions 573+574+580 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 574H+580K+573P (e . g . S E Q I D N O : 135).
A variant albumin may comprise alterations at positions corresponding to positions 492+550+573+574 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492G+550K+573P+574H (e.g. SEQ ID NO: 257) or 492D+550K+573P+574H (e.g. SEQ ID NO: 256).
A variant albumin may comprise alterations at positions corresponding to positions 492+550+573+580 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492D+550K+573P+580K (e.g . S EQ I D NO: 260) or 492G+550K+573P+580K (e.g. SEQ ID NO: 261 ).
A variant albumin may comprise alterations at positions corresponding to positions
492+550+574+580 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof).
A variant albumin may comprise alterations at positions corresponding to positions 492+573+574+580 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492D+573P+574H+580K (e.g . S EQ I D NO: 1 14) or 492G+573P+574H+580K (e.g. SEQ I D NO: 1 15), 492F+573P+574G+580K (e.g. SEQ ID NO: 238), 492G+573P+574G+580K ( e . g . S E Q I D N O : 2 3 4 ) , 492D+573P+574G+580K (e.g. SEQ I D NO: 235), 492F+573P+574H+580R (e.g. SEQ I D NO: 239), 492D+573P+574H+580K (e.g. SEQ ID NO: 264), 492G+573P+574H+580R (e.g. SEQ ID NO: 244), 492D+573P+574H+580R (e.g. SEQ ID NO: 236) or 492F+573P+574H+580K (e.g. SEQ ID NO: 237).
A variant albumin may comprise alterations at positions corresponding to positions 550+573+574+580 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 550K+573P+574H+580K (e.g . S EQ I D NO: 265).
A variant albumin may comprise alterations at positions corresponding to positions 492+550+573+574+580 of SEQ ID NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492D+550K+573P+574H+580K (e.g. SEQ ID NO: 266) or 492G+550K+573P+574H+580K (e.g. SEQ I D NO: 267). Such alterations may comprise 492D+550K+573P+574H (e.g. SEQ ID NO: 256).
Particularly preferred variants include:
a variant albumin with alterations at positions corresponding to positions 492 and 580 in
SEQ ID NO: 2, such as (i) E492G and Q580K, or (ii) E492D and Q580K (or equivalent positions of other albumins or variants or fragment thereof);
a variant albumin comprising alterations at positions corresponding to positions 492 and 574 in SEQ I D NO: 2, such as (i) E492G and K574H, (ii) E492D and K574H , (iii) E492D and K574K, or (iv) E492G and K574K (or equivalent positions of other albumins or variants or fragment thereof);
a variant albumin comprising alterations at positions corresponding to positions 492 and 550 in SEQ I D NO: 2, such as (i) E492G and D550K, or (ii) E492D and D550K (or equivalent positions of other albumins or variants or fragment thereof);
a variant albumin comprising alterations at positions corresponding to positions 550 and
573 in SEQ ID NO: 2, such as (i) D550K and K573P, (ii) D550L and K573P or (iii) D550M and K573P (or equivalent positions of other albumins or variants or fragment thereof);
a variant albumin comprising alterations at positions corresponding to positions 550 and
574 in SEQ I D NO: 2, such as (i) D550K and K574H, or (ii) D550L and K574H (or equivalent positions of other albumins or variants or fragment thereof);
a variant albumin comprising alterations at positions corresponding to positions 550 and 580 in SEQ I D NO: 2, such as (i) D550M and Q580K, (ii) D550L and Q580K or (iii) D550K and Q580K (or equivalent positions of other albumins or variants or fragment thereof);
a va ri ant albumin with alterations (e.g. comprising alterations) at positions corresponding to positions 580 and 573 in SEQ I D NO: 2, such as Q580K and K573P (or equivalent positions of other albumins or variants or fragment thereof) and preferably one or more (several) other alterations such at a position selected from 492, 550 and 574. If there is a K at position 580 and a P at position 573 it is preferred that the polypeptide comprises an additional alteration at a position selected from the group consisting of 492, 550, and 574;
a variant albu m i n with alterations (e.g. comprising alterations) at positions corresponding to positions 492 and 573 in SEQ ID NO: 2, such as:
a variant albumin comprising alterations at positions corresponding to positions 492 and 573 in SEQ ID NO: 2, such as (i) E492D and K573P (or equivalent positions of other albumins or variants or fragment thereof) or (ii) E492G and K573P or (iii) E492G and K573A (or equivalent positions of other albumins or variants or fragment thereof), and preferably one or more (several) other alterations such at a position selected from 550, 574 and 580. If there is a
G at position 492 and an A or P at position 573 it is preferred that the polypeptide comprises an additional alteration at a position selected from the group consisting of 550, 574 and 580 (or equivalent positions of other albumins or variants or fragment thereof);
a variant albumin comprising alterations at positions corresponding to positions 573 and 574 in SEQ ID NO: 2, such as K573P and K574H (or equivalent positions of other albumins or variants or fragment thereof, and preferably one or more (several) alterations such as at a position selected from 492, 550 and 580. If there is a P at position 573 and an N at position 574 it is preferred that the polypeptide comprises an additional alteration at a position selected from the group consisting of 492, 550, and 580 (or equivalent positions of other albumins or variants or fragment thereof);
a variant albumin comprising alterations at positions corresponding to positions 574 and 580 in SEQ ID NO: 2, such as 574H and 580K (or equivalent positions of other albumins or variants or fragment thereof, and preferably one or more (several) alterations such as at a position selected from 492, 550 and 573. If there is a N at position 574 and a K at position 580 it is preferred that the polypeptide comprises an additional alteration at a position selected from the group consisting of 492, 550, and 573 (or equivalent positions of other albumins or variants or fragment thereof);
a variant albumin comprising alterations at positions corresponding to positions 492, 550 and 573 in SEQ ID NO: 2, such as E492G, D550K and K573P e.g. SEQ ID NO: 254 or such as E492D, D550K and K573P e.g. SEQ ID NO: 253;
a variant albumin comprising alterations at positions corresponding to positions 550, 573 and 580 in SEQ ID NO: 2, such as D550K, K573P and Q580K e.g. SEQ ID NO: 133;
a variant albumin comprising alterations at positions corresponding to positions 550, 573 and 580 in SEQ ID NO: 2, such as D550L, K573P and Q580K e.g. SEQ I D NO:248 or such as D550M, K573P and Q580K e.g. SEQ ID NO:252;
a variant albumin comprising alterations at positions corresponding to positions 550, 573 and 574 in SEQ ID NO: 2, such as D550L, K573P and K574H e.g. SEQ ID NO:246;
a variant albumin comprising alterations at positions corresponding to positions 550, 573 and 574 in SEQ ID NO: 2, such as D550K, K573P and K574H e.g. SEQ ID NO: 131 ;
a variant albumin comprising alterations at positions corresponding to positions 492, 573 and 580 in SEQ ID NO: 2, such as E492G, K573P and Q580K e.g. SEQ ID NO: 242;
a variant albumin comprising alterations at positions corresponding to positions 492, 573 and 580 in SEQ ID NO: 2, such as E492D, K573P and Q580K e.g. SEQ ID NO: 234;
a variant albumin comprising alterations at positions corresponding to positions 492, 550, 573 and 574 in SEQ I D NO: 2, such as E492D, D550K, K573P and K574H e.g. SEQ ID NO: 256;
a variant albumin comprising alterations at positions corresponding to positions 492,
550, 573 and 574 in SEQ I D NO: 2, such as E492G, D550K, K573P and K574H e.g. SEQ ID NO: 257;
a variant albumin comprising alterations at positions corresponding to positions 573, 574 and 580 in SEQ ID NO: 2, such as K573P, K574H and Q580K e.g. SEQ ID NO: 135;
a variant albumin with alterations (e.g. comprising alterations) at positions corresponding to positions 492, 573, 574 and 580 in SEQ I D NO: 2, such as E492D, K573P, K574H and Q580K (or equivalent positions of other albumins or variants or fragment thereof), e.g. SEQ ID NO: 1 14;
a variant albumin with alterations (e.g. comprising alterations) at positions corresponding to positions 492, 573, 574 and 580 in SEQ ID NO: 2, such as E492G, K573P, K574H and Q580K (or equivalent positions of other albumins or variants or fragment thereof), e.g. SEQ ID NO: 1 15.
It is preferred that the variant albumin does not consist of SEQ ID NO: 2 with only the following alterations: K573P+K574N+A577T+A578R+S579C+Q580K+A581 D+G584A;
K573P+A577E+A578S+Q580K+A582T; E492G+K573A; E492G+N503K+K573A;
E492G+N503H+K573A; E492G+K573P; E492G+N503K+K573P; E492G+N503H+K573P.
Such variant albumins are disclosed in WO201 1/051489.
It is preferred that the variant albumin does not consist of SEQ I D NO: 2 with amino aci d s 573 to 585 replaced with (a ) KKLVAASQAALG L , (b) P KFVAASQAALA, (c)
PNLVTRCKDALA, (d) PKLVESSKATLG or (e) PKLVASTQAALA.
It is preferred that the variant albu min , a fragment thereof or fusion polypeptide comprising the variant albumin or fragment thereof has altered binding affinity to FcRn and/or an altered plasma half-life compared with the corresponding parent or reference albumin, fragment thereof, or fusion polypeptide comprising the variant albumin or fragment thereof and/or an altered binding affinity to FcRn.
In a particularly preferred embodiment the parent or reference albumin is HSA (SEQ ID
NO: 2) and the variant albumin, a fragment thereof or fusion polypeptide comprising the variant albumin or fragment thereof has altered binding affinity to FcRn and/or an altered plasma half- life compared with the HSA, the corresponding fragment or fusion polypeptide comprising HSA or fragment thereof and/or an altered binding affinity to FcRn.
The correlation between binding of albumin to its receptor and plasma half-life has been realized by the present inventors based on the natural occurring allele of HSA D494N. The inventors have previously analyzed this allele and found that it has a lower affinity to its receptor
FcRn than the affinity of WT HSA to FcRn.
Further, it has been disclosed that a transgenic mouse having the natural mouse FcRn replaced with human FcRn has a higher serum albumin level than normal mouse (J Exp Med.
(2003) 197(3):315-22). It has previously been discovered that human FcRn has a higher affinity to mouse serum albumin than mouse FcRn has to mouse serum albumin and, therefore, the observed increase in serum albumin in the transgenic mice corresponds with a higher affinity between serum albumin and its receptor, confirming the correlation between albumin binding to
FcRn and plasma half-life. In addition, variants of albumin that have little or no binding to FcRn have been shown to have reduced half-life in a mouse model, Kenanova et al (2009) J. Nucl. Med.; 50 (Supplement 2): 1582).
One way to determine whether the affinity of a variant albumin to FcRn is higher or lower than the parent or reference albumin is to use the Surface Plasmon Resonance assay (SPR) as described below. The skilled person will understand that other methods might be useful to determine whether the affinity of a variant albumin to FcRn is higher or lower than the affinity of the parent or reference albumin to FcRn, e.g., determination and comparison of the binding constants KD. The binding affinity (KD) between a first molecule (e.g. ligand) and a second molecule (e.g. receptor) is a function of the kinetic constants for association (on rate, ka) and dissociation (off-rate, kd) according to KD= kd/ka. Thus, according to the invention variant albumins having a KD that is lower than the KD for natural HSA is considered to have a higher plasma half-life than HSA and variant albumins having a KD that is higher than the KD for natural HSA is considered to have a lower plasma half-life than HSA.
I n an embodiment of the invention, the variants of albumin or fragments thereof, or fusion polypeptides comprising variant albumin or a fragment thereof according to the invention have a plasma half-life that is longer than the plasma half-life of the parent or reference albumin fragment thereof or fusion polypeptide comprising the parent or reference albumin or a fragment thereof and/or an stronger binding affinity to FcRn.
I n a further embodiment the variants of albumin or fragments thereof, or fusion polypeptides comprising variant albumin or fragments thereof according to the invention have a plasma half-life that is shorter than the plasma half-life of the parent or reference albumin fragment thereof or fusion polypeptide comprising the parent or reference albumin or a fragment thereof and/or an weaker binding affinity to FcRn.
In addition to alterations at two or more positions selected from 492, 550, 573, 574 and/or 580 (or equivalent position of other albumins or variants or fragments thereof) the variant albumin or fragments thereof, or fusion polypeptides comprising variant albumin or fragments thereof according to the invention may contain additional substitutions, deletions or insertions in other positions of the molecules. Such additional substitutions, deletions or insertions may be useful in order to alter other properties of the molecules such as but not limited to altered glycosylation ; i ntrod uction of reactive grou ps of the su rface such a thiol groups, removing/generating a carbamoylation site; etc.
Residues that might be altered in order to provide reactive residues on the surface and which advantageously could be applied to the invention has been disclosed in WO2010/092135 (incorporated herein by reference). Particular preferred residues include the positions corresponding to positions in SEQ ID NO: 2.
As examples of alterations that can be made in SEQ I D NO: 2 or in corresponding positions in other albumins in order to provide a reactive thiol group on the surface includes alterations corresponding to following alterations in SEQ ID NO: 2: L585C, D1 C, A2C, D562C, A364C, A504C, E505C, T79C, E86C, D129C, D549C, A581 C, D121 C, E82C, S270C, A578C, L595LC, D 1 DC, A2AC, D562DC, A364AC, A504AC, E505EC, T79TC, E86EC, D129DC, D549DC, A581AC, A581AC, D121 DC, E82EC, S270SC, A579AC, C360*, C316*, C75*, C168*, C558*, C361 *, C91 *, C124*, C169* and C567*. Alternatively a cysteine residue may be added to the N or C terminal of albumin. The term 'reactive thiol' means and/or includes a thiol group provided by a Cys which is not disulphide bonded to a Cysteine and/or which is sterically available for binding to a partner such as a conjugation partner.
Fusion polypeptides
A second aspect of the invention relates to fusion polypeptides. Therefore, the variants of albumin or fragments thereof according to the invention may be fused with a non-albumin polypeptide fusion partner. The fusion partner may in principle be any polypeptide but generally it is preferred that the fusion partner is a polypeptide having therapeutic, prophylactic (including vaccine), diagnostic, imaging or other beneficial properties. Such properties may be referred to as 'pharmaceutically beneficial properties' . Fusion polypeptides comprising albumin or fragments thereof are known in the art. It has been found that such fusion polypeptides comprising albumin or a fragment thereof and a fusion partner polypeptide have a longer plasma half-life compared to the unfused fusion partner polypeptide alone. According to the invention it is possible to alter the plasma half-life of the fusion polypeptides according to the invention compared to the corresponding fusion polypeptides of the prior art. 'Alter' includes both increasing the plasma half-life or decreasing the plasma half-life. Increasing the plasma half-life is preferred. The invention allows tailoring of half-life to a term desired.
One or more (several) therapeutic, prophylactic (including vaccine), diagnostic, imaging or other beneficial polypeptides may be fused to the N-terminus, the C-terminus of albumin, inserted into a loop in the albumin structure or any combination thereof. It may or it may not comprise linker sequences separating the various components of the fusion polypeptide.
Teachings relating to fusions of albumin or a fragment thereof are known in the art and the skilled person will appreciate that such teachings can also be applied to the invention. WO 2001 /79271 A (particularly page 9 and/or Table 1 ), WO 2003/59934 (particularly Table 1 ), WO03/060071 (particularly Table 1 ) and WO01/079480 (particularly Table 1 ) (each incorporated herein by reference in their entirety) also contain examples of therapeutic, prophylactic (including vaccine), diagnostic, imaging or other beneficial polypeptides that may be fused to albumin or fragments thereof, and these examples apply also to the invention.
Further preferences for the second aspect of the invention include those of the first aspect of the invention and those provided below the twelfth aspect of the invention. The skilled person understands that any aspect of the invention may be combined with another aspect or aspects of the invention and/or with one or more (several) of the preferences for the aspects of the invention and/or other disclosures made herein. Polynucleotides
A third aspect of the invention relates to isolated polynucleotides that encode any of the variants or fusion polypeptides of the invention . The polynucleotide may be an isolated polynucleotide. The polynucleotide may be comprised in a vector (such as a plasmid) and/or in a host cell.
Further preferences for the third aspect of the invention include those of the first aspect of the invention and those provided below the twelfth aspect of the invention. The skilled person understands that any aspect of the invention may be combined with another aspect or aspects of the invention and/or with one or more (several) of the preferences for the aspects of the invention and/or other disclosures made herein.
Nucleic Acid Constructs
A fourth aspect of the invention relates to n ucleic acid constructs comprising a polynucleotide encoding a variant or fusion polypeptide of the invention operably linked to one or more (several) control sequences that direct the expression of the coding sequence in a suitable host cell under conditions compatible with the control sequences.
A polynucleotide may be manipulated in a variety of ways to provide for expression of a variant. Manipulation of the polynucleotide prior to its insertion into a vector may be desirable or necessary depending on the expression vector. The techniques for modifying polynucleotides utilizing recombinant DNA methods are well known in the art.
The control sequence may be a promoter sequence, which is recognized by a host cell for expression of the polynucleotide. The promoter sequence contains transcriptional control sequences that mediate the expression of the variant. The promoter may be any nucleic acid sequence that shows transcriptional activity in the host cell including mutant, truncated, and hybrid promoters, and may be obtained from genes encoding extracellular or intracellular polypeptides either homologous or heterologous to the host cell.
I n a yeast host, useful promoters are obtained from the genes for Saccharomyces cerevisiae enolase (ENO-1 ), Saccharomyces cerevisiae protease A (PRA1 ), Saccharomyces cerevisiae protease B (PRB1 ), Saccharomyces cerevisiae translation elongation factor (TEF1 ),
Saccharomyces cerevisiae translation elongation factor (TEF2), Saccharomyces cerevisiae galactokinase (GAL1 ), Saccharomyces cerevisiae alcohol dehydrogenase/glyceraldehyde-
3-phosphate dehydrogenase (ADH1 , ADH2/GAP), Saccharomyces cerevisiae triose phosphate isomerase (TPI), Saccharomyces cerevisiae metallothionein (CU P1 ), and Saccharomyces cerevisiae 3-phosphoglycerate kinase. Other useful promoters for yeast host cells are described by Romanos et al., 1992, Yeast 8: 423-488.
The skilled person knows useful promoters for use in rice and mammalian cells, such as
CHO or HEK. In a rice host, useful promoters are obtained from cauliflower mosaic virus 35S RNA gene (CaMV35S), maize alcohol dehydrogenase (Adh1 ) and alpha Amy3.
In a mammalian host cell, such as CHO or H EK, useful promoters are obtained from Cytomegalovirus (CMV) and CAG hybrid promoter (hybrid of CMV early enhancer element and chicken beta-actin promoter), Simian vacuolating virus 40 (SV40).
The control sequence may also be a suitable transcription terminator sequence, which is recognized by a host cell to terminate transcription. The terminator sequence is operably linked to the 3'-terminus of the polynucleotide encoding the variant. Any terminator that is functional in the host cell may be used.
P referred term i n ators for yeast h ost cel ls are obtai n ed from th e gen es for Saccharomyces cerevisiae enolase, Saccharomyces cerevisiae cytochrome C (CYC 1 ), Saccharomyces cerevisiae alcohol dehydrogenase (ADH 1 ) and Saccharomyces cerevisiae glyceraldehyde-3-phosphate dehydrogenase. Other useful terminators for yeast host cells are described by Romanos et al., 1992, supra. The skilled person knows useful terminators for use in rice and mammalian cells, such as CHO or H EK. For example, in a rice host, preferred terminators are obtained from Agrobacterium tumefaciens nopaline synthase (Nos) and cauliflower mosaic virus 35S RNA gene (CaMV35S).
The control sequence may also be a suitable leader sequence, a nontranslated region of an mRNA that is important for translation by the host cell. The leader sequence is operably linked to the 5'-terminus of the polynucleotide encoding the variant. Any leader sequence that is functional in the host cell may be used.
Suitable leaders for yeast host cells are obtained from the genes for Saccharomyces cerevisiae enolase (E N O-1 ), Saccharomyces cerevisiae 3-phosphoglycerate kinase, Saccharomyces cerevisiae alpha-facto r, a n d Saccharomyces cerevisiae alcohol dehydrogenase/glyceraldehyde-3-phosphate dehydrogenase (ADH2/GAP).
The control sequence may also be a polyadenylation sequence, a sequence operably linked to the 3'-terminus of the variant-encoding sequence and, when transcribed, is recognized by the host cell as a signal to add polyadenosine residues to transcribed mRNA. Any polyadenylation sequence that is functional in the host cell may be used.
Useful polyadenylation sequences for yeast host cells are described by Guo and Sherman, 1995, Mol. Cellular Biol. 15: 5983-5990.
The control sequence may also be a signal peptide coding region that encodes a signal peptide linked to the N-terminus of a variant and directs the variant into the cell's secretory pathway. The 5'-end of the coding sequence of the polynucleotide may inherently contain a signal peptide coding region naturally linked in translation reading frame with the segment of the coding region that encodes the variant. Alternatively, the 5'-end of the coding sequence may contain a signal peptide coding region that is foreign to the coding sequence. The foreign signal peptide coding region may be required where the coding sequence does not naturally contain a signal peptide coding region. Alternatively, the foreign signal peptide coding region may simply replace the natural signal peptide coding region in order to enhance secretion of the variant. However, any signal peptide coding region that directs the expressed variant into the secretory pathway of a host cell may be used.
U sefu l si gn a l peptid es for yeast h ost cel ls a re obta i n ed from the genes for Saccharomyces cerevisiae alpha-factor and Saccharomyces cerevisiae invertase. Other useful signal peptide coding sequences are described by Romanos et al., 1992, supra. The skilled person knows useful signal peptides for use in rice and mammalian cells, such as CHO or HEK.
Where both signal peptide and propeptide regions are present at the N-terminus of a variant, the propeptide region is positioned next to the N-terminus of the variant and the signal peptide region is positioned next to the N-terminus of the propeptide region.
Further preferences for the fourth aspect of the invention include those of the first aspect of the invention and those provided below the twelfth aspect of the invention. The skilled person understands that any aspect of the invention may be combined with another aspect or aspects of the invention and/or with one or more (several) of the preferences for the aspects of the invention and/or other disclosures made herein.
Preparation of Variants
A fifth aspect of the invention relates to a method for preparing or obtaining a variant albumin or fragment thereof, or fusion polypeptides comprising the variant albumin or fragments thereof, or associates of variant albumin or fragment thereof comprising:
(a) introducing into a parent albumin or fragments thereof, or fusion polypeptides comprising the parent albumin or fragments thereof an alteration at two or more positions selected from 492, 550, 573, 574 and 580, preferably as described for the first aspect of the invention; and
(b) recovering the variant albumin or fragment thereof, or fusion polypeptides comprising the variant albumin or fragment thereof.
Preferred alterations are as described in relation to the first aspect of the invention. The resultant variant albumin or fragment thereof may have altered FcRn-binding affinity compared to the FcRn-binding affinity of a reference such as a parent albumin or fragment which does not comprise the alterations. More preferably, the resultant variant albumin or fragment thereof has a stronger FcRn-binding affinity.
The invention includes a method for preparing a polypeptide which is a variant of albumin, fragment thereof or fusion polypeptide comprising said variant albumin or fragment thereof having a binding affinity to FcRn which is altered compared to the binding affinity of a reference albumin, fragment or fusion thereof to FcRn, comprising:
(a) providing a nucleic acid encoding a parent albumin such as an albumin having at least 60% sequence identity to SEQ ID NO: 2;
(b) modifying the sequence of step (a), to encode a polypeptide which is a variant albumin, fragment thereof or fusion polypeptide comprising said variant albumin or fragment thereof comprising alterations at two or more positions corresponding to positions selected from selected among two or more of the group consisting of positions 492, 550, 573, 574 and 580 in SEQ ID NO: 2;
(c) optionally, introducing the modified sequence of step (b) in a suitable host cell;
(d) optionally, growing the cells in a suitable growth medium under condition leading to expression of the polypeptide; and
(e) optionally, recovering the polypeptide from the growth medium;
wherein the polypeptide has an altered binding affinity to FcRn and/or an altered plasma half-life compared with the half-life of a parent albumin, reference albumin, fragment thereof or fusion polypeptide comprising said parent albumin, reference albumin or fragment or fusion thereof.
It is preferred that the parent albumin and/or the variant albumin comprises or consists of:
(a) a polypeptide having at least 60% sequence identity to the mature polypeptide of SEQ ID NO: 2;
(b) a polypeptide encoded by a polynucleotide that hybridizes under low stringency conditions with (i) the mature polypeptide coding sequence of SEQ I D NO: 1 , or (ii) the f u II- length complement of (i);
(c) a polypeptide encoded by a polynucleotide having at least 60% identity to the mature polypeptide coding sequence of SEQ ID NO: 1 ; and/or
(d) a fragment of the mature polypeptide of SEQ ID NO: 2. The variants can be prepared by those skilled persons using any mutagenesis procedure known in the art, such as site-directed mutagenesis, synthetic gene construction, semi-synthetic gene construction, random mutagenesis, shuffling, etc.
Site-directed mutagenesis is a technique in which one or more (several) mutations (alterations) are created at one or more (several) defined sites in a polynucleotide encoding the parent.
Site-directed mutagenesis can be accomplished in vitro by PCR involving the use of oligonucleotide primers containing the desired mutation. Site-directed mutagenesis can also be performed in vitro by cassette mutagenesis involving the cleavage by a restriction enzyme at a site in the plasmid comprising a polynucleotide encoding the parent and subsequent ligation of an oligonucleotide containing the mutation in the polynucleotide. Usually the restriction enzyme that digests at the plasmid and the oligonucleotide is the same, permitting ligation of the plasmid and insert to one another. See, e.g., Scherer and Davis, 1979, Proc. Natl. Acad. Sci. USA 76:
4949-4955; and Barton et al., 1990, Nucleic Acids Res. 18: 7349-4966. Site-directed mutagenesis can also be accomplished in vivo by methods known in the art. See, e.g., U .S. Patent Application Publication NO: 2004/0171 154; Storici et al., 2001 , Nature Biotechnol. 19: 773-776; Kren et al., 1998, Nat. Med. 4: 285-290; and Calissano and Macino, 1996, Fungal Genet. Newslett. 43: 15-16.
Any site-directed mutagenesis procedure can be used in the invention. There are many commercial kits available that can be used to prepare variants.
Synthetic gene construction entails in vitro synthesis of a designed polynucleotide molecule to encode a polypeptide of interest. Gene synthesis can be performed utilizing a number of techniques, such as the multiplex microchip-based technology described by Tian et al. (2004, Nature 432: 1 050-1054) and similar technologies wherein oligonucleotides are synthesized and assembled upon photo-programmable microfluidic chips.
Single or multiple amino acid substitutions, deletions, and/or insertions can be made and tested using known methods of mutagenesis, recombination, and/or shuffling, followed by a relevant screening procedure, such as those disclosed by Reidhaar-Olson and Sauer, 1988, Science 241 : 53-57; Bowie and Sauer, 1989, Proc. Natl. Acad. Sci. USA 86: 2152-2156; WO 95/17413; or WO 95/22625. Other methods that can be used include error-prone PCR, phage display (e.g., Lowman et al., 1991 , Biochemistry 30: 10832-10837; U.S. Patent NO: 5,223,409; WO 92/06204) and region-directed mutagenesis (Derbyshire et al., 1986, Gene 46: 145; Ner ei a/., 1988, DNA 7: 127).
Mutagenesis/shuffling methods can be combined with high-throughput, automated screening methods to detect activity of cloned, mutagenized polypeptides expressed by host cells (Ness et al., 1999, Nature Biotechnology 17: 893-896). Mutagenized DNA molecules that encode active polypeptides can be recovered from the host cells and rapidly sequenced using standard methods in the art. These methods allow the rapid determination of the importance of individual amino acid residues in a polypeptide.
Semi-synthetic gene construction is accomplished by combining aspects of synthetic gene construction, and/or site-directed mutagenesis, and/or random mutagenesis, and/or shuffling. Semi-synthetic construction is typified by a process utilizing polynucleotide fragments that are synthesized, in combination with PCR techniques. Defined regions of genes may thus be synthesized de novo, while other regions may be amplified using site-specific mutagenic primers, while yet other regions may be subjected to error-prone PCR or non-error prone PCR amplification. Polynucleotide sub sequences may then be shuffled.
Further preferences for the fifth aspect of the invention include those of the first aspect of the invention and those provided below the twelfth aspect of the invention. The skilled person understands that any aspect of the invention may be combined with another aspect or aspects of the invention and/or with one or more (several) of the preferences for the aspects of the invention and/or other disclosures made herein. Methods of Production
A sixth aspect of the invention relates to methods of preparation of a variant according to the invention. The variants of the invention can be prepared using techniques well known to the skilled person. One convenient way is by cloning nucleic acid encoding the parent albumin or a fragment thereof or fusion polypeptide comprising albumin or a fragment thereof, modifying said nucleic acid to introduce the desired substitution(s) at two or more positions corresponding to positions selected from 492, 550, 573, 574 and 580 of the mature polypeptide of SEQ ID NO: 2 (or equivalent positions in other albumins or fragments thereof), preferably as described for the first or fifth aspects of the invention, preparing a suitable genetic construct where the modified nucleic acid is placed in operative connection with suitable regulatory genetic elements, such as promoter, terminator, activation sites, ribosome binding sites etc., introducing the genetic construct into a suitable host organism , culturing the transformed host organism under conditions leading to expression of the variant and recovering the variant. All these techniques are known in the art and it is within the skills of the average practitioner to design a suitable method for preparing a particular variant according to the invention.
The variant polypeptide of the invention may also be connected to a signal sequence in order to have the variant polypeptide secreted into the growth medium during culturing of the transformed host organism. It is generally advantageous to have the variant polypeptide secreted into the growth medium in order to ease recovery and purification.
Tech niq ues for preparing variant polypeptides have also been disclosed in WO
2009019314 (included by reference) and these techniques may also be applied to the invention.
Albumins have been successfully expressed as recombinant proteins in a range of hosts including fungi (including but not limited to Aspergillus (WO06066595), Kluyveromyces (Fleer 1991 , Bio/technology 9, 968-975), Pichia (Kobayashi 1998 Therapeutic Apheresis 2, 257-262) and Saccharomyces (Sleep 1990, Bio/technology 8, 42-46)), bacteria (Pandjaitab 2000, J. Allergy Clin. Immunol. 105, 279-285)), animals (Barash 1993, Transgenic Research 2, 266-276) and plants (including but not limited to potato and tobacco (Sijmons 1990, Bio/technology 8, 217 and Farran 2002, Transgenic Research 11 , 337-346) and rice e.g. Oryza sativa) and mammalian cells such as CHO and HEK. The variant polypeptide of the invention is preferably produced recombinantly in a suitable host cell. In principle any host cell capable of producing a polypeptide in su itable amounts may be used and it is within the skills of the average practitioner to select a suitable host cell according to the invention. A preferred host organism is yeast, preferably selected among Saccharomycacae, more preferred Saccharomyces cerevisiae.
The variant polypeptides of the invention may be recovered and purified from the growth medium using a combination of known separation techniques such as filtration, centrifugation, chromatography, and affinity separation techniques etc. It is within the skills of the average practitioner to purify the variants of the invention using a particular combination of such known separation steps. As an example of purification techniques that may be applied to the variants of the invention can be mentioned the teaching of WO00/44772.
The variant polypeptides of the invention may be used for delivering a therapeutically beneficial compound (including prophylactically beneficial compound such as a vaccine) to an animal or a human individual in need thereof. Such therapeutically beneficial compounds include, but are not limited, to labels and readily detectable compounds for use in diagnostics, such as various imaging techniques; pharmaceutical active compounds such as drugs, or specifically binding moieties such as antibodies. The variants of the invention may even be connected to two or more different therapeutically beneficial compounds, e.g. , an antibody and a drug, which gives the combined molecule the ability to bind specifically to a desired target and thereby provide a high concentration of the connected drug at that particular target.
Further preferences for the sixth aspect of the invention include those of the first aspect of the invention and those provided below the twelfth aspect of the invention. The skilled person understands that any aspect of the invention may be combined with another aspect or aspects of the invention and/or with one or more (several) of the preferences for the aspects of the invention and/or other disclosures made herein.
Conjugates
A seventh aspect of the invention relates to conjugates (conjugations). Therefore, the variants of albumin or fragments thereof or fusion polypeptides according to the invention may be conjugated to a second molecule ('conjugation partner') using techniques known within the art. The conjugation partner may be a therapeutic, prophylactic (including vaccine), diagnostic, imaging or other beneficial moiety. Said conjugation partner may be a polypeptide or a non- polypeptide chemical. The conjugation partner may be a polypeptide, chemical (e.g. chemically synthesised drug) or a nucleic acid (e.g. DNA, RNA, siRNA).
Said second molecule may comprise a diagnostic or imaging moiety, and in this embodiment the conjugate may be useful as a diagnostic tool such as in imaging; or the second molecule may be a therapeutic or prophylactic (e.g. vaccine) compound and in this embodiment the conjugate may be used for therapeutic or prophylactic (e.g. vaccination) purposes where the conjugate will have the therapeutic or prophylactic properties of the therapeutic or prophylactic compound as well as the desirable plasma half-life provided by the albumin part of the conjugate. Conjugates of albumin and a therapeutic molecule are known in the art and it has been verified that such conjugates have long plasma half-life compared with the non- conjugated, free therapeutic molecule as such. According to the invention it is possible to alter the binding affinity to FcRn and/or plasma half-life of the conjugate according to the invention compared to the corresponding conjugates of the prior art. 'Alter' includes both increasing the plasma half-life and decreasing the plasma half-life binding affinity to FcRn and/or increasing the binding affinity and decreasing the binding affinity to FcRn. Increasing the plasma half-life and/or binding affinity to FcRn is preferred. The conjugates may conveniently be linked via a free thiol group present on the surface of HSA (amino acid residue 34 of mature HSA) using well known chemistry.
In one particular preferred aspect the variant albumin or fragment thereof is conjugated to a beneficial therapeutic or prophylactic (including vaccine) compound and the conjugate is used for treatment of a condition in a patient in need thereof, which condition is responsive to the particular selected therapeutic compound. Techniques for conjugating such a therapeutically useful compou nd to the variant albumin or fragment thereof are known in the art. WO 2009/019314 (incorporated herein by reference in its entirety) discloses examples of techniques suitable for conjugating a therapeutically compound to a polypeptide which techniques can also be applied to the invention. Further WO 2009/019314 discloses examples of compounds and moieties that may be conjugated to substituted transferrin and these examples may also be applied to the invention. The teaching of WO 2009/019314 is included herein by reference.
HSA contains in its natural form one free thiol group (at Cys34) that conveniently may be used for conjugation. As a particular embodiment within this aspect the variant albumin or fragment thereof may comprise further modifications provided to generate additional free thiol groups on the surface. This has the benefit that the payload of the variant albumin or fragment thereof is increased so that more than one molecule of the therapeutic (e.g. prophylactic) compound can be conjugated to each molecule of variant albumin or fragment thereof, or two or more (several) different therapeutic compounds may be conjugated to each molecule of variant albumin or fragment thereof, e.g., a compound having targeting properties such as an antibody specific for example a tumour; and a cytotoxic drug conjugated to the variant albumin or fragment thereof thereby creating a highly specific drug against a tumour. Teaching of particular residues that may be modified to provide for further free thiol groups on the surface can be found in co-pending patent application WO 2010/092135, which is incorporated by reference.
The conjugation partner may alternatively be conjugated to a fusion polypeptide (described herein), resulting in a molecule comprising a fusion partner fused to the albumin as well as a conjugation partner conjugated to the same albumin or even to the fusion partner.
Further preferences for the seventh aspect of the invention include those of the first aspect of the invention and those provided below the twelfth aspect of the invention. The skilled person understands that any aspect of the invention may be combined with another aspect or aspects of the invention and/or with one or more (several) of the preferences for the aspects of the invention and/or other disclosures made herein. Associates
An eighth aspect of the invention relates to associates. Therefore, the variants of albumin or fragments thereof or fusion polypeptides may further be used in form of "associates".
In this connection the term "associate" is intended to mean a compound comprising a variant of albumin or a fragment thereof and another compound bound or associated to the variant albumin or fragment thereof by non-covalent binding. As an example of such an associate can be mentioned an associate consisting of variant albumin and a lipid associated to albumin by a hydrophobic interaction. Such associates are known in the art and they may be prepared using well known techniques. As an example of a preferred associate according to the invention can be mentioned, an associate comprising variant albumin and a taxane, a taxol or taxol derivative (e.g. paclitaxel). Further examples of associates comprise a therapeutic, prophylactic (including vaccine), diagnostic, imaging or other beneficial moiety.
The half-life of an albumin associate according to the invention may be longer or shorter than the half-life of the 'other compound' alone. The half-life of an albumin associate according to the invention may be longer or shorter than the half-life of the analogous / equivalent albumin associate comprising or consisting of a reference albumin such as native HSA (instead of an albumin variant or derivative according to the invention) and the 'other compound'. Likewise, the binding affinity to FcRn of an albumin associate according to the invention may be stronger or weaker than the binding affinity to FcRn of the analogous / equivalent albumin associate comprising or consisting of a reference albumin such as native HSA (instead of an albumin variant or derivative according to the invention) and the 'other compound'. Methods for the preparation of associates are well-known to the skilled person, for example, formulation (by association) of HSA with Lipo-compounds is described in Hussain, R. and Siligardi, G. (2006) International Journal of Peptide Research and Therapeutics, Vol. 12, NO: 3, pp. 31 1-315.
Further preferences for the eighth aspect of the invention include those of the first aspect of the invention and those provided below the twelfth aspect of the invention. The skilled person understands that any aspect of the invention may be combined with another aspect or aspects of the invention and/or with one or more (several) of the preferences for the aspects of the invention and/or other disclosures made herein.
Compositions
A ninth aspect of the invention relates to compositions. Therefore the invention is also directed to the use of a variant of albumin or a fragment thereof or fusion polypeptides comprising variant albumin or fragments thereof, or a conjugate comprising a variant of albumin or a fragment thereof, or an associate comprising a variant of albumin or a fragment thereof for the manufacture of a pharmaceutical composition, wherein the variant of albumin or a fragment thereof or fusion polypeptides comprising variant albumin or fragments thereof, or a conjugate comprising a variant of albumin or a fragment thereof, or an associate comprising a variant of albumin or a fragment thereof has an altered binding affinity to FcRn and/or an altered plasma half-life compared with HSA or the corresponding fragment thereof or fusion polypeptide comprising HSA or fragment thereof or conjugate comprising HSA.
In this connection the corresponding fragment of HSA is intended to mean a fragment of HSA that aligns with and has same number of amino acids as the fragment of the variant albumin with which it is compared. Similarly the corresponding fusion polypeptide comprising HSA or conjugate comprising HSA is intended to mean molecules having same size and amino acid sequence as the fusion polypeptide of conjugate comprising variant albumin, with which it is compared.
The composition may comprise a pharmaceutically acceptable carrier or excipient such as water, polysorbate 80 or those specified in the US Pharmacopoeia for human albumin.
Further preferences for the ninth aspect of the invention include those of the first aspect of the invention and those provided below the twelfth aspect of the invention. The skilled person understands that any aspect of the invention may be combined with another aspect or aspects of the invention and/or with one or more (several) of the preferences for the aspects of the invention and/or other disclosures made herein.
Nanoparticles
A tenth aspect of the invention relates to a nanoparticle comprising a variant, fusion, conjugate, associate, nanoparticle, composition or polynucleotide as disclosed herein.
Techniques for incorporation of a molecule into nano- or microparticles are known in the art. Preferred methods for preparing nano- or microparticles that may be applied to the albumin, variant, fragment, fusion, conjugate or associate thereof according to the invention is disclosed in WO 2004/071536 or WO2008/007146 or Oner & Groves (Pharmaceutical Research, Vol 10(9), 1993, pages 1387 to 1388) which are incorporated herein by reference. Preferably the average diameter of a nano-particle is from 5 to 1000 nm, more preferably 5, 10, 20, 30, 40, 50, 80, 100, 130, 150, 200, 300, 400, 500, 600, 700, 800, 900, or 999 to 5, 10, 20, 30, 40, 50, 80, 1 00, 1 30, 1 50, 200, 300, 400, 500, 600, 700, 800, 900, or 1 000 nm. An advantage of a microparticle less than 200 nm diameter, and more particularly less than 130 nm, is that is amenable to sterilisation by filtration through a 0.2 μηη (micron) filter. Preferably, the average diameter of a micro-particle is from 1 000 nm (1 pm (micron)) to 1 00 pm (micron), more preferably from 1 , 2, 5, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100 to 1 , 2, 5, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100 m (micron).
Further preferences for the tenth aspect of the invention include those of the first aspect of the invention and those provided below the twelfth aspect of the invention. The skilled person understands that any aspect of the invention may be combined with another aspect or aspects of the invention and/or with one or more (several) of the preferences for the aspects of the invention and/or other disclosures made herein.
Uses
An eleventh aspect of the invention relates to use of a variant albumin, fragment, fusion or conjugate thereof or nanoparticle or associate thereof. Use may be, for example, in a method of treatment, prophylaxis, diagnosis or imaging. The variant albumin or fragments thereof or fusion polypeptides comprising variant albumin or fragments thereof according to the invention have the benefit that their binding affinity to FcRn and/or plasma half-life is altered compared to the parent or reference albumin or fragments thereof or fusion polypeptides comprising parent or reference albumin or fragments thereof. This has the advantage that the binding affinity to FcRn and/or plasma half-life of conjugates comprising variant albumin or a fragment thereof or fusion polypeptide comprising variant albumin or a fragment thereof, or an associate comprising variant albumin or a fragment thereof according to the invention can be selected in accordance with the particular therapeutic purpose.
In some situations, it would be advantageous to use an albumin, variant, fragment, fusion, conjugate or associate or composition thereof having a longer plasma half-life than the reference molecule or composition since this would have the benefit that the administration of the albumin, variant, fragment, fusion, conjugate or associate or composition thereof would be needed less frequently or at a reduced dose (and consequently with fewer side effects) compared to the situation where the reference molecule or composition was used. With respect to the use of a variant, fusion, conjugate, associate, nanoparticle, composition or polynucleotide the albumin moiety may comprise one more alterations as disclosed herein.
In other situations, it would be advantageous to use an albumin, variant, fragment, fusion, conjugate or associate or composition thereof having a shorter plasma half-life than the reference molecule or composition since this would have the benefit that the administration of the albumin, variant, fragment, fusion, conjugate or associate or composition thereof can be carried out at a higher dose compared to the situation where the reference molecule or composition was used with the benefit that the administered compound clears from the recipient more quickly than if the reference molecule or composition was used. With respect to the use of a variant, fusion, conjugate, associate, nanoparticle, composition or polynucleotide the albumin moiety may comprise one more alterations as disclosed herein.
For example for a conjugate, associate or fusion polypeptide used for imaging purposes in animals or human beings, where the imaging moiety has an very short half-life and a conjugate or a fusion polypeptide comprising HSA has a plasma half-life that is far longer than needed for the imaging purposes it would be advantageous to use a variant albumin or fragment thereof of the invention having a shorter plasma half-life than the parent or reference albumin or fragment thereof, to provide conjugates of fusion polypeptides having a plasma half- life that is sufficiently long for the imaging purpose but sufficiently short to be cleared form the body of the particular patient on which it is applied.
In another example for a conjugate, an associate or fusion polypeptide comprising a therapeutic compound effective to treat or alleviate a particular condition in a patient in need for such a treatment it would be advantageous to use the variant albumin or fragment thereof having a longer plasma half-life than the parent or reference albumin or fragment thereof, to provide associates or conjugates or fusion polypeptides having longer plasma half-lives which would have the benefit that the admin istration of the associate or conjugate or fusion polypeptide of the invention would be needed less frequently or at reduced dose with less side effects compared to the situation where the parent or reference albumin or associates thereof or fragment thereof was used . For example, the invention provides a method of treating a proliferative disease in an individual, comprising administering the individual an effective amount of an associate according to the invention in which the associate comprises a taxane, a taxol or taxol derivative (e.g. paclitaxel).
In a further aspect the invention relates to compositions comprising the variant albumin, associates thereof or fragment thereof, variant albumin fragment or associates thereof or fusion polypeptide comprising variant albumin or fragment thereof according to the invention. The compositions are preferably pharmaceutical compositions. The composition may be prepared using techniques known in the area such as disclosed in recognized handbooks within the pharmaceutical field. Since the albumin, variant, fragment, fusion, conjugate or associate thereof has a binding affinity to FcRn and/or plasma half-life which is modulated (i.e. stronger or weaker and/or longer or shorter) than that of a reference molecule, the composition also has a binding affinity to FcRn and/or modulated plasma half-life relative to an equivalent composition comprising the reference molecule in place of the albumin, variant, fragment, fusion, conjugate or associate thereof as described herein. The composition may be a vaccine. The polypeptide according to the invention may be an active pharmaceutical or an excipient. Optionally, the composition is provided in unit dosage form.
Preferably the albumin, variant, fragment, fusion, conjugate or associate thereof has a plasma half-life that is longer than the plasma half-life of the reference molecule e.g. the same composition except that the albu min component (e.g. albumin, variant, fragment, fusion, conjugate or associate) is wild-type albumin (e.g. HSA) or a variant, fragment, fusion, conjugate or associate.
In a particular embodiment the compositions comprise a variant albumin or a fragment thereof according to the invention and a compound comprising a pharmaceutically beneficial moiety and an albumin binding domain (ABD). According to the invention ABD means a site, moiety or domain capable of binding to circulating albumin in vivo and thereby conferring transport in the circulation of the ABD and any compound or moiety bound to said ABD. ABD's are known in the art and have been shown to bind very tight to albumin so a compound comprising an ABD bound to albumin will to a certain extent behave as a single molecule. The inventors have realized by using the variant albumin or fragment thereof according to the invention together with a compound comprising a pharmaceutically beneficial moiety and an
ABD makes it possible to alter the binding affinity to FcRn and/or plasma half-life of the compound comprising a pharmaceutically beneficial moiety and an ABD compared to the situation where said compound were injected as such in a patient having need thereof or administered in a formulation comprising natural albumin or a fragment thereof.
The variant albumin or fragments thereof, conjugates comprising variant albumin or a fragment thereof or fusion polypeptide comprising variant albumin or a fragment thereof, or an associate comprising variant albumin or a fragment thereof according to the invention may also be incorporated into nano- or microparticles using techniques well known within the art. A preferred method for preparing nano- or microparticles that may be applied to the variant albumins or fragments thereof according to the invention is disclosed in WO 2004/071536 or WO2008/007146 or Oner & Groves (Pharmaceutical Research, Vol 10(9), 1993, pages 1387 to 1388) which are incorporated herein by reference.
Further preferences for the eleventh aspect of the invention include those of the first aspect of the invention and those provided below the twelfth aspect of the invention. The skilled person understands that any aspect of the invention may be combined with another aspect or aspects of the invention and/or with one or more (several) of the preferences for the aspects of the invention and/or other disclosures made herein.
Method for altering the FcRn-binding affinity or half-life of a molecule
A twelfth aspect of the invention provides a method for altering the FcRn-binding affinity or half-life of a molecule comprising:
(a) where the molecule is a polypeptide, fusing or conjugating the molecule to a polypeptide disclosed herein or to a conjugate disclosed herein; associating the molecule to a polypeptide disclosed herein or to a conjugate disclosed herein; incorporating the molecule in a nanoparticle disclosed herein or a composition disclosed herein;
(b) where the molecule is not a polypeptide, conjugating the molecule to a polypeptide disclosed herein or to a conjugate disclosed herein; associating the molecule to a polypeptide disclosed herein or to a conjugate a disclosed herein ; incorporating the molecule in a nanoparticle disclosed herein or a composition disclosed herein.
Examples of 'molecule' include those useful in therapy, prophylaxis (including those used in vaccines either as an active pharmaceutical ingredient or as an excipient), imaging and diagnosis, such as those described herein.
Further preferences for the twelfth aspect of the invention include those of the first aspect of the invention and those provided below this twelfth aspect of the invention. The skilled person understands that any aspect of the invention may be combined with another aspect or aspects of the invention and/or with one or more (several) of the preferences for the aspects of the invention and/or other disclosures made herein.
Preferences for all aspects of the invention are provided below. The skilled person understands that any aspect of the invention may be combined with another aspect or aspects of the invention and/or with one or more (several) of the preferences for the aspects of the invention and/or other disclosures made herein.
The variant of albumin or a fragment thereof or fusion polypeptides comprising variant albumin or fragments thereof, fragment thereof, conjugate, nanoparticle, associate or composition may have a plasma half-life that is either longer or shorter, preferably longer, than the plasma half-life than a corresponding albumin or a fragment thereof or fusion polypeptides comprising albumin or fragments thereof, fragment thereof, conjugate, nanoparticle, associate or composition or a binding to FcRn that is stronger or weaker, preferably weaker. Preferably the variant of albumin or a fragment thereof or fusion polypeptides comprising variant albumin or fragments thereof, fragment thereof, conjugate, nanoparticle, associate or composition has a plasma half-life that is longer than the plasma half-life of HSA or the corresponding albumin or a fragment thereof or fusion polypeptides comprising albumin or fragments thereof, fragment thereof, conjugate, nanoparticle, associate or composition.
Alternatively, this may be expressed as the variant of albumin or a fragment thereof or fusion polypeptides comprising variant albumin or fragments thereof, fragment thereof, conjugate, nanoparticle, associate or composition having a KD to FcRn (e.g. shFcRn) that is lower than the corresponding KD for HSA to FcRn or the corresponding fragment thereof or fusion polypeptide comprising HSA or fragment thereof. Preferably, the KD for the variant of albumin or a fragment thereof or fusion polypeptides comprising variant albumin or fragments thereof, fragment thereof, conjugate, nanoparticle, associate or composition is less than 0.9X KD for HSA to FcRn, more preferred less than 0.5X KD for HSA to FcRn, more preferred less than 0.1 X KD for HSA to FcRn, even more preferred less than 0.05X KD for HSA to FcRn, even more preferred less than 0.02X KD for HSA to FcRn and most preferred less than 0.01 X KD for HSA to FcRn (where X means 'multiplied by'). The KD of the variant of albumin or a fragment thereof or fusion polypeptides comprising variant albumin or fragments thereof, fragment thereof, conjugate, nanoparticle, associate or composition may be between the KD of WT albumin (e.g. SEQ ID No. 2) for FcRn and the KD of HSA K573P (SEQ ID No. 3) for FcRn. Such KDs represent binding affinities that are higher than the binding affinity between HSA and FcRn. A higher binding affinity indicates a longer half-life, for example plasma half-life.
Alternatively, the variant of albumin or a fragment thereof or fusion polypeptides comprising variant albumin or fragments thereof, fragment thereof, conjugate, nanoparticle, associate or composition has a plasma half-life that is shorter than the plasma half-life of HSA or the corresponding fragment thereof or fusion polypeptide comprising HSA or fragment thereof.
This may be expressed as the variant of albumin or a fragment thereof or fusion polypeptides comprising variant albumin or fragments thereof, fragment thereof, conjugate, nanoparticle, associate or com position having a KD to FcRn that is higher than the corresponding KD for HSA to FcRn or the corresponding of albumin or a fragment thereof or fusion polypeptides comprising albumin or fragments thereof, fragment thereof, conjugate, nanoparticle, associate or composition. Preferably, the KD for the variant of albumin or a fragment thereof or fusion polypeptides comprising variant albumin or fragments thereof, fragment thereof, or a conjugate comprising a variant of albumin or a fragment thereof is more than 2X KD for HSA to FcRn, more preferred more than 5X KD for HSA to FcRn, more preferred more than 10X KD for HSA to FcRn, even more preferred more than 25X KD for HSA to FcRn, most preferred more than 50X KD, more than 60X, more than 70X KD, more than 80X, more than 90X or more than 100X KD for HSA to FcRn. The variant of albumin or a fragment thereof or fusion polypeptides comprising variant albumin or fragments thereof, fragment thereof, conjugate, nanoparticle, associate or composition may be a null binder to FcRn.
The variant of albumin or a fragment thereof or fusion polypeptides comprising variant albumin or fragments thereof, fragment thereof, or a conjugate or nanoparticle or associate or composition comprising a variant of albumin or a fragment thereof is preferably the variant of albumin or a fragment thereof or fusion polypeptides comprising variant albumin or fragments thereof, fragment thereof, or a conjugate or nanoparticle or associate or composition comprising a variant of albumin or a fragment thereof according to the invention. A lower binding affinity indicates a shorter half-life, for example plasma half-life.
One advantage of the invention is that it allows the half-life of albumin, a variant of albumin or a fragment thereof or fusion polypeptides comprising variant albumin or fragments thereof, fragment thereof, conjugate, nanoparticle, associate or composition to be tailored in order to achieve a binding affinity or half-life which meets the needs of the user.
When determining and/or comparing KD, one or more (and preferably all) of the following parameters may be used:
Instrument: Biacore 3000 instrument (GE Healthcare)
Flow cell: CM5 sensor chip
FcRn: human FcRn, preferably soluble human FcRn, optionally coupled to a tag such as
GST or H is, most preferably His such as 6 histidines at the C-terminus of the beta-2- microglobulin (SEQ ID NO: 31 ).
Quantity of FcRn: 1200-2500 RU
Coupling chemistry: amine coupling chemistry (e.g. as described in the protocol provided by the manufacturer of the instrument).
Coupling method: The coupling may be performed by injecting 20 μg ml of the protein in
10 mM sodium acetate pH 5.0 (GE Healthcare). Phosphate buffer (67 mM phosphate buffer,
0.15 M NaCI, 0.005% Tween 20) at pH 5.5) may be used as running buffer and dilution buffer. Regeneration of the surfaces may be done using injections of HBS-EP buffer (0.01 M HEPES,
0.15 M NaCI, 3 mM EDTA, 0.005% surfactant P20) at pH 7.4 (Biacore AB).
Quantity of injection of test molecule (e.g. HSA or variant) 20-0.032μΜ
Flow rate of injection: constant, e.g. 30 μΙ/ml Temperature of injection: 25 °C
Data evaluation software: BIAevaluation 4.1 software (BIAcore AB).
The preferred method for determining KD is provided in Example 2. The invention discloses that two or more positions selected among the group consisting of positions 492, 550, 573, 574 and 580 in SEQ ID NO: 2 (and therefore equivalent positions in albumins and fragments from human serum and albumin and non-human serum albumins) may be altered in order to modulate (increase of decrease) the binding affinity and/or half-life e.g. plasma half-life of an albu min , fragment, fusion , conjugate, associate, nanoparticle or composition. An alteration may be a substitution, insertion or deletion. Substitution is preferred.
A substitution or insertion may or may not comprise introduction of a conserved amino acid, i.e. conserved in relation to the amino acid at the position of interest. Examples of conserved amino acids are shown by the groups of Fig. 3: aliphatic, aromatic, hydrophobic, charged, polar, positive, tiny and small. At position 492 of SEQ ID NO: 2 (or equivalent position of other albumins or variants or fragments thereof), it is preferred that the alteration is a substitution, such as from the native amino acid to A, C, D, E, F, G, H, I , K, L, M, N, P, Q, R, S, T, V, W, Y, more preferred to G, D, F, H , M or R, even more preferred to G or D and most preferred to G. I n SEQ I D NO: 2 the native amino acid at position 492 is E, therefore a substitution to E is not preferred.
At position 550 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof), it is preferred that the alteration is a substitution, such as from the native amino acid to A, C, D, E, F, G, H, I, K, L, M, N, P, Q, R, S, T, V, W, Y, more preferred to K, L, M, E or R, even more preferred to K, L or M and most preferred to K. In SEQ ID NO: 2 the native amino acid at position 550 is D, therefore a substitution to D is not preferred.
At position 573 of SEQ ID NO: 2 (or equivalent position of other albumins or variants or fragments thereof), it is preferred that the alteration is a substitution, such as from the native amino acid to A, C, D, E, F, G, H, I, K, L, M, N, P, Q, R, S, T, V, W, Y, more preferred to P, Y, W, H, F, T, I or V, even more preferred to P, Y or W and most preferred to P. In SEQ ID NO: 2 the native amino acid at position 573 is K, therefore a substitution to K is not preferred.
At position 574 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof), it is preferred that the alteration is a substitution, such as from the native amino acid to A, C, D, E, F, G, H, I, K, L, M, N, P, Q, R, S, T, V, W, Y, more preferred to D, F, G, H, N, S or Y, even more preferred to H, D, F or G and most preferred to H. In SEQ ID NO: 2 the native amino acid at position 574 is K, therefore a substitution to K is not preferred.
At position 580 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof), it is preferred that the alteration is a substitution, such as from the native amino acid to A, C, D, E, F, G, H, I, K, L, M, N, P, Q, R, S, T, V, W, Y, more preferred to K or R, most preferred to K. In SEQ I D NO: 2 the native amino acid at position 580 is Q, therefore a substitution to Q is not preferred.
A variant albumin may comprise alterations at positions corresponding to positions 492+550 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492D+550K (e.g. SEQ I D NO: 231 ), or 492G+550K (e.g. SEQ ID NO: 240).
A variant albumin may comprise alterations at positions corresponding to positions 492+573 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492F+573P (e.g. SEQ I D NO: 109), 492G+573P (e.g. SEQ ID NO: 1 10), 492H+573P (e.g. SEQ ID NO: 1 1 1 ) or 492R+573P (e.g. SEQ I D NO: 1 13) or more preferably 492D+573P (e.g. SEQ ID NO: 108). However, it is preferred that the variant does not consist of SEQ ID NO: 2 with only alterations 492G+K573A, E492G+K573A, E492G+N503K+K573A, E492G+N503H + K573A, E492G+K573P, E492G+N503K+K573P or E492G+N503H+K573P.
A variant albumin may comprise alterations at positions corresponding to positions 492+574 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492D+574H (e.g. SEQ I D NO: 232), 492G+574H (e.g. SEQ ID NO: 241 ) or 492D+574H (e.g. SEQ ID NO: 232).
A variant albumin may comprise alterations at positions corresponding to positions 492+580 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof).
A variant albumin may comprise alterations at positions corresponding to positions 550+573 of SEQ ID NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise D550K+K573P (e.g. SEQ ID NO: 1 17).
A variant albumin may comprise alterations at positions corresponding to positions 550+574 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 550K+574H (e.g. SEQ ID NO: 130), 550M+574H
(e.g. SEQ ID NO: 249), 550M+574H (e.g. SEQ ID NO: 249) or 550L+574H (e.g . S EQ I D NO: 245).
A variant albumin may comprise alterations at positions corresponding to positions 550+580 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 550K+580K (e.g. SEQ ID NO: 131 ), 550M+580K (e.g. SEQ ID NO: 251 ) or 550L+580K (e.g. SEQ ID NO: 247).
A variant albumin may comprise alterations at positions corresponding to positions 573+574 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 574D+573P (e.g. SEQ ID NO: 121 ), 574F+573P (e.g. SEQ ID NO: 122), 574G+573P (e.g. SEQ ID NO: 123), 574H+573P (e.g . S EQ I D NO: 124), 574N+573P (e.g. SEQ ID NO: 125) or 574S+573P (e . g . S E Q I D N O : 126). It is preferred that the variant does not consist of SEQ I D NO: 2 with only alterations K573P+K574N+A577T+A578R+S579C+Q580K+A581 D+G584A.
A variant albumin may comprise alterations at positions corresponding to positions 573+580 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 580K+573P ( e . g . S E Q I D N O : 128) or 580R+573P (e.g. SEQ ID NO: 129). However, it is preferred that the variant does not consist of SEQ ID NO: 2 with only alterations K573P+A577E+A578S+Q580K+A582T.
A variant albumin may comprise alterations at positions corresponding to positions 574+580 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof).
A variant albumin may comprise alterations at positions corresponding to positions 492+550+573 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492G+550K+573P (e.g. SEQ ID NO: 254) or 492D+550K+573P (e.g. SEQ ID NO: 253).
A variant albumin may comprise alterations at positions corresponding to positions 492+550+574 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise E492G+D550K+K574H (e.g. SEQ ID NO: 255).
A variant albumin may comprise alterations at positions corresponding to positions
492+550+580 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492D+550K+580K (e.g. SEQ ID NO: 258) or 492G+550K+580K (e.g. SEQ ID NO: 259).
A variant albumin may comprise alterations at positions corresponding to positions 492+573+574 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492D+573P+574H (e . g . S E Q I D N O : 233).
A variant albumin may comprise alterations at positions corresponding to positions 492+573+580 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492D+573P+580K (e.g. SEQ ID NO: 234) or 492G+573P+580K (e.g. SEQ ID NO: 242).
A variant albumin may comprise alterations at positions corresponding to positions 492+574+580 of SEQ I D NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492D+574H+580K (SEQ I D NO: 262) or 492G+574H+580K (e.g. SEQ ID NO: 263).
A variant albumin may comprise alterations at positions corresponding to positions 550+573+574 of S EQ I D NO : 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 550K+574H+573P (e.g. SEQ ID NO: 131 ), 550L+573P+574H (e.g. SEQ ID NO: 246) or 550M+573P+574H (e.g. SEQ ID NO: 250).
A variant albumin may comprise alterations at positions corresponding to positions 550+573+580 of SEQ ID NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 550L+573P+580K (e.g. SEQ ID NO: 248) or 550M+573P+580K (e.g. SEQ ID NO: 252).
A variant albumin may comprise alterations at positions corresponding to positions 550+574+580 of SEQ ID NO: 2 (or equivalent position of other albumins or variants or fragments thereof).
A variant albumin may comprise alterations at positions corresponding to positions
573+574+580 of SEQ ID NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 574H+580K+573P (e.g. SEQ ID NO: 135).
A variant albumin may comprise alterations at positions corresponding to positions 492+550+573+574 of SEQ ID NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492G+550K+573P+574H (e.g. SEQ ID NO: 257) or492D+550K+573P+574H (e.g. SEQ ID NO: 256).
A variant albumin may comprise alterations at positions corresponding to positions 492+550+573+580 of SEQ ID NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492D+550K+573P+580K (e.g. SEQ ID NO: 260)or492G+550K+573P+580K(e.g. SEQ ID NO: 261).
A variant albumin may comprise alterations at positions corresponding to positions 492+550+574+580 of SEQ ID NO: 2 (or equivalent position of other albumins or variants or fragments thereof).
A variant albumin may comprise alterations at positions corresponding to positions
492+573+574+580 of SEQ ID NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492D+573P+574H+580K (e.g. SEQ ID NO: 114)or492G+573P+574H+580K (e.g. SEQ ID NO: 115), 492F+573P+574G+580K (e.g. SEQ ID NO: 238), 492G+573P+574G+580K (e.g. SEQ I D NO : 234), 492D+573P+574G+580K (e.g. SEQ ID NO: 235), 492F+573P+574H+580R (e.g. SEQ ID NO: 239), 492D+573P+574H+580K (e.g. SEQ ID NO: 264), 492G+573P+574H+580R (e.g. SEQ ID NO: 244), 492D+573P+574H+580R (e.g. SEQ ID NO: 236) or 492F+573P+574H+580K (e.g. SEQ ID NO: 237).
A variant albumin may comprise alterations at positions corresponding to positions 550+573+574+580 of SEQ ID NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 550K+573P+574H+580K (e.g. SEQ ID NO: 265). A variant albumin may comprise alterations at positions corresponding to positions 492+550+573+574+580 of SEQ ID NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492D+550K+573P+574H+580K (e.g. SEQ I D NO: 266) or 492G+550K+573P+574H+580K (e.g. SEQ I D NO: 267). Such alterations may comprise 492D+550K+573P+574H (e.g. SEQ ID NO: 256).
Particularly preferred variants include:
a variant albumin comprising alterations at positions corresponding to positions 492 and 580 in SEQ ID NO: 2, such as (i) E492G and Q580K, or (ii) E492D and Q580K (or equivalent positions of other albumins or variants or fragment thereof);
a variant albumin comprising alterations at positions corresponding to positions 492 and 574 in SEQ I D NO: 2, such as (i) E492G and K574H, (ii) E492D and K574H , (iii) E492D and K574K, or (iv) E492G and K574K (or equivalent positions of other albumins or variants or fragment thereof);
a variant albumin comprising alterations at positions corresponding to positions 492 and
550 in SEQ ID NO: 2, such as (i) E492G and D550K, or (ii) E492D and D550K (or equivalent positions of other albumins or variants or fragment thereof);
a variant albumin comprising alterations at positions corresponding to positions 550 and
573 in SEQ ID NO: 2, such as (i) D550K and K573P, (ii) D550L and K573P or (iii) D550M and K573P (or equivalent positions of other albumins or variants or fragment thereof);
a variant albumin comprising alterations at positions corresponding to positions 550 and
574 in SEQ I D NO: 2, such as (i) D550K and K574H, or (ii) D550L and K574H (or equivalent positions of other albumins or variants or fragment thereof);
a variant albumin comprising alterations at positions corresponding to positions 550 and 580 in SEQ I D NO: 2, such as (i) D550M and Q580K, (ii) D550L and Q580K or (iii) D550K and Q580K (or equivalent positions of other albumins or variants or fragment thereof);
a variant albu m i n with alterations (e.g. comprising alterations) at positions corresponding to positions 580 and 573 in SEQ I D NO: 2, such as Q580K and K573P (or equivalent positions of other albumins or variants or fragment thereof) and preferably one or more (several) other alterations such at a position selected from 492, 550 and 574. If there is a K at position 580 and a P at position 573 it is preferred that the polypeptide comprises an additional alteration at a position selected from the group consisting of 492, 550, and 574.
a variant albumin with alterations (e.g. comprising alterations) at positions corresponding to positions 492 and 573 in SEQ I D NO: 2, such as a variant albumin comprising alterations at positions corresponding to positions 492 and 573 in SEQ I D NO: 2, such as (i) E492D and K573P or (ii) E492G and K573P or (iii) E492G and K573A (or equivalent positions of other albumins or variants or fragment thereof), and preferably one or more (several) other alterations such at a position selected from 550, 574 and 580. If there is a G at position 492 and an A or P at position 573 it is preferred that the polypeptide comprises an additional alteration at a position selected from the group consisting of 550,574 and 580 (or equivalent positions of other albumins or variants or fragment thereof);
a variant albumin with alterations (e.g. comprising alterations) at positions corresponding to positions 573 and 574 in SEQ ID NO: 2, such as K573P and K574H (or equivalent positions of other albumins or variants or fragment thereof, and preferably one or more (several) alterations such as at a position selected from 492, 550 and 580. If there is a P at position 573 and an N at position 574 it is preferred that the polypeptide comprises an additional alteration at a position selected from the group consisting of 492, 550, and 580 (or equivalent positions of other albumins or variants or fragment thereof);
a variant albumin comprising alterations at positions corresponding to positions 574 and 580 in SEQ ID NO: 2, such as 574H and 580K (or equivalent positions of other albumins or variants or fragment thereof. If there is a N at position 574 and a K at position 580 it is preferred that the polypeptide comprises an additional alteration at a position selected from the group consisting of 492, 550, and 573 (or equivalent positions of other albumins or variants or fragment thereof);
a variant albumin comprising alterations at positions corresponding to positions 492, 550 and 573 in SEQ ID NO: 2, such as E492G, D550K and K573P e.g. SEQ ID NO: 254 or such as E492D, D550K and K573P e.g. SEQ ID NO: 253;
a variant albumin comprising alterations at positions corresponding to positions 550, 573 and 580 in SEQ ID NO: 2, such as D550K, K573P and Q580K e.g. SEQ ID NO: 133;
a variant albumin comprising alterations at positions corresponding to positions 550, 573 and 580 in SEQ ID NO: 2, such as D550L, K573P and Q580K e.g. SEQ ID NO: 248 or such as D550M, K573P and Q580K e.g. SEQ ID NO:252;
a variant albumin comprising alterations at positions corresponding to positions 550, 573 and 574 in SEQ ID NO: 2, such as D550L, K573P and K574H e.g. SEQ ID NO:246;
a variant albumin comprising alterations at positions corresponding to positions 550, 573 and 574 in SEQ ID NO: 2, such as D550K, K573P and K574H e.g. SEQ ID NO: 131 ;
a variant albumin comprising alterations at positions corresponding to positions 492, 573 and 580 in SEQ ID NO: 2, such as E492G, K573P and Q580K e.g. SEQ ID NO: 242;
a variant albumin comprising alterations at positions corresponding to positions 492, 573 and 580 in SEQ ID NO: 2, such as E492D, K573P and Q580K e.g. SEQ ID NO: 234;
a variant albumin comprising alterations at positions corresponding to positions 492, 550, 573 and 574 in SEQ I D NO: 2, such as E492D, D550K, K573P and K574H e.g. SEQ ID NO: 256;
a variant albumin comprising alterations at positions corresponding to positions 492, 550, 573 and 574 in SEQ I D NO: 2, such as E492G, D550K, K573P and K574H e.g. SEQ I D NO: 257; a variant albumin comprising alterations at positions corresponding to positions 573, 574 and 580 in SEQ ID NO: 2, such as K573P, K574H and Q580K e.g. SEQ ID NO: 135;
a variant albumin comprising alterations at positions corresponding to positions 492, 573, 574 and 580 in SEQ I D NO: 2, such as E492D, K573P, K574H and Q580K (or equivalent positions of other albumins or variants or fragment thereof), e.g. SEQ ID NO: 1 14;
a variant albumin comprising alterations at positions corresponding to positions 492, 573, 574 and 580 in SEQ ID NO: 2, such as E492G, K573P, K574H and Q580K (or equivalent positions of other albumins or variants or fragment thereof), e.g. SEQ ID NO: 1 15.
It is preferred that the alteration at position 492 is conserved relative to D or E. It is preferred that the alteration at position 574 is conserved relative to H. It is preferred that the alteration at position 580 is conserved relative to K.
Advantageously, the polypeptide retains substantially the same tertiary structure (or, for a fragment, the relevant part of the structure) as a reference or parent albumin such as HSA. The skilled person understand the term 'substantially the same tertiary structure' bearing in mind that some degree of variation in tertiary structure is expected as all proteins have some degree of structural flexibility. This applies particularly to polypeptides having a higher binding affinity to FcRn than the parent or reference albumin (e.g. HSA) has to FcRn.
One or more (several) of the His residues may or may not be maintained relative to the parent albumin. For example, with reference to SEQ I D NO: 2, one or more (several) of the following His residues may be maintained: 3, 9, 39, 67, 105, 128, 146, 242, 247, 288, 338, 367, 440, 464, 510, 535. One or more (several), preferably all, of the His residues in domain I are maintained {i.e. 3, 9, 39, 67, 105, 128, 146.). One or more (several), preferably all, of the His residues in domain I I are maintained {i.e. 242, 247, 288, 338, 367). One or more (several), preferably all, of the His residues in domain I II are maintained {i.e. 440, 464, 510, 535). One or more (several) or all three of His 464, 510, 535 may be maintained.
It is preferred that at least 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16 or 17 of the disulphide bonds of the albumin are maintained in the polypeptide. For a polypeptide derived from a full length albumin, it is preferred that all disulphide bonds usually present in that albumin are maintained. For a polypeptide derived from a fragment of albumin, it is preferred that all disulphide bonds usually present in that fragment are maintained. It is preferred that Cys34 (or equivalent in non-human albumins) is maintained.
For all aspects of the invention fusion partner polypeptides and/or conjugates may comprise one or more (several) of: 4-1 BB ligand, 5-helix, A human C-C chemokine, A human
L105 chemokine, A human L105 chemokine designated huL105_3., A monokine induced by gamma-interferon (M I G), A partial CXCR4B protein , A platelet basic protein (PBP), a1 - antitrypsin , ACRP-30 Homologue; Complement Com ponent C 1 q C, Adenoid-expressed chemokine (ADEC), aFGF; FGF-1 , AGF, AGF Protein, albumin, an etoposide, angiostatin, Anthrax vaccine, Antibodies specific for collapsin, antistasin, Anti-TGF beta family antibodies, antithrombin I I I , APM-1 ; ACRP-30; Famoxin, apo-lipoprotein species, Arylsulfatase B, b57 Protein, BCMA, Beta-thromboglobulin protein (beta-TG), bFGF; FGF2, Blood coagulation factors, BMP Processing Enzyme Furin, BMP-10, BMP-12, BMP-15, BMP-17, BMP-18, BMP- 2B, BMP-4, BM P-5, BMP-6, BMP-9, Bone Morphogenic Protein-2, calcitonin, Calpain-10a, Calpain-10b, Calpain-10c, Cancer Vaccine, Carboxypeptidase, C-C chemokine, MCP2, CCR5 variant, CCR7, CCR7, CD1 1 a Mab, CD1 37; 4-1 BB Receptor Protein, CD20 Mab, CD27, CD27L, CD30, CD30 ligand, CD33 immunotoxin, CD40, CD40L, CD52 Mab, Cerebus Protein, Chemokine Eotaxin., Chemokine hlL-8, Chemokine hMCP1 , Chemokine hMCPI a, Chemokine hMCPI b, Chemokine hMCP2, Chemokine hMCP3, Chemokine hSDFI b, Chemokine MCP-4, chemokine TECK and TECK variant, Chemokine-like protein I L-8M1 Full-Length and Mature, Chemokine-like protein I L-8M10 Full-Length and Mature, Chemokine-like protein IL-8M3, Chemokine-like protein I L-8M8 Full-Length and Mature, Chemokine-like protein I L-8M9 Full- Length and Mature, Chemokine-like protein PF4-414 Full-Length and Mature, Chemokine-like protein PF4-426 Full-Length and Mature, Chemokine-like protein PF4-M2 Fu ll-Length and Mature, Cholera vaccine, Chondromodulin-like protein , c-kit ligand; SCF; Mast cell growth factor; MGF; Fibrosarcoma-derived stem cell factor, CNTF and fragment thereof (such as CNTFAx15"(Axokine™)), coagu lation factors i n both pre and active forms , col lagen s, Complement C5 Mab, Connective tissue activating protein-Ill, CTAA1 6.88 Mab, CTAP-III, CTLA4-l g , CTLA-8, CXC3, CXC3, CXCR3; CXC chemokine receptor 3, cyanovirin-N, Darbepoetin, designated exodus, designated huL105_7., DIL-40, DNase, EDAR, EGF Receptor Mab, ENA-78, Endostatin, Eotaxin, Epithelial neutrophil activating protein-78, EPO receptor; EPOR, erythropoietin (EPO) and EPO mimics, Eutropin, Exodus protein, Factor IX, Factor VII, Factor VIII, Factor X and Factor XIII , FAS Ligand Inhibitory Protein (DcR3), FasL, FasL, FasL, FGF, FGF-12; Fibroblast growth factor homologous factor-1 , FGF-15, FGF-16, FGF-18, FGF-3; INT-2, FGF-4; gelonin, HST-1 ; HBGF-4, FGF-5, FGF-6; Heparin binding secreted transforming factor-2, FGF-8, FGF-9; Glia activating factor, fibrinogen, flt-1 , flt-3 ligand, Follicle stimulating hormone Alpha subunit, Follicle stimulating hormone Beta subunit, Follitropin, Fractalkine, fragment, myofibrillar protein Troponin I, FSH, Galactosidase, Galectin-4, G-CSF, GDF-1 , Gene therapy, Glioma-derived growth factor, glucagon, glucagon-like peptides, Glucocerebrosidase, glucose oxidase, Glucosidase, Glycodelin-A; Progesterone-associated endometrial protein, GM- CSF, gonadotropin, Granulocyte chemotactic protein-2 (GC P-2), Granulocyte-macrophage colony stimulating factor, growth hormone, Growth related oncogene-alpha (GRO-alpha), Growth related oncogene-beta (GRO-beta), Growth related oncogene-gamma (GRO-gamma), hAPO-4; TROY, hCG, Hepatitus B surface Antigen, Hepatitus B Vaccine, HER2 Receptor Mab, hirudin, HIV gp120, HIV gp41 , HIV Inhibitor Peptide, HIV Inhibitor Peptide, HIV Inhibitor Peptide, H IV protease inhibiting peptides, HIV-1 protease inhibitors, H PV vaccine, Human 6CKine protein, Human Act-2 protein, Human adipogenesis inhibitory factor, human B cell stimulating factor-2 receptor, Human beta-chemokine H 1305 (MCP-2), Human C-C chemokine DGWCC, Human CC chemokine ELC protein, Human CC type chemokine interleukin C, Human CCC3 protein, Human CCF1 8 chemokine, Human CC-type chemokine protein designated SLC (secondary lymphoid chemokine), Human chemokine beta-8 short forms, Human chemokine C10, Human chemokine CC-2, Human chemokine CC-3, Human chemokine CCR-2, Human chemokine Ckbeta-7 , H u m a n ch emokine ENA-78, Human chemokine eotaxin, Human chemokine GRO alpha, Human chemokine GROalpha, Human chemokine GRObeta, Human chemokine HCC-1 , Human chemokine HCC-1 , Human chemokine I-309, Human chemokine IP- 10, Human chemokine L105_3, Human chemokine L105_7, Human chemokine MIG, Human chemokine MIG-beta protein, Human chemokine MI P-1 alpha, Human chemokine Ml Pl beta, Human chemokine MIP-3alpha, Human chemokine MIP-3beta, Human chemokine PF4, Human chemokine protein 331 D5, Human chemokine protein 61 1 64, Human chemokine receptor CXCR3, Human chemokine SDFI alpha, Human chemokine SDFI beta, Human chemokine ZSIG-35, Human Chr19Kine protein, Human CKbeta-9, Human CKbeta-9, Human CX3C 1 1 1 amino acid chemokine, Human DNAX interleukin-40, Human DVic-1 C-C chemokine, Human EDI RF I protein sequence, Human EDI RF I I protein sequence, Human eosinocyte CC type chemokine eotaxin, Human eosinophil-expressed chemokine (EEC), Human fast twitch skeletal muscle troponin C, Human fast twitch skeletal muscle troponin I , Human fast twitch skeletal muscle Troponin subunit C, Human fast twitch skeletal muscle Troponin subunit I Protein, Human fast twitch skeletal muscle Troponin subunit T, Human fast twitch skeletal muscle troponin T, Human foetal spleen expressed chemokine, FSEC, Human GM-CSF receptor, Human gro-alpha chemokine, Human gro-beta chemokine, Human gro-gamma chemokine, H uman I L-16 protein , Hu man I L-1 RD10 protein sequence, Human IL-1 RD9, Human IL-5 receptor alpha chain, Human I L-6 receptor, Human I L-8 receptor protein hlL8RA, Human I L-8 receptor protein hlL8RB, Human IL-9 receptor protein, Human IL-9 receptor protein variant #3, Human IL-9 receptor protein variant fragment, Human IL-9 receptor protein variant fragment#3, Human interleukin 1 delta, Human Interleukin 10, Human Interleukin 10, Human interleukin 18, Human interleukin 18 derivatives, Human interleukin-1 beta precursor, Human interleukin-1 beta precursor., Human interleukin-1 receptor accessory protein , Human interleukin-1 receptor antagonist beta, Human interleukin-1 type-3 receptor, Human Interleukin-10 (precursor), Human Interleukin-10 (precursor), Human interleukin-1 1 receptor, Human interleukin-12 40 kD subunit, Human interleukin-1 2 beta-1 receptor, Human interleukin-12 beta-2 receptor, Human Interleukin-12 p35 protein, Human Interleukin-12 p40 protein, Human interleukin-12 receptor, Human interleukin-13 alpha receptor, Human interleukin-13 beta receptor, Human interleukin- 15, Human interleukin-1 5 receptor from clone P 1 , Human interleukin-17 receptor, Human interleukin-1 8 protein (I L-18), Human interleukin-3, human interleukin-3 receptor, Human interleukin-3 variant, Human interleukin-4 receptor, Human interleukin-5, Human interleukin-6, Human interleukin-7, Human interleukin-7., Human interleukin-8 (IL-8), Human intracellular IL-1 receptor antagonist, Human IP-10 and HIV-1 gp120 hypervariable region fusion protein, Human IP-10 and human Muc-1 core epitope (VNT) fusion protein, human liver and activation regulated chemokine (LARC), Human Lkn-1 Full-Length and Mature protein, Human mammary associated chemokine (MACK) protein Full-Length and Mature, Human mature chemokine Ckbeta-7, Human mature gro-alpha, Human mature gro-gamma polypeptide used to treat sepsis, Human MCP-3 and human Muc-1 core epitope (VNT) fusion protein, Human MH O protein, Human MM A protein, Human monocyte chemoattractant factor hMCP-1 , Human monocyte chemoattractant factor h MCP-3, Human monocyte chemotactic proprotein (MCPP) sequence, Human neurotactin chemokine like domain, Human non-ELR CXC chemokine H 174, Human non-ELR CXC chemokine IP10, Human non-ELR CXC chemokine Mig, Human PAI-1 mutants, Human protein with I L-1 6 activity, H uman protein with I L-16 activity, Human secondary lymphoid chemoki ne (S LC), H u man S I SD protein , H u man STC P-1 , Human stromal cell-derived chemokine, SDF-1 , Human T cell mixed lymphocyte reaction expressed chemokine (TMEC), Human thymus and activation regulated cytokine (TARC), Human thymus expressed, Human TNF-alpha, H uman TN F-alpha, H uman TN F-beta (LT-alpha), Human type CC chemokine eotaxin 3 protein sequence, Human type II interleukin-1 receptor, Human wild-type interleukin-4 (hlL-4) protein, Human ZCHEMO-8 protein, Humanized Anti-VEGF Antibodies, and fragments thereof, Humanized Anti-VEGF Antibodies, and fragments thereof, Hyaluronidase, ICE 10 kD subunit., ICE 20 kD subunit, ICE 22 kD subunit, lduronate-2-sulfatase, Iduronidase, IL-1 alpha, IL-1 beta, I L-1 inhibitor (IL-1 i)., IL-1 mature, IL-10 receptor, IL- 1 , IL-1 1 , IL-12 p40 subunit., IL- 13, IL-14, IL-15, IL-15 receptor, IL-1 , IL-17 receptor, 11-17 receptor, 11-17 receptor, IL-19, IL-1 i fragments, IL1 -receptor antagonist, I L-21 (TI F), I L-3 containing fusion protein. , I L-3 mutant proteins, IL-3 variants, IL-3 variants, IL-4, I L-4 mutein, IL-4 mutein Y124G, IL-4 mutein Y124X, IL-4 muteins, II-5 receptor, I L-6, I I-6 receptor, I L-7 receptor clone, I L-8 receptor, I L-9 mature protein variant (Met1 17 version), immunoglobulins or immunoglobulin-based molecules or fragment of either (e.g. a Small Modular ImmunoPharmaceutical™ ("SMIP") or dAb, Fab' fragments, F(ab')2, scAb, scFv or scFv fragment), including but not limited to plasminogen, Influenza Vaccine, Inhibin alpha, Inhibin beta, insulin, insulin-like growth factor, Integrin Mab, inter-alpha trypsin inhibitor, inter-alpha trypsin inhibitor, Interferon gamma-inducible protein (IP- 10), interferons (such as interferon alpha species and sub-species, interferon beta species and sub-species, interferon gamma species and sub-species), interferons (such as interferon alpha species and sub-species, interferon beta species and sub-species, interferon gamma species and su b-species), Interleukin 6, Interleukin 8 (IL-8) receptor, Interleukin 8 receptor B, lnterleukin-1 alpha, lnterleukin-2 receptor associated protein p43, interleukin-3, interleukin-4 muteins, lnterleukin-8 (I L-8) protein., interleukin-9, lnterleukin-9 (I L-9) mature protein (Thr1 17 version), interleukins (such as IL10, I L1 1 and I L2), interleukins (such as IL10, I L1 1 and I L2),
Japanese encephalitis vaccine, Kalikrein Inhibitor, Keratinocyte growth factor, Kunitz domain protein (such as aprotinin, amyloid precursor protein and those described in WO 03/066824, with or without albumin fusions), Kunitz domain protein (such as aprotinin, amyloid precursor protein and those described in WO 03/066824, with or without albumin fusions), LACI , lactoferrin, Latent TGF-beta binding protein I I, leptin, Liver expressed chemokine-1 (LVEC-1 ), Liver expressed chemokine-2 (LVEC-2), LT-alpha, LT-beta, Luteinization Hormone, Lyme Vaccine, Lymphotactin, Macrophage derived chemokine analogue MDC (n+1 ), Macrophage derived chemokine analogue MDC-eyfy, Macrophage derived chemokine analogue MDC-yl, Macrophage derived chemokine, MDC, Macrophage-derived chemokine (MDC), Maspin ; Protease Inhibitor 5, MCP-1 receptor, MCP-1 a, MCP-1 b, MCP-3, MCP-4 receptor, M-CSF, Melanoma inhibiting protein, Membrane-bound proteins, Met1 17 human interleukin 9, MI P-3 alpha, MIP-3 beta, MIP-Gamma, MIRAP, Modified Rantes, monoclonal antibody, MP52, Mutant Interleukin 6 S1 76R, myofibrillar contractile protein Troponin I , Natriuretic Peptide, Nerve Growth Factor-beta, Nerve Growth Factor-beta2, Neuropilin-1 , Neuropilin-2, Neurotactin, Neurotrophin-3 , N eu rotroph i n-4, Neurotrophin-4a, Neurotrophin-4b, Neurotrophin-4c, Neurotrophin-4d , Neutrophil activating peptide-2 (NAP-2), NOGO-66 Receptor, NOGO-A, NOGO-B, NOGO-C, Novel beta-chemokine designated PTEC, N-terminal modified chemokine GroHEK/hSDF-1 al pha , N-terminal modified chemokine GroHEK/hSDF-1 beta., N-terminal modified chemokine met-hSDF-1 alpha , N-terminal modified chemokine met-hSDF-1 beta, OPGL, Osteogenic Protein-1 ; O P-1 ; BMP-7, Osteogenic Protein-2, OX40; ACT-4, OX40L, Oxytocin (Neurophysin I), parathyroid hormone, Patched, Patched-2, PDGF-D, Pertussis toxoid, Pituitary expressed chemokine (PGEC), Placental Growth Factor, Placental Growth Factor-2, Plasminogen Activator lnhibitor-1 ; PAI-1 , Plasminogen Activator lnhibitor-2; PAI-2, Plasminogen Activator lnhibitor-2; PAI-2, Platelet derived growth factor, Platelet derived growth factor Bv-sis, Platelet derived growth factor precursor A, Platelet derived growth factor precursor B, Platelet Mab, platelet-derived endothelial cell growth factor (PD-ECGF), Platelet-Derived Growth Factor A chain , Platelet-Derived Growth Factor B chain, polypeptide used to treat sepsis, Preproapolipoprotein "milano" variant, Preproapolipoprotein "paris" variant, pre-thrombin, Primate CC chemokine "I LI NCK", Primate CXC chemokine "I BICK", proinsulin, Prolactin, Prolactin2, prosaptide, Protease inhibitor peptides, Protei n C , Protei n S , pro-thrombin, prourokinase, RANTES, RANTES 8-68, RANTES 9-68, RANTES peptide, RANTES receptor, Recombinant interleukin-16, Resistin, restrictocin, Retroviral protease inhibitors, ricin, Rotavirus Vaccine, RSV Mab, saporin, sarcin, Secreted and Transmembrane polypeptides, Secreted and Transmembrane polypeptides, serum cholinesterase, serum protein (such as a blood clotting factor), Soluble BMP Receptor Kinase Protein-3, Soluble VEGF Receptor, Stem Cell Inhibitory Factor, Straphylococcus Vaccine, Stromal Derived Factor-1 alpha, Stromal Derived Factor-1 beta, Substance P (tachykinin), T1249 peptide, T20 peptide, T4 Endonuclease, TACI , Tare, TGF-b e t a 1 , T G F-beta 2, Thr1 17 human interleukin 9, thrombin, thrombopoietin, Thrombopoietin derivativel , Th rombopoietin derivative2 , Th rombopoietin derivatives , Thrombopoietin derivative^ Th rombopoietin derivatives, Thrombopoietin derivative6, Thrombopoietin derivative/, Thymus expressed chemokine (TECK), Thyroid stimulating Hormone, tick anticoagulant peptide, Tim-1 protein, TNF-alpha precursor, TNF-R, TNF-RII; TNF p75 Receptor; Death Receptor, tPA, transferrin, transforming growth factor beta, Troponin peptides, Truncated monocyte chemotactic protein 2 (6-76), Truncated monocyte chemotactic protein 2 (6-76), Truncated RANTES protein (3-68), tumour necrosis factor, Urate Oxidase, urokinase, Vasopressin (Neurophysin II), VEGF R-3; flt-4, VEGF Receptor; KDR; flk-1 , VEGF- 1 10, VEGF-121 , VEGF-138, VEGF-145, VEGF-162, VEGF-165, VEGF-182, VEGF-189, VEGF- 206, VEGF-D, VEGF-E; VEGF-X, von Willebrand's factor, Wild type monocyte chemotactic protein 2, Wild type monocyte chemotactic protein 2, ZTGF-beta 9, alternative antibody scaffolds e.g. anticalin(s), adnectin(s), fibrinogen fragment(s), nanobodies such as camelid nanobodies, infestin, and/or any of the molecules mentioned in WO01/79271 (particularly page 9 and/or Table 1 ), WO 2003/59934 (particularly Table 1 ), WO03/060071 (particularly Table 1 ) or WO01/079480 (particularly Table 1 ) (each incorporated herein by reference in their entirety).
Furthermore, conjugates may comprise one or more (several) of chemotherapy drugs such as: 13-cis-Retinoic Acid, 2-CdA, 2-Chlorodeoxyadenosine, 5-Azacitidine, 5-Fluorouracil, 5- FU , 6-Mercaptopurine, 6-MP, 6-TG , 6-Thioguanine, A, Abraxane, Accutane®, Actinomycin-D, Adriamycin®, Adrucil®, Agrylin®, Ala-Cort®, Aldesleukin, Alemtuzumab, ALIMTA, Alitretinoin, Alkaban-AQ®, Alkeran®, Al l-transretinoic Acid, Alpha Interferon, Altretamine, Amethopterin, Amifostine, Aminoglutethimide, Anagrelide, Anandron®, Anastrozole, Arabinosylcytosine, Ara-C, Aranesp®, Aredia®, Arimidex®, Aromasin®, Arranon®, Arsenic Trioxide, Asparaginase, ATRA, Avastin®, Azacitidine, BCG, BCN U , Bevacizumab, Bexarotene, BEXXAR®, Bicalutamide, BiCNU, Blenoxane®, Bleomycin, Bortezomib, Busulfan, Busulfex®, C225, Calcium Leucovorin, Campath®, Camptosar®, Camptothecin-1 1 , Capecitabine, Carac™, Carboplatin, Carmustine, Carmustine Wafer, Casodex®, C C-5013, CCNU, CDDP, CeeNU, Cerubidine®, Cetuximab, Chlorambucil, Cisplatin, Citrovorum Factor, Cladribine, Cortisone, Cosmegen®, C P T-1 1 , Cyclophosphamide, Cytadren®, Cytarabine, Cytarabine Liposomal, Cytosar-U®, Cytoxan®, Dacarbazine, Dacogen, Dactinomycin, Darbepoetin Alfa, Dasatinib, Daunomycin, Daunorubicin, Daunorubicin Hydrochloride, Daunorubicin Liposomal, DaunoXome®, Decadron, Decitabine, Delta-Cortef®, Deltasone®, Denileukin diftitox, DepoCyt™, Dexamethasone, Dexamethasone acetate, Dexamethasone Sodium Phosphate, Dexasone, Dexrazoxane, DHAD, DIC, Diodex, Docetaxel, Doxil®, Doxoru bicin , Doxorubicin liposomal , Droxia™, DTI C, DTI C-Dome®, Duralone®, Efudex®, Eligard™, Ellence™, Eloxatin™, Elspar®, Emcyt®, Epirubicin, Epoetin alfa, Erbitux™, Erlotinib, Erwinia L-asparaginase, Estramustine, Ethyol, Etopophos®, Etoposide, Etoposide Phosphate, Eulexin®, Evista®, Exemestane, Fareston®, Faslodex®, Femara®, Filgrastim, Floxuridine, Fludara®, Fludarabine, Fluoroplex®, Fluorouracil, Fluorouracil (cream), Fluoxymesterone, Flutamide, Folinic Acid, FUDR®, Fulvestrant, G-CSF, Gefitinib, Gemcitabine, Gemtuzumab ozogamicin , Gemzar®, Gleevec™, Gl iadel® Wafer, G M-CSF, Goserelin, Granulocyte - Colony Stimulating Factor, Granulocyte Macrophage Colony Stimulating Factor, Halotestin®, Herceptin®, Hexadrol, Hexalen®, Hexamethylmelamine, HMM, Hycamtin®, Hydrea®, Hydrocort Acetate®, Hydrocortisone, Hydrocortisone Sodium Phosphate, Hydrocortisone Sodium Succinate, Hydrocortone Phosphate, Hydroxyurea, Ibritumomab, Ibritumomab Tiuxetan, Idamycin®, Idarubicin, Ifex®, I FN-alpha, Ifosfamide, IL-1 1 , IL-2, Imatinib mesylate, Imidazole Carboxamide, Interferon alfa, Interferon Alfa-2b (PEG Conjugate), lnterleukin-2, lnterleukin-1 1 , Intron A® (interferon alfa-2b), Iressa®, Irinotecan, Isotretinoin, Kidrolase®, Lanacort®, Lapatinib, L-asparaginase, LCR, Lenalidomide, Letrozole, Leucovorin, Leukeran, Leukine™, Leuprolide, Leurocristine, Leustatin™, Liposomal Ara-C, Liquid Pred®, Lomustine, L-PAM , L-Sarcolysin, Lupron®, Lu pron De pot®, M, Matulane®, Maxidex, Mechlorethamine, Mechlorethamine Hydrochloride, Medralone®, Med rol®, Megace®, Megestrol, Megestrol Acetate, Melphalan, Mercaptopurine, Mesna, Mesnex™, Methotrexate, Methotrexate Sodium, Methylprednisolone, Meticorten®, Mitomycin, Mitomycin-C, Mitoxantrone, M-Prednisol®, MTC, MTX, Mustargen®, Mustine, Mutamycin®, Myleran®, Mylocel™, Mylotarg®, Navelbine®, Nelarabine, Neosar®, Neulasta™, Neumega®, Neupogen®, Nexavar®, Nilandron®, Nilutamide, Nipent®, Nitrogen Mustard, Novaldex®, Novantrone®, Octreotide, Octreotide acetate, Oncospar®, Oncovin®, Ontak®, Onxal™, Oprevelkin, Orapred®, Orasone®, Oxaliplatin, a taxol or taxol derivative e.g. Paclitaxel or Paclitaxel Protein-bound, Pamidronate, Panitumumab, Panretin®, Paraplatin®, Pediapred®, P E G I nterferon , Pegaspa rgase , Pegfi lg rasti m , P E G-INTRON™, PEG-L- asparaginase, PEMETREXED, Pentostatin, Phenylalanine Mustard, Platinol®, Platinol-AQ®, Prednisolone, Prednisone, Prelone®, Procarbazine, PROCRIT®, Proleukin®, Prolifeprospan 20 with Carmustine Implant, Purinethol®, R, Raloxifene, Revlimid®, Rheumatrex®, Rituxan®, Rituximab, Roferon-A® (Interferon Alfa-2a), Rubex®, Rubidomycin hydrochloride, Sandostatin®, Sandostatin LAR®, Sargramostim, Solu-Cortef®, Solu-Medrol®, Sorafenib, SPRYCEL™, STI- 571 , Streptozocin, SU 1 1248, Sunitinib, Sutent®, Tamoxifen, Tarceva®, Targretin®, Taxol®, Taxotere®, Tem od a r®, Temozolomide, Teniposide, TESPA, Thalidomide, Thalomid®, TheraCys®, Thioguanine, Thioguanine Tabloid®, Thiophosphoamide, Thioplex®, Thiotepa, TICE®, Toposar®, Topotecan, Toremifene, Tositumomab, Trastuzumab, Tretinoin, Trexall™, Trisenox®, TS PA, TYKE RB®, VCR, Vectibix™, Velban®, Velcade®, VePesid®, Vesanoid®, Viadur™, Vidaza®, Vinblastine, Vinblastine Sulfate, Vincasar Pfs®, Vincristine, Vinorelbine, Vinorelbine tartrate, VLB, VM-26, Vorinostat, VP-16, Vumon®, Xeloda®, Zanosar®, Zevalin™, Zinecard®, Zoladex®, Zoledronic acid, Zolinza, Zometa®; radiopharmaceuticals such as: Carbon- 1 1 , Carbon-14, Chromium-51 , Cobalt-57, Cobalt-58, Erbium-169, Fluorine-18, Gallium-67, Gold- 198, l ndium-1 1 1 , l ndium-1 13m, lodine-123, lodine-125, lodine-131 , l ron-59, Krypton-81 m, Nitrogen-1 3 , Oxygen-15, Phosphorous-32, Rhenium-186, Rubidium-82, Samarium-153, Selenium-75, Strontium-89, Technetium-99m, Thallium-201 , Tritium, Xenon-127, Xenon-133, Yttrium-90; imaging agents such as Gadolinium, magnetite, manganese, technetium, 1125, 1131 , P32, TI201 , lopamidol, PET-FDG. Further fusion partners, conjugation partners and/or molecules for inclusion in a nanoparticle, associate or composition according to the invention include: acromegaly drugs e.g. somatuline, lanreotide, octreotide, Sandostatin; antithrombotics e.g. bivalirudin, Angiomax, dalteparin, Fragmin, enoxaparin, Lovenox, Drotrecogin alfa (e.g. Activated), Xigris, heparin; assisted reproductive therapy compounds e.g. choriogonadotropin, Ovidrel, follitropin, alpha/beta; enzymes e.g. hyaluronidase, Hylenex; diabetes drugs e.g. exenatide, Byetta, glucagon, insulin, liraglutide, albiglutide, GLP-1 agonists, exendin or an exendin analog; compounds useful in diagnosis e.g. protirelin, Thyrel TRH Thypinone, secretin (e.g. synthetic human), Chirhostim, thyrotropin (e.g. alpha), Thyrogen' erythropoiesis drugs e.g. Darbepoetin alfa, Aranesp, Epoetin alfa, Epogen, Eprex, drugs for the treatment of genetic defects e.g. pegademase, drugs for the treatment of growth failure e.g. Adagen, mecasermin, rinfabate, drugs for the treatment of cystic fibrosis e.g. Dornase alfa, Pulmozyme, drugs for the treatment of metaoblic disorders e.g. Agalsidase beta, Fabrazyme, alglucosidase alpha, Myozyme, Laronidase, Aldurazyme, drugs for the treatment of genital wart intralesional e.g. Interferon alfa- n3, Alferon N , drugs for the treatment of granulomatous disease e.g. Interferon gamma-1 b, Actimmune; drugs for the treatment of growth failure e.g. pegvisomant, Somavert, somatropin, Genotropin, Nutropin, Humatrope,Serostim, Protropin; drugs for the treatment of heart failure e.g. nesiritide, Natrecor; drugs for the treatment of hemophilia e.g. a coagulation factor e.g. Factor VI I I , H el ixate FS , Kogenate FS , Factor IX, BeneFIX, Factor Vi l a , Novoseven, desmopressin, Stimate, DDAVP; hemopoetic drugs e.g. Filgrastim (G-CSF), Neupogen, Oprelvekin, Neumega, Pegfilgrastim, Neulasta, Sargramostim, Leukine; drugs for the treatment of hepatitis C e.g. Interferon alfa-2a, Roferon A, Interferon alfa-2b, Intron A, Interferon alfacon-1 , Infergen, Peginterferon alfa-2a, Pegasys, Peginterferon alfa-2b, PEG-lntron; drugs for the treatment of H IV e.g. enfuvirtide, Fuzeon; Fabs e.g. Fab (antithrombin), Abciximab, ReoPro; monoclonal antibodies e.g. Daclizumab, Zenapax; antiviral monoclonal antibodies e.g. Palivizumab, Synagis; monoclonal antibodies for the treatment of asthma e.g. Omalizumab, Xolair; monoclonal antibodies for use in diagnostic imaging e.g. Arcitumomab, CEA-Scan, Capromab Pendetide, ProstaScint, Satumomab Pendetide, OncoScint CR/OV, Fabs for use in diagnostic imaging e.g. Nofetumomab, Verluma; iimmuno-supressant monoclonal antibodies e.g. Basiliximab, Simulect, Muromonab-CD3, Orthoclone OKT3; monoclonal antibodies for the treatment of malignancy e.g. Alemtuzumab, Campath, Ibritumomab tiuxetan, Zevalin, Rituximab, Rituxan, Trastuzumab, Herceptin ; monoclonal antibodies for the treatment of rheumatoid arthritis (RA) e.g. Adalimumab, Humira, Infliximab, Remicade; monoclonal a nti bod i es for u se as a rad i o-immuno-therapeutic e.g. Tositumomab and Iodine I131, Tositumomab, Bexxar; drugs for the treatment of macular degeneration e.g. pegaptanib,
Macugen; drugs for the treatment of malignancy e.g. Aldesleukin, Proleukin, lnterleukin-2,
Asparaginase, Elspar, Rasburicase, Elitek, Denileukin diftitox, Ontak, Pegaspargase, Oncaspar, goserelin, leuprolide; drugs for the treatment of multiple sclerosis (MS) e.g. Glatiramer acetate (e.g. copolymer-1 ), Copaxone, Interferon beta-1 a, Avonex, Interferon beta-1 a, Rebif, Interferon beta-1 b, Betaseron; drugs for the treatment of mucositis e.g. palifermin, Kepivance; drug for the treatment of dystonia e.g., neurotoxin, Botulinum Toxin Type A, BOTOX, BOTOX Cosmetic, Botulinum Toxin Type B, MYOBLOC; drugs for the treatment of osteoporosis e.g. teriparatide,Forteo; drugs for the treatment of psoriasis e.g. Alefacept, Amevive; drugs for the treatment of RA e.g. abatacept, Orencia, Anakinra, Kineret, Etanercept, Enbrel; thrombolytics e.g. Alteplase, Activase, rtPA, Anistreplase, Eminase, Reteplase, Retavase, Streptokinase, Streptase, Tenecteplase, TNKase, Urokinase, Abbokinase, Kinlytic; drugs for the treatment of osteoporosis e.g. calcitonin (e.g. salmon), Miacalcin, Fortical, drugs for the treatment of skin ulcers e.g. Becaplermin, Regranex, Collagenase, Santyl.
Such polypeptides and chemical compounds may be referred to as diagnostic moieties, therapeutic moieties, prophylactic moieties or beneficial moieties.
Preferably the fusion partner and/or conjugation partner is not an albumin, variant or fragment thereof.
One or more (several) therapeutic or prophylactic polypeptides may be fused to the N- terminus, the C-terminus of albumin, inserted into a loop in the albumin structure or any combination thereof. It may or it may not comprise linker sequences separating the various components of the fusion polypeptide.
Teachings relating to fusions of albumin or a fragment thereof are known in the art and the skilled person will appreciate that such teachings can also be applied to the invention. WO 2001/79271A and WO 2003/59934 (incorporated herein by reference) also contain examples of therapeutic and prophylactic polypeptides that may be fused to albumin or fragments thereof, and these examples apply also to the invention. The invention is further defined in the following embodiments:
1 . A polypeptide which is a variant of albumin, fragment thereof or fusion polypeptide comprising said variant albumin or a fragment thereof having an altered binding affinity to FcRn compared with the binding affinity of a parent albumin, reference albumin, fragment thereof or fusion polypeptide comprising said parent albumin, reference albumin or fragment or fusion thereof to FcRn, wherein the polypeptide comprises alterations at two or more positions selected from positions corresponding to positions 492, 550, 573, 574 and 580 in SEQ ID NO: 2.
2. The polypeptide according to embodiment 1 wherein the polypeptide comprises alterations at two or more positions selected from positions corresponding to positions (a) 492 and 580; (b) 492 and 574; (c) 492 and 550; (d) 550 and 573; (e) 550 and 574; (f) 550 and 580 in SEQ ID NO: 2.
3. The polypeptide according to embodiment 1 or 2 comprising alterations at positions corresponding to positions 492 and 580 of SEQ I D NO: 1 , further comprising one or more (several) alterations at positions corresponding to positions selected from the group consisting of 550, 573 and 574 of SEQ ID NO: 2.
4. The polypeptide according to embodiment 1 , 2 or 3 comprising alterations at positions corresponding to positions 492 and 574 of SEQ I D NO: 1 , further comprising one or more (several) alterations at positions corresponding to positions selected from the group consisting 550, 573 and 580 of SEQ ID NO:
5. The polypeptide according to any preceding embodiment comprising alterations at positions corresponding to positions 492 and 550 of SEQ ID NO: 1 , further comprising one or more (several) alterations at positions corresponding to positions selected from the group consisting 573 , 574 and 580of SEQ ID NO: 2.
6. The polypeptide according to any preceding embodiment comprising alterations at positions corresponding to positions 550 and 573 of SEQ ID NO: 1 , further comprising one or more (several) alterations at positions corresponding to positions selected from the group consisting 492, 574, and 580 of SEQ ID NO: 2.
7. The polypeptide according to any preceding embodiment comprising alterations at positions corresponding to positions 550 and 574 of SEQ ID NO: 1 , further comprising one or more (several) alterations at positions corresponding to positions selected from the group consisting 492, 573 and 580 of SEQ ID NO: 2.
8. The polypeptide according to any preceding embodiment comprising alterations at positions corresponding to positions 550 and 580 of SEQ I D NO: 1 , further comprising an alteration at a position corresponding to position 492, 573 and 574 of SEQ ID NO: 2.
9. The polypeptide according to any preceding embodiment in which:
(a) at a position corresponding to position 492 of SEQ I D NO: 2 there is an alteration to generate an amino acid from the group consisting of A, C, D, F, H, I , K, L, M, N, P, Q, R, S, T, V, W, Y, preferably D and at a position corresponding to position 573 of SEQ ID NO: 2 there is an alteration to generate an amino acid from the group consisting of C, D, E, F, G, H, I, L, M, N, Q, R, S, T, V, W, Y, preferably Y, W or H, or
(b) at a position corresponding to position 492 of SEQ I D NO: 2 there is an alteration to generate G and at a position corresponding to position 573 there is an alteration to generate A or P and the polypeptide comprises an additional alteration at a position selected from the group consisting of 550, 574 and 580 (or equivalent positions of other albumins or variants or fragment thereof).
10. The polypeptide according to any preceding embodiment in which:
(a) at a position corresponding to position 573 of SEQ ID NO : 2 there is an alteration to generate an amino acid from the group consisting of A, C, D, E, F, G, H, I, L, M, N, Q, R, S, T, V, W, Y, preferably Y, W or H and at a position corresponding to position 574 of SEQ ID NO: 2 there is an alteration to generate an amino acid from the group consisting of A, C, D, E, F, G, H, I , L, M, P, Q, R, S, T, V, W, Y, H , D, F, G, N , S or Y, more preferably H , D, F or G, most preferably H, or
(b) at a position corresponding to position 573 of SEQ I D NO: 2 there is a P and at a position corresponding to position 574 of SEQ ID NO: 2 there is an N and the polypeptide comprises an additional alteration at a position selected from the group consisting of 492, 550, and 580 (or equivalent positions of other albumins or variants or fragment thereof);
1 1 . The polypeptide according to any preceding embodiment in which:
(a) at a position corresponding to position 573 of SEQ I D NO: 2 there is an alteration to generate an amino acid from the group consisting of A, C, D, E, F, G, H, I, L, M, N, Q, R, S, T, V, W, Y, preferably Y, W or H and at a position corresponding to position 580 of SEQ ID NO: 2 there is an alteration to generate an amino acid from the group consisting of C, D, E, F, G, H, I, L, M, N, P, R, S, T, V, W, Y, or
(b) at a position corresponding to position 573 of SEQ I D NO: 2 there is an alteration to generate a P and at a position corresponding to position 580 of SEQ I D NO: 2 there is an alteration to generate a K and the polypeptide comprises an additional alteration at a position selected from the group consisting of 492, 550, and 574 (or equivalent positions of other albumins or variants or fragment thereof).
12. The polypeptide according to any preceding embodiment in which:
(a) at a position corresponding to position 574 of SEQ I D NO : 2 there is an alteration to generate an amino acid from the group consisting of A, C, D, E, F, G, H, I , L, M, P, Q, R, S, T, V, W, Y, H , D, F, G, N , S or Y, more preferably H , D, F or G, most preferably H and at a position corresponding to position 580 of SEQ ID NO: 2 there is an alteration to generate at a position corresponding to position to an amino acid from the group consisting of A, C, D, E, F, G, H, I, L, M, N, P, R, S, T, V, W, Y, or
(b) at a position corresponding to position 574 of SEQ I D NO: 2 there is an alteration to generate an N and at a position corresponding to position 580 of SEQ I D NO: 2 there is an alteration to generate a K and the polypeptide comprises an additional alteration at a position selected from the group consisting of 492, 550, and 573 (or equivalent positions of other albumins or variants or fragment thereof).
13. The polypeptide according to any preceding embodiment, wherein the polypeptide comprises alterations at three or more positions selected from positions corresponding to positions 492, 550, 573, 574 and 580 in SEQ ID NO: 2.
14. The polypeptide according to any preceding embodiment wherein the alteration at the position corresponding to position 492, 550, 573, 574 and/or 580 is a substitution.
15. The polypeptide of embodiment 14 wherein the substitution at the position corresponding to position 492 is to G, D, F, H, M or R.
16. The polypeptide of embodiment 14 or 15 wherein the substitution at the position corresponding to position 492 is to G or D.
17. The polypeptide of any of embodiments 13 to 15 wherein the substitution at the position corresponding to position 492 is to G.
18. The polypeptide of any of embodiments 13 to 15 wherein the substitution at the position corresponding to position 492 is to D.
19. The polypeptide of any of embodiments 13 to 18 wherein the substitution at the position corresponding to position 550 is to K, L, M E, or R.
20. The polypeptide of any of embodiments 13 to 19 wherein the substitution at the position corresponding to position 550 is to K, L or M.
21 . The polypeptide of any of embodiments 13 to 20 wherein the substitution at the position corresponding to position 550 is to K.
22. The polypeptide of any of embodiments 13 to 21 wherein the alteration at the position corresponding to position 573 is a substitution to P, Y, W, H, F, T, I or V.
23. The polypeptide of any of embodiments 13 to 22 wherein the substitution at the position corresponding to position 573 is to P, Y or W.
24. The polypeptide of any of embodiments 13 to 23 wherein the substitution at the position corresponding to position 573 is to a P.
25. The polypeptide of any of embodiments 13 to 24 wherein the alteration at the position corresponding to position 574 is a substitution to H, G, D, F, N, S or Y.
26. The polypeptide of any of embodiments 13 to 25 wherein the substitution at the position corresponding to position 574 is to D, F, G or H.
27. The polypeptide of any of embodiments 13 to 26 wherein the substitution at the position corresponding to position 574 is to H.
28. The polypeptide of any of embodiments 13 to 27 wherein the substitution at the position corresponding to position 580 is to K or R.
29. The polypeptide of any of embodiments 13 to 28 wherein the substitution at the position corresponding to position 580 is to K.
30. The polypeptide of any preceding embodiment wherein the polypeptide comprises alterations at positions corresponding to positions (i) 573 and 580; (ii) 492 and 573; or (iii) 573 and 574 of SEQ ID NO: 2.
31. The polypeptide of any preceding embodiment wherein the polypeptide comprises alterations at two or more positions corresponding to the following positions of SEQ ID NO: 2:
580K + 573P (e.g. SEQ ID NO: 128); 580R + 573P (e.g. SEQ ID NO: 129); 574D + 573P (e.g.
SEQ ID NO: 121 ); 574F + 573P (e.g. SEQ ID NO: 122); 574G + 573P (e.g. SEQ ID NO: 123);
574H + 573P (e.g. SEQ I D NO: 124); 574N + 573P (e.g. SEQ ID NO: 125); 574S + 573P (e.g.
SEQ I D NO: 126); 550K + 580K (e.g. SEQ I D NO: 132); 550K + 574H (e.g. SEQ ID NO: 130); 550K + 573P (e.g. SEQ I D NO: 1 17); 492D + 573P (e.g. SEQ ID NO: 108); 492F + 573P (e.g.
SEQ ID NO: 109); 492H + 573P (e.g. SEQ ID NO: 1 1 1 ); 492R + 573P (e.g. SEQ I D NO: 1 12);
574H + 580K (e.g. SEQ ID NO: 134); 550L + 574H (e.g. SEQ ID NO: 245); 550L + 580K (e.g.
SEQ ID NO: 247); 550M + 580K (e.g. SEQ ID NO: 251 ); 492D + 550K (e.g. SEQ ID NO: 231 ); 550M + 574H (e.g. SEQ ID NO: 249); 492D + 574H (e.g. SEQ ID NO: 232); 492G + 550K (e.g. SEQ I D NO: 240); 550M + 574H (e.g. SEQ I D NO: 249); or 492G + 574H (e.g. SEQ I D NO: 241 ).
32. The polypeptide according to any preceding embodiment wherein the polypeptide comprises three or more alterations at positions selected from the group consisting of positions corresponding to the following positions of SEQ ID NO: 2: 492, 550, 573, 574 and 580.
33. The polypeptide of embodiment 32 wherein the polypeptide comprises alterations at positions corresponding to the following positions of SEQ ID NO: 2: 574H + 580K + 573P (e.g. SEQ ID NO: 135); 550K + 574H + 573P (e.g. SEQ ID NO: 131 ); 492D + 550K + 573P (e.g. SEQ ID NO: 253); 550M + 573P + 580K (e.g. SEQ ID NO: 252); 550L + 573P + 580K (e.g. SEQ ID NO: 253); 492G + 573P + 580K (e.g. SEQ I D NO: 242); 550M + 573P + 574H (e.g. SEQ I D NO: 250); 492G + 550K + 573P (e.g. SEQ I D NO: 254); 550L + 573P + 574H (e.g. SEQ I D NO: 246); 492D + 573P + 580K (e.g. SEQ I D NO: 234); 492D + 573P + 574H (e.g. SEQ ID NO: 233); 492G + 574H + 580K (e.g. SEQ ID NO: 263); 492G + 550K + 580K (e.g. SEQ ID NO: 259); 492D + 550K + 580K (e.g. SEQ I D NO: 258); 492D + 574H + 580K (e.g. SEQ ID NO: 262); or 492G + 550K + 574H (e.g. SEQ ID NO: 255).
34. The polypeptide according to any preceding embodiment wherein the polypeptide comprises four or more alterations at positions selected from the group consisting of positions corresponding to the following positions of SEQ ID NO: 2: 492, 550, 573, 574 and 580.
35. The polypeptide of embodiment 34 wherein the polypeptide comprises alterations at positions corresponding to the following positions of SEQ I D NO: 2: 492G + 573P + 574H + 580K (e.g. SEQ ID NO: 1 15); 492D + 573P + 574H + 580K (e.g. SEQ ID NO: 1 14); 550K + 573P + 574H + 580K (e.g. SEQ ID NO: 265); 492G + 550K + 573P + 580K (e.g. SEQ I D NO: 261 ); 492F + 573P + 574H + 580K (e.g. SEQ ID NO: 237); 492G + 573P + 574G + 580K (e.g. SEQ ID NO: 243); 492D + 573P + 574H + 580R (e.g. SEQ ID NO: 236); 492G + 573P + 574H + 580R (e.g. SEQ I D NO: 244); 492D + 550K + 573P + 580K (e.g. SEQ I D NO: 260); 492D + 550K + 573P + 574H (e.g. SEQ I D NO: 256); 492F + 573P + 574H + 580R (e.g. SEQ I D NO: 239); 492D + 573P + 574G + 580K (e.g. SEQ ID NO: 235); or 492F + 573P + 574G + 580K (e.g. SEQ ID NO: 238);
36. The polypeptide according to any preceding embodiment wherein the polypeptide comprises five or more alterations at positions selected from the group consisting of positions corresponding to the following positions of SEQ ID NO: 2: 492, 550, 573, 574 and 580.
37. The polypeptide according to embodiment wherein the polypeptide comprises alterations at positions corresponding to the following positions of SEQ I D NO: 2: 492G + 550K + 573P + 574H + 580K (e.g. SEQ ID NO: 267) or 492D + 550K + 573P + 574H + 580K (e.g. SEQ ID NO: 266).
38. The polypeptide according to any preceding embodiment comprising alterations corresponding to the following positions in SEQ ID NO: 2: 492G+573P+574H+580K (e.g. SEQ ID NO: 1 15); 492G+550K+573P+574H (e.g. SEQ ID NO: 257); 492D+550K+573P+574H (e.g. SEQ ID NO: 256); 492G+550K+573P (e.g. SEQ ID NO: 254); 492D+550K+573P (e.g. SEQ ID NO: 253); 550M+573P+580K (e.g. SEQ ID NO: 252); 550L+573P+580K (e.g. SEQ ID NO: 248); 550L+573P+574 H (e.g . SEQ I D NO : 246); 492G+573 P+580 K (e.g. SEQ I D NO: 242); 492D+573P+580 K (e.g . S EQ I D N O: 234); 573 P+574H +580 K (e.g . SEQ I D NO : 1 35); 550K+573P+580K (e.g. SEQ I D NO: 1 33); 550K+573P+574H (e.g. SEQ I D NO: 1 31 ); or 492D+573P+574H+580K (SEQ ID NO: 1 14).
39. The polypeptide of any of embodiments 1 to 38 wherein the reference albumin is HSA (SEQ ID No: 2) or a fragment thereof, or a fusion polypeptide comprising HSA or a fragment thereof, most preferably SEQ ID NO: 2.
40. The polypeptide according to any of embodiments 1 to 39, having a stronger binding affinity to FcRn and/or longer plasma half-life than a parent albumin, reference albumin, fragment thereof or fusion polypeptide comprising said parent albumin, reference albumin or fragment or fusion thereof.
41 . The polypeptide according any of embodiments 1 to 40, wherein the sequence identity of the polypeptide to SEQ ID NO: 2 is more than 80%, preferably more than 90%, more preferred more than 95%, more preferred more than 96%, even more preferred more than 97%, more preferred more than 98% and most preferred more than 99%.
42. A fusion polypeptide comprising a polypeptide according to any of embodiments 1 to 41 and a fusion partner polypeptide selected from a therapeutic, prophylactic, diagnostic, imaging or other beneficial moiety.
43. A method for preparing a polypeptide which is a variant of albumin, fragment thereof or fusion polypeptide comprising said variant albumin or fragment thereof having a binding affinity to FcRn which is altered compared to the binding affinity of a reference albumin, fragment or fusion thereof to FcRn, comprising:
(a) Providing a nucleic acid encoding a parent albumin having at least 80% sequence identity to SEQ ID NO: 2;
(b) Modifying the sequence of step (a), to encode a polypeptide which is a variant albumin, fragment thereof or fusion polypeptide comprising said variant albumin or fragment thereof comprising alterations at two or more positions corresponding to positions selected from 492, 550, 573, 574 and 580 in SEQ ID NO: 2, preferably as described in any of embodiments 1 to 13;
(c) Introducing the modified sequence of step (b) in a suitable host cell;
(d ) Growing the cells in a su itable growth mediu m under condition leading to expression of the polypeptide; and
(e) Recovering the polypeptide from the growth medium;
wherein the polypeptide has an altered binding affinity to FcRn and/or an altered plasma half-life compared with the half-life of a parent albumin, reference albumin, fragment thereof or fusion polypeptide comprising said parent albumin, reference albumin or fragment or fusion thereof.
44. The method of embodiment 43 wherein the substitution at the position corresponding to position 492 is a substitution to G, D, F, H, M or R, preferably G or D
45. The method of embodiment 44 wherein the substitution at the position corresponding to position 492 is to G.
46. The method of embodiment 44 wherein the substitution at the position corresponding to position 492 is to D.
47. The method of any of embodiments 43 to 46 wherein the substitution at the position corresponding to position 550 is to K, L, M, E or R, preferably K, L or M, most preferably K.
48. The method of any of embodiments 43 to 47 wherein the substitution at the position corresponding to position 573 is a substitution to P, Y, W, H, F, T, I or V, preferably P, Y, or W, most preferably P.
49. The method of any of embodiments 43 to 48 wherein the substitution at the position corresponding to position 574 is a substitution to H, D, F, G, N, S or Y, preferably H, D, F,or G, most preferably H.
50. The method of any of embodiments any of embodiments 43 to 49 wherein the substitution at the position corresponding to position 580 is a substitution to K or R, most preferably R.
51 . The method of any of embodiments 43 to 50 wherein the two or more alteraions are at positions corresponding to positions (i) 573 and 580; (ii) 492 and 573; or (iii) 573 and 574 of SEQ ID NO: 2.
52. The method according to any of embodiments 43 to 51 wherein the two or more alterations are at positions corresponding to the following positions of SEQ I D NO: 2: 580K + 573P (e.g. SEQ I D NO: 128); 580R + 573P (e.g. SEQ I D NO: 129); 574D + 573P (e.g. SEQ ID NO: 121 ); 574F + 573P (e.g. SEQ ID NO: 122); 574G + 573P (e.g. SEQ ID NO: 123); 574H + 573P (e.g. SEQ ID NO: 124); 574N + 573P (e.g. SEQ ID NO: 125); 574S + 573P (e.g. SEQ ID NO: 126); 550K + 580K (e.g. SEQ I D NO: 132); 550K + 574H (e.g. SEQ I D NO: 130); 550K + 573P (e.g. SEQ ID NO: 1 17); 492D + 573P (e.g. SEQ ID NO: 108); 492F + 573P (e.g. SEQ ID NO: 109); 492H + 573P (e.g. SEQ I D NO: 1 1 1 ); 492R + 573P (e.g. SEQ ID NO: 1 12); 574H + 580K (e.g. SEQ ID NO: 134); 550L + 574H (e.g. SEQ ID NO: 245); 550L + 580K (e.g. SEQ ID NO: 247); 550M + 580K (e.g. SEQ ID NO: 251 ); 492D + 550K (e.g. SEQ ID NO: 231 ); 550M + 574H (e.g. SEQ ID NO: 249); 492D + 574H (e.g. SEQ ID NO: 232); 492G + 550K (e.g. SEQ ID NO: 240); 550M + 574H (e.g. SEQ ID NO: 249); or 492G + 574H (e.g. SEQ ID NO: 241 ).
53. The method according to any of embodiments 43 to 52 comprising three or more alterations at positions selected from the group consisting of positions corresponding to the following positions of SEQ ID NO: 2: 492, 550, 573, 574 and 580.
54. The method according to any of embodiments 43 to 53 comprising alterations at positions corresponding to the following positions of SEQ ID NO: 2: 574H + 580K + 573P (e.g. SEQ ID NO: 135); 550K + 574H + 573P (e.g. SEQ ID NO: 131); 492D + 550K + 573P (e.g. SEQ ID NO: 253); 550M + 573P + 580K (e.g. SEQ ID NO: 252); 550L + 573P + 580K (e.g. SEQ ID NO: 253); 492G + 573P + 580K (e.g. SEQ ID NO: 242); 550M + 573P + 574H (e.g. SEQ ID NO: 250); 492G + 550K + 573P (e.g. SEQ ID NO: 254); 550L + 573P + 574H (e.g. SEQ ID NO: 246); 492D + 573P + 580K (e.g. SEQ ID NO: 234); 492D + 573P + 574H (e.g. SEQ ID NO: 233); 492G + 574H + 580K (e.g. SEQ ID NO: 263); 492G + 550K + 580K (e.g. SEQ ID NO: 259); 492D + 550K + 580K (e.g. SEQ ID NO: 258); 492D + 574H + 580K (e.g. SEQ ID NO: 262); or 492G + 550K + 574H (e.g. SEQ ID NO: 255).
55. The method according to any of embodiments 43 to 54 comprising four or more alterations at positions selected from the group consisting of positions corresponding to the following positions of SEQ ID NO: 2: 492, 550, 573, 574 and 580.
56. The method according to any of embodiments 43 to 55 comprising alterations at positions corresponding to the following positions of SEQ ID NO: 2: 492G + 573P + 574H + 580K (e.g. SEQ ID NO: 115); 492D + 573P + 574H + 580K (e.g. SEQ ID NO: 114); 550K + 573P + 574H + 580K (e.g. SEQ ID NO: 265); 492G + 550K + 573P + 580K (e.g. SEQ ID NO: 261); 492F + 573P + 574H + 580K (e.g. SEQ ID NO: 237); 492G + 573P + 574G + 580K (e.g. SEQ ID NO: 243); 492D + 573P + 574H + 580R (e.g. SEQ ID NO: 236); 492G + 573P + 574H + 580R (e.g. SEQ ID NO: 244); 492D + 550K + 573P + 580K (e.g. SEQ ID NO: 260); 492D + 550K + 573P + 574H (e.g. SEQ ID NO: 256); 492F + 573P + 574H + 580R (e.g. SEQ ID NO: 239); 492D + 573P + 574G + 580K (e.g. SEQ ID NO: 235); or 492F + 573P + 574G + 580K (e.g. SEQ ID NO: 238);
57. The method according to any of embodiments 43 to 56 comprising five or more alterations at positions selected from the group consisting of positions corresponding to the following positions of SEQ ID NO: 2: 492, 550, 573, 574 and 580.
58. The polypeptide according to any of embodiments 43 to 57 comprising alterations at positions corresponding to the following positions of SEQ ID NO: 2: 492G + 550K + 573P + 574H + 580K (e.g. SEQ ID NO: 267) or 492D + 550K + 573P + 574H + 580K (e.g. SEQ ID NO: 266).
59. The method according to any of embodiments 43 to 58 comprising alterations at positions corresponding to the following positions in SEQ ID NO: 2: 492G+573P+574H+580K
(e.g. SEQ ID NO: 115); 492G + 550K + 573P + 574H (e.g. SEQ ID NO: 257);
492D+550K+573P+574H (e.g. SEQ ID NO: 256); 492G+550K+573P (e.g. SEQ ID NO: 254);
492D+550K+573P (e.g. SEQ ID NO: 253); 550M+573P+580K (e.g. SEQ ID NO: 252); 550L+573P+580K (e.g. SEQ ID NO: 248); 550L+573P+574H (e.g. SEQ ID NO: 246);
492G+573P+580K (e.g. SEQ ID NO: 242); 492D+573P+580K (e.g. SEQ ID NO: 234);
573P+574H+580K (e.g. SEQ ID NO: 135); 550K+573P+580K (e.g. SEQ ID NO: 133);
550K+573P+574H (e.g. SEQ ID NO: 131); or 492D+573P+574H+580K (SEQ ID NO: 114). 60. The method any of embodiments 43 to 59 wherein the reference albumin is HSA (SEQ ID No: 2) or a fragment thereof, or a fusion polypeptide comprising HSA or a fragment thereof, most preferably SEQ ID NO: 2.
61 . The method according any of embodiments 43 to 60, wherein the sequence identity of the polypeptide to SEQ ID NO: 2 is more than 80%, preferably more than 90%, more preferred more than 95%, more preferred more than 96%, even more preferred more than 97%, more preferred more than 98% and most preferred more than 99%.
62. A conjugate comprising a polypeptide according to any of embodiments 1 to 42 or obtainable by a method according to any of embodiments 43 to 61 and a conjugation partner. 63. The conjugate according to embodiment 62 wherein the conjugation partner is a therapeutic, prophylactic, diagnostic, imaging or other beneficial moiety.
64. An associate comprising a polypeptide according to any of embodiments 1 to 42 or obtainable by a method according to any of embodiments 43 to 61 and a therapeutic, prophylactic, diagnostic, imaging or other beneficial moiety.
65. A nanoparticle or microparticle com prisi ng a polypeptide accord i ng to any of embodiments 1 to 42 or obtainable by a method according to any of embodiments 43 to 61 , a conjugate according to embodiment 62 or 63 or an associate according to embodiment 64.
66. A composition comprising a polypeptide according to any of embodiments 1 to 42 or obtainable by a method according to any of embodiments 43 to 61 , a conjugate according to embodiment 62 or 63, an associate according to embodiment 64 or a nanoparticle or microparticle according embodiment 65, wherein the binding affinity of the polypeptide, fusion polypeptide, conjugate, associate or nanoparticle or microparticle to FcRn is stronger than the binding affinity of a composition comprising the corresponding parent albumin , reference albumin , fragment thereof or fusion polypeptide, conjugate, associate or nanoparticle or microparticle comprising said parent albumin, reference albumin or fragment or fusion thereof to FcRn.
67. A composition according to embodiment 66 where the binding affinity of the polypeptide, fusion polypeptide, conjugate, associate or nanoparticle or microparticle to FcRn is stronger than the binding affinity of HSA to FcRn.
68. A composition according to embodiment 66 or 67, wherein the binding coefficient of the variant of the polypeptide, fusion polypeptide, conjugate, associate or nanoparticle or microparticle to FcRn is less than 0.9X KD of HSA to FcRn, more preferred less than 0.5X KD of HSA to FcRn, more preferred less than 0.1 X KD of HSA to FcRn, even more preferred less than 0.05X KD of HSA to FcRn, even more preferred less than 0.02X KD of HSA to FcRn and most preferred less than 0.01X KD of HSA to FcRn.
69. The composition according to any of embodiments 66 to 68, comprising a polypeptide according to any of embodiments 1 to 42 or obtainable by a method according to any of embodiments 43 to 61 , a conjugate according to embodiment 62 or 63, an associate according to embodiment 64 o r a n anoparticle or microparticle according embodiment 65, further comprising a compound comprising an antibody binding domain (ABD) and a therapeutic, prophylactic, diagnostic, imaging or other beneficial moiety.
70. The com position accord i ng to a ny of em bod i ments 66 to 69, comprising a pharmaceutically acceptable carrier or excipient.
71 . Use of a polypeptide according to any of embodiments 1 to 42 or obtainable by a method according to any of embodiments 43 to 61 , a conjugate according to embodiment 62 or 63, an associate according to embodiment 64 or a nanoparticle or microparticle according embodiment 65 or a composition according to any of embodiments 66 to 70 to alter the binding affinity to FcRn or half-life, preferably in plasma, of a therapeutic, prophylactic, diagnostic, imaging or other beneficial moiety.
72. The use according to embodiment 71 wherein the binding affinity to FcRn is increased relative to the binding affinity of a reference comprising or consisting of HSA (SEQ ID NO: 2) or a fragment, fusion, conjugate, associate, nanoparticle or microparticle thereof to FcRn.
73. The use according to embodiment 71 wherein the binding affinity to FcRn is decreased relative to the binding affinity of a reference comprising or consisting of HSA (SEQ ID NO: 2) or a fragment, fusion, conjugate, associate, nanoparticle or microparticle thereof to FcRn.
74. A method for altering the binding affinity to FcRn or half-life preferably in plasma, of a molecule comprising:
(a) where the molecule is a polypeptide, fusing or conjugating the molecule to a polypeptide according to any of embodiments 1 to 42 or obtainable by a method of embodiments 43 to 61 , or to a conjugate according to embodiment 62 or 63; associating the molecule to a polypeptide according to any of embodiments 1 to 42 or obtainable by a method of embodiments 43 to 61 or to a conjugate according to embodiment 62 or 63; incorporating the molecule in an associate according to embodiment 64, in nanoparticle or microparticle according to embodiment 65 or a composition according to any of embodiments 66 to 70;
(b) where the molecule is not a polypeptide, conjugating the molecule to a polypeptide according to any of embodiments 1 to 42 or obtainable by a method of embodiments 43 to 61 , or to a conjugate according to embodiment 62 or 63; associating the molecule to a polypeptide according to any of embodiments 1 to 42 or obtainable by a method of embodiments 43 to 61 or to a conjugate according to embodiment 62 or 63; incorporating the molecule in an associate according to embodiment 64, in nanoparticle or microparticle according to embodiment 65 or a composition according to any of embodiments 66 to 70.
75. A method accord ing to embodiment 74 wherein the molecule is a therapeutic, prophylactic, diagnostic, imaging or other beneficial moiety.
76. A polypeptide, fusion polypeptide, conjugate, associate, nanoparticle or microparticle or composition thereof according to any of embodiments 1 to 42 or 62 to 70 or obtainable by the method of embodiments 43 to 61 wherein the polypeptide, fusion polypeptide, conjugate, associate, nanoparticle or microparticle or composition comprises one or more (several) moiety selected from those described herein.
77. A nucleic acid encoding the polypeptide or fusion polypeptide of any of embodiments 1 to 76.
78. A vector comprising a nucleic acid according to embodiment 77.
79. A host cell comprising a nucleic acid according to embodiment 77 or a vector according to embodiment 78.
80. A host cell according to embodiment 79 wherein the host cell is a eukaryote, preferably a yeast (such as Saccharomyces cerevisiae) or a mammalian cell (such as CHO or HEK) or a plant cell (such as rice).
81 . A method of prophylaxis, treatment or diagnosis comprising administering a polypeptide, fusion polypeptide, conjugate, composition, associate, nanoparticle or microparticle or polynucleotide according to any of embodiments 1 to 42 or 62 to 70 or obtainable by the method of any of embodiments 43 to 61 to a subject.
The invention is further described by the following examples that should not be construed as limiting the scope of the invention.
Examples
Example 1. Preparation of HSA variant expression plasmids for HSA
HSA variants were expressed using standard molecular biology techniques, such as described in Sambrook, J. and D.W. Russell, 2001 (Molecular Cloning: a laboratory manual, 3rd ed. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y).
Method 1 : Permutation libraries at positions E492, D550, K574H and Q580K were produced by PCR amplification of pDB4081 , encoding wild type HSA (described below), using a mutagenic forward primer and non-mutagenic reverse primer as shown in Table 2. The PCR conditions are shown in Tables 3 and 4. 2 μΙ of reaction product was assessed by agarose gel electrophoresis and the remainder treated with 5μΙ of 10x buffer 4 (New England Biolabs - 50 mM potassium acetate, 20mM Tris-acetate, 10mM magnesium acetate, 1 mM DTT pH 7.9 at 25~C) and 1 μΙ Dpn\ (NEB) for 1 hour at 37° C. The reactions were purified by addition of 50 μΙ water, prior to application of a vacuum for 20 minutes. A further 50 μΙ of water was added and a vacuum applied until the well was dry. Plasmid DNA was recovered by addition of 30 μΙ of water and incubation for 1 minute. 2 μΙ of this purified product was transformed into 15 μΙ TOP10 E. coli cells by standard protocols. Clones were selected and grown overnight in LB supplemented with 100 μg ml ampicillin and purified using a Qiagen QIAprep 96 miniprep kit according to manufacturer's instructions. Variants were verified by sequencing.
pDB4081 was made by the ligation of a synthetic DNA fragment, Bsa\/Sph\ digested, which had been generated by gene assembly (DNA2.0 Inc, USA), containing 3' region of the PRB1 promoter, modified fusion leader sequence, nucleotide sequence encoding HSA and 5' region of the modified AD \ terminator) into H/ndl I l/Sp/7 l-d i gested pDB4005. pDB4005 is described in WO 201 1/051489 (incorporated herein by reference). Table 2: Plasmid and amino acid substitution and relevant primers
Figure imgf000071_0001
Variant Variant SEQ ID Oligol Oligo 2 Plasmid No. (SEQ ID No.) (SEQ ID No.) name
HSA D550N 61 HSA-30 (165) REV D550 (213) pDB4793
HSA D550P 62 HSA-31 (166) REV D550 (213) pDB4794
HSA D550Q 63 HSA-32 (167) REV D550 (213) pDB4795
HSA D550R 64 HSA-33 (168) REV D550 (213) pDB4796
HSA D550S 65 HSA-34 (169) REV D550 (213) pDB4797
HSA D550T 66 HSA-35 (170) REV D550 (213) pDB4798
HSA D550V 67 HSA-36 (171 ) REV D550 (213) pDB4799
HSA D550Y 68 HSA-37 (172) REV D550 (213) pDB4800
HSA D550W 69 HSA-38 (173) REV D550 (213) pDB4801
HSA K574A 70 HSA-39 (174) REV K574 (214) pDB4802
HSA K574C 71 HSA-40 (175) REV K574 (214) pDB4803
HSA K574D 72 HSA-41 (176) REV K574 (214) pDB4804
HSA K574E 73 HSA-42 (177) REV K574 (214) pDB4805
HSA K574F 74 HSA-43 (178) REV K574 (214) pDB4806
HSA K574G 75 HSA-44 (179) REV K574 (214) pDB4807
HSA K574H 76 HSA-45 (180) REV K574 (214) pDB4808
HSA K574I 77 HSA-46 (181 ) REV K574 (214) pDB4809
HSA K574L 78 HSA-47 (182) REV K574 (214) pDB4810
HSA K574M 79 HSA-48 (183) REV K574 (214) pDB481 1
HSA K574N 80 HSA-49 (184) REV K574 (214) pDB4812
HSA K574P 81 HSA-50 (185) REV K574 (214) pDB4813
HSA K574Q 82 HSA-51 (186) REV K574 (214) pDB4814
HSA K574R 83 HSA-52 (187) REV K574 (214) pDB4815
HSA K574S 84 HSA-53 (188) REV K574 (214) pDB4816
HSA K574T 85 HSA-54 (189) REV K574 (214) pDB4817
HSA K574V 86 HSA-55 (190) REV K574 (214) pDB4818
HSA K574Y 87 HSA-56 (191 ) REV K574 (214) pDB4819
HSA K574W 88 HSA-57 (192) REV K574 (214) pDB4820
HSA Q580A 89 HSA-58 (193) REV Q580 (215) pDB4821
HSA Q580C 90 HSA-59 (194) REV Q580 (215) pDB4822
HSA Q580D 91 HSA-60 (195) REV Q580 (215) pDB4823
HSA Q580E 92 HSA-61 (196) REV Q580 (215) pDB4824
HSA Q580F 93 HSA-62 (197) REV Q580 (215) pDB4825
HSA Q580G 94 HSA-63 (198) REV Q580 (215) pDB4826 Variant Variant SEQ ID Oligol Oligo 2 Plasmid No. (SEQ ID No.) (SEQ ID No.) name
HSA Q580H 95 HSA-64 (199) REV Q580 (215) pDB4827
HSA Q580I 96 HSA-65 (200) REV Q580 (215) pDB4828
HSA Q580K 97 HSA-66 (201 ) REV Q580 (215) pDB4829
HSA Q580L 98 HSA-67 (202) REV Q580 (215) pDB4830
HSA Q580M 99 HSA-68 (203) REV Q580 (215) pDB4831
HSA Q580N 100 HSA-69 (204) REV Q580 (215) pDB4832
HSA Q580P 101 HSA-70 (205) REV Q580 (215) pDB4833
HSA Q580R 102 HSA-71 (206) REV Q580 (215) pDB4834
HSA Q580S 103 HSA-72 (207) REV Q580 (215) pDB4835
HSA Q580T 104 HSA-73 (208) REV Q580 (215) pDB4836
HSA Q580V 105 HSA-74 (209) REV Q580 (215) pDB4837
HSA Q580Y 106 HSA-75 (210) REV Q580 (215) pDB4838
HSA Q580W 107 HSA-76 (21 1 ) REV Q580 (215) pDB4839
Table 3: PCR reaction components
Figure imgf000073_0001
Table 4: PCR reaction conditions
Figure imgf000073_0002
Method 2: Variants described in Table 5 and 6 were produced as described in WO2012/150319, Example 6, Method 2 'Production of combination Variants with K573P' (incorporated herein by reference), with the following modifications. For variants shown in Table 5, a fragment encoding the K573P mutation was removed from pDB4673 via the Sa/I and Bsu36\ restriction sites and inserted into similarly digested parent plasmid, as indicated in Table 5. pDB4673 (HSA K573P) was constructed by insertion of the fragment produced by digestion of pD4283 (described in WO 201 1/051489, incorporated herein by reference) with Sa/I and HinD\ \\ restriction enzymes into similarly digested pDB4081 . For variants shown in Table 6, the fragment encoding K573P+K574H+Q580K was removed from pDB5032 using the Sa/I and Bsu36\ restriction sites and inserted into similarly digested parent plasmid, as indicated in Table 6.
Table 5: Plasmid and amino acid substitution
Figure imgf000074_0001
Table 6: Plasmid and amino acid substitution
Figure imgf000074_0002
Method 3: Variants described in Table 7 were produced as described in WO2012/150319, Example 6, Method 1 (incorporated herein by reference), using the plasmids indicated in the tables as templates for the PCR reactions. The conditions described in Tables 8 and 9 were used to produce pDB4997-5004, pDB 5042-46, and pDB5030-1 . Those described in Tables 10 and 1 1 were used to produce pDB5027-29 and pDB5032. The mutated PCR products were digested using Sa/I and Bsu36\ restriction enzymes and inserted into similarly digested pDB4081 , encoding wild type HSA (as described above).
Table 7: Plasmid and amino acid substitution and relevant primers
Figure imgf000074_0003
Variant Variant Oligo 1 Oligo 2 Template Plasmid
S E Q I D ( S E Q I D (SEQ ID No.) plasmid Name No. No.)
HSA D550K+K573P 1 17 KBf4 (216) KBr30 (217) pDB4790 pDB5043
HSA D550L+K573P 1 18 KBf4 (216) KBr30 (217) pDB4791 pDB5044
HSA D550M+K573P 1 19 KBf4 (216) KBr30 (217) pDB4792 pDB5045
HSA D550R+K573P 120 KBf4 (216) KBr30 (217) pDB4796 pDB5046
HSA K574D+K573P 121 KBf4 (216) KBr21 (218) pDB4673 pDB4997
HSA K574F+K573P 122 KBf4 (216) KBr22 (219) pDB4673 pDB4998
HSA K574G+K573P 123 KBf4 (216) KBr23 (220) pDB4673 pDB4999
HSA K574H+K573P 124 KBf4 (216) KBr24 (221 ) pDB4673 pDB5000
HSA K574N+K573P 125 KBf4 (216) KBr25 (222) pDB4673 pDB5001
HSA K574S+K573P 126 KBf4 (216) KBr26 (223) pDB4673 pDB5002
HSA K574Y+K573P 127 KBf4 (216) KBr27 (224) pDB4673 pDB5005
HSA Q580K+K573P 128 KBf4 (216) KBr28 (225) pDB4673 pDB5003
HSA Q580R+K573P 129 KBf4 (216) KBr29 (226) pDB4673 pDB5004
HSA D550K+K574H 130 KBf4 (216) KBr31 (227) pDB4790 pDB5027
HSA 131 KBf4 (216) KBr24 (221 ) pDB4790 pDB5028
D550K+K574H+K573P
HSA D550K+Q580K 132 KBf4 (216) KBr32 (228) pDB4790 pDB5029
HSA 133 KBf4 (216) KBr28 (225) pDB5043 pDB5030
D550K+Q580K+K573P
HSA K574H+Q580K 134 KBf4 (216) KBr33 (229) pDB4808 pDB5031
HSA 135 KBf4 (216) KBr34 (230) pDB5003 pDB5032
K574H+Q580K+K573P
Table 8: PCR reaction components
Figure imgf000075_0001
Table 9: PCR reaction conditions
Figure imgf000075_0002
Figure imgf000076_0001
Table 10: PCR reaction components
Figure imgf000076_0002
Table 11 : PCR reaction conditions
Figure imgf000076_0003
Preparation of expression plasmids and transformation of S. cerevisiae was performed as described in WO2012/150319 (incorporated herein by reference), employing the 24 (pDB5027-9, pDB5032 and pDB5091 -92), or 48 hour stocking method (all remaining variants). The host strain for pDB4990-5, pDB4997-5004, pDB5042-5046, pDB5027-32 and pDB5091 -2 was S. cerevisiae DYB7 (Payne et al (2008) Applied and Environmental Microbiology Vol 74(24): 7759-7766) with four copies of PDI integrated into the genome. The host strain for the remaining plasmids was S. cerevisiae BXP10cir° as described in WO2012/150319 (incorporated herein by reference). Protein isolation and purification from shake flask was performed as described in WO201 1/051489.
Example 2. SPR analysis of binding affinity of WT HSA and variants to shFcRn
SP R analyses were performed on a Biacore 3000 instrument (GE Healthcare). Immobilisation was carried out on CM5 chips coupled with His-tagged shFcRn (FcRn produced by GeneArt, Life Technologies) using GE Healthcare amine coupling chemistry as per the manufacturer's instructions. Immobilised levels of shFcRn-HIS (shFcRn with a 6-His tail on the C-terminus of beta-2-microglobulin) were 1200 - 2500RU and achieved by injecting 20μg/mL shFcRn diluted using sodium acetate pH4.5 (G E Healthcare). Chip surface was left to stabilize with a constant flow (5μΙ_Ληίη) of running buffer - Di-basic/Mono-basic phosphate buffer pH5.5 ((67 mM phosphate buffer, 0.15 M NaCI, 0.005% Tween 20) at pH 5.5)) at 25 °C (i.e. ambient temperature) overnight. After ligand stabilization, the chip surface was conditioned by injecting 5-12 x 45μΙ_ Di-basic/Mono-basic phosphate buffer at 30μΙ_Ληίη followed by HBS_EP (0.01 M H EPES, 0.15 M NaCI , 3mM EDTA, 0.005% surfactant P20) at pH 7.4 (GE Healthcare)) regeneration steps (12s) in between each injection. Surfaces were then checked for activity by injecting 3χ45μΙ_ positive control (wt HSA (S EQ I D NO : 2)) at 30μΙ_Ληίη, followed by 12s regeneration pulse. Kinetic measurements were performed by injecting dilutions (100μΜ - 0.016μΜ) of HSA and HSA variants at 30μΙ_ Ήη over immobilised shFcRn, at 25°C (i.e. ambient temperature). The reference cell value was then subtracted and Biaevaluation software 4.1 used to obtain kinetic data and confirm KD values. The variants were wild-type albumin (SEQ ID NO: 2) and albumins with alterations at one or more of positions 492, 550, 573, 574 and 580. The variants were analysed by SPR to determine their binding response (RU) to shFcRn. Some variants were further characterized to determine KD values. The data are shown in Tables 12 to 18. Table 12: Binding affinity of selected albumin variants (with a single alteration) to shFcRn
Run Analyte SEQ ID No. Binding % i ncrease i n bi ndi ng response (RU) affinity compared to WT
A WT HSA 2 157 -
HSA-K573P 3 247 57.3
HSA-E492D 34 192 22.5
HSA-E492F 35 200 27.6
HSA-E492G 36 187 19.4
HSA-E492H 37 186 18.8
HSA-E492M 41 184 17.5
HSA-E492R 45 184 17.5
B WT HSA 2 150 -
HSA-K573P 3 21 1 40.7
HSA-D550E 53 175 16.8
HSA-D550K 58 186 24.4
HSA-D550L 59 187 24.6
HSA-D550M 60 177 18.0
HSA-D550R 64 177 18.2
C WT HSA 2 171 -
HSA-K573P 3 223 30.4
HSA-K574D 72 214 25.4
HSA-K474F 74 219 27.1
HSA-K574G 75 225 31.7 Run Analyte SEQ ID No. Binding % i ncrease i n bi ndi ng response (RU) affinity compared to WT
HSA-K574H 76 230 34.7
HSA-K574N 80 205 20.4
HSA-K574S 84 205 20.1
HSA-K574Y 87 207 21.1
D WT HSA 2 159 -
HSA-K573P 3 227 42.8
HSA-Q580K 97 212 33.4
HSA-Q580R 102 195 22.3
The data in Table 12 show that variants comprising an alteration at one of positions 492, 550, 574 and 580 show a higher FcRn binding affinity than WT albumin. Table 13: Binding affinity of albumin variants (with two alterations) to shFcRn
Figure imgf000078_0001
conjunction with the K573P variant show higher binding affinity than WT albumin.
Table 14: Binding affinity of albumin variants to shFcRn
Fold
Mean
SEQ ID Ka Kd KD difference
Analyte KD
NO: (103/Ms) (10"3/s) (μΜ) from HSA-
(μΜ)
WT
WT HSA (run 1 ) 2 10.9 40 3.67
4.02 -
WT HSA (run 2) 2 9.15 39.9 4.36
HSA-K573P (run 1 ) 3 5.99 2.15 0.358
0.39 10.3
HSA-K573P (run 2) 3 5.95 2.53 0.426
HSA-E492D+K573P (run 1 ) 108 1 1.5 1 .68 0.147
0.162 24.8
HSA-E492D+K573P (run 2) 108 1 1.1 1 .96 0.177
HSA-E492G+K573P (run 1 ) 1 10 12.7 2.15 0.17 0.175 23.0 Fold
Mean
SEQ ID Ka Kd KD difference
Analyte KD
NO: (103/Ms) (10"3/s) (μΜ) from HSA-
(μΜ)
WT
HSA-E492G+K573P (run 2) 1 10 1 1.3 2.04 0.18
Variants HSA-E492D+K573P and HSA-E492G+K573P were selected for further analysis based on their apparent slow 'off rates' (i.e. dissociation constants (Kd)). The data in Table 14 show that variants HSA-E492D+K573P and HSA-E492G+K573P show higher affinity for shFcRn as compared to WT albumin and the single K573P variant.
Table 15: Binding affinity of albumin variants (with two alterations) to shFcRn
Figure imgf000079_0001
The data in Table 15 show that introduction of substitutions to the K573P variant which had individually demonstrated an improvement in affinity for shFcRn (Table 12), produced a range of binding responses, from a reduction in affinity as compared to WT albumin, through to variants showing increased affinity.
Table 16: Binding affinity of albumin variants to shFcRn
Analyte SEQ ID Ka Kd KD Mean KD Fold
NO: (103/Ms) (10"3/s) (μΜ) (μΜ) difference from WT HSA
WT HSA 2 17.3 63.0 3.6 3.6 -
HSA-K573P (run 1 ) 3 8.1 3.4 0.42
0.46 7.8
HSA-K573P (run 2) 3 8.0 4.0 0.5
HSA-E492H+K573P (run 1 ) 1 1 1 1 1.2 3.7 0.33 0.37 9.7 Analyte SEQ ID Ka Kd KD Mean KD Fold
NO: (103/Ms) (10"3/s) (μΜ) (μΜ) difference from WT HSA
HSA-E492H+K573P (run 2) 1 1 1 1 1.4 4.7 0.41
HSA-E492R+K573P 1 13 10.3 4.3 0.42 0.42 8.6
HSA-K574G+K573P (run 1 ) 123 1 1.5 5.5 0.48
0.53 6.8
HSA-K574G+K573P (run 2) 123 1 1.7 6.8 0.58
HSA-K574H+K573P (run 1 ) 124 12.6 2.7 0.22
0.25 14.4
HSA-K574H+K573P (run 2) 124 10.8 3.08 0.29
HSA-Q580K+K573P (run 1 ) 128 13.4 1 .9 0.14
0.15 24.0
HSA-Q580K+K573P (run 2) 128 13.2 2.19 0.16
The data in Table 16 show that variants HSA-K574H+K573P and HSA-Q580K+K573P possess considerably higher affinity for shFcRn as compared to WT albumin and the single K573P variant. Table 17: Binding affinity of albumin variants to shFcRn
Figure imgf000080_0001
550, 573, 574 and 580. For example, HSA-K574H+K573P showed increased binding compared to WT albumin (Table 16). Introduction of the K574H mutation into HSA-Q580K+K573P (Table 16) to produce HSA-K574H+Q580K+K573P resulted in no improvement in binding affinity (compared to WT albumin) over HSA-Q580K+K573P. In contrast, introduction of this mutation into HSA-D550K+K573P, producing HSA-D550K+K574H+K573P, has resulted in an increase in affinity compared to WT albumin and HSA-D550K+K573P.
Table 18: Binding affinity of albumin variants to shFcRn
Figure imgf000081_0001
The data in Table 18 show that variants containing four substitutions at positions 492 573P, 574 and 580 show a marked increase in binding affinity as compared to variants containing three mutations, such as HSA-K574H+Q580K+K573P (Table 17), variants containing two mutations, such as K574H+K573P and Q580K+K573P (Table 16) and the single K573P variant or WT albumin.
Example 3. Production of further combination variants.
Single or multiple further mutations were introduced into the template plasmids listed in Table 19 to produce the indicated variants. Mutagenic forward primers and non-mutagenic reverse primers were used to introduce the desired changes, as listed in Table 20. The template plasmids were methylated using the components described in Table 21 , incubated at 37°C for one hour and were then purified using a Qiagen QiaQuick PCR purification kit according to manufacturer's instructions. The methylated template was then used in mutagenic PCR reactions (Tables 22 and 23) according to the oligonucleotide/template combination detailed in Table 19 to produce the required HSA variants. Following PCR reaction, 5 μΙ of each PCR reaction mixture was visualised by agarose gel electrophoresis to assess production of the plasmid and 5 μΙ of plasmid was retained for further analysis.
Table 19. Variants and associated templates and oligonucleotides for mutagenic PCR amplification
Figure imgf000082_0001
Variant Oligo Oligo Template Seq ID forward reverse no.
HSA E492D+D550K+K573P+Q580K 28-f 28-r pDB5030 260
HSA E492G+D550K+K573P+Q580K 29-f 29-r pDB5030 261
HSA E492D+K574H+Q580K 28-f 28-r pDB5031 262
HSA E492G+K574H+Q580K 29-f 29-r pDB5031 263
HSA E492D+K573P+K574H+Q580K 28-f 28-r pDB5032 264
HSA D550K+K573P+K574H+Q580K 01 -f 01 -r pDB5032 265
HSA E492D+D550K+K573P+K574H+Q580K 01 -f 01 -r pDB5091 266
HSA E492G+D550K+K573P+K574H+Q580K 01 -f 01 -r pDB5092 267
(f = forward, r = reverse)
Table 20. Oligonucleotides for mutagenic PCR amplification.
Figure imgf000083_0001
08-r ACCTTCTTCAGCGAAACAAGTTTCCTTATCATCAGC 283
28-f GAAGACCATGTTTCTCTGCTTTGGACGTCGACGAAACTTACGTTC 284
28-r CAAAGCAGAGAAACATGGTCTTCTGTTAACCAAAGA 285
29-f GAAGACCATGTTTCTCTGCTTTGGGTGTCGACGAAACTTACGTTC 286
29-r CAAAGCAGAGAAACATGGTCTTCTGTTAACCAAAGA 287
(f = forward, r = reverse)
Table 21. Methylation reaction components
Figure imgf000084_0001
Table 22: PCR reaction components
Figure imgf000084_0002
Table 23: PCR reaction conditions for construction of variants listed in Table 19
Figure imgf000084_0003
40 μΙ of each of the PCR-generated plasmids was prepared for in vivo recombination by the addition of 0.5 μΙ of each of Dpn\, Nsi\ and Pvu\ restriction enzymes (New England Biolabs), followed by incubation at 37°C for one hour. 3 μΙ of the prepared plasmid, 3 μΙ of Acc65\/Bam \- digested pDB3936 and 1 μΙ of salmon sperm DNA were used to transform S. cerevisiae according to the protocol described in Example 1 . The host strain used was a S. cerevisiae strain derived from DYB7 ura3 (Payne et a/ 2008, Appl. Environ. Microbiol. 74(24): 7759-7766) with two additional copies of PDI1 integrated into the genome. Single colonies were patched onto BMMD+CSM-leu plates (as described in WO 2012/150319, incorporated herein by reference) to enable assessment of expression prior to production of yeast stocks. Stocks were produced by the 48-hour method described in Example 1. 200 μΙ of each yeast strain was used to inoculate 10 ml BMMS in 50 ml shake flask, followed by incubation at 30°C, 200 rpm for four days. Culture supernatant was harvested by centrifugation at 3000 rpm for 5 minutes.
Albumin variants were purified from 10ml_ shake flasks using a single chromatographic step with an albumin affinity matrix (AlbuPure™ - ProMetic Biosciences, Inc.). Micro-scale affinity chromatography was performed on an automated platform (Perkin Elmer, Janus) with 200μΙ_ custom packed Atoll columns with 8 run in parallel in a 96-well format using the same procedure as described in WO 201 1/051489 (incorporated herein by reference), with volumes scaled down appropriately.
Final concentration of the samples was determined by Absorbance at 280nm using a UV m icroplate and a plate reader, read i ngs were blan k corrected agai nst P BS and the concentrations calculated based on an extinction coefficient of 0.52AU/cm for a 1 mg/ml_ solution (AU: absorbance units). Example 4. Analysis of further combination variants.
Kinetic analyses using bio-layer interferometry were performed on an Octet Red-96 instrument (ForteBio). Immobilisation of GST-tagged shFcRn (shFcRn-GST/FLAG) was carried out on AR2G biosensors using ForteBio amine coupling chemistry following the instructions from the manufacturer. shFcRn-GST/FLAG- refers to GST- tag (glutathione-transferase) and a FLAG-tag (DYKDDDDK) on the C-terminal of the alpha chain of FcRn . Prior to use, the GST/FLAG-tagged shFcRn was purified using IgG affinity chromatography. More specifically, the GST/FLAG-tagged shFcRn was captured on a GSTrap column and eluted with reduced glutathione. sh FcRn was d ialyzed into PBS into pH 7.4 and further pu rified using IgG Sepharose™ 6 Fast Flow (GE Healthcare). The shFcRn was captured on the resin in 50mM Na- acetate, 1 50m M N aCI pH 5.5 a nd el uted with P BS p H 7.4. The el uted shFcRn was concentrated to 2-5mg/ml_ and stored at -20°C until use. Immobilised level of sh FcRn- GST/FLAG was at a response level more than 1 nm, and achieved using a FcRn concentration of 2-10μg/mL in sodium acetate, followed by ethanolamine quenching of the amine coupling reaction. The sensors were either used directly or soaked in 15% (w/v) sucrose and dried until use.
Kinetic analyses were performed using micro-scale affinity purified albumin variants diluted 2-, 4- and 8-fold in BMMD fermentation media (as described WO 201 1/051489, incorporated herein by reference) supplemented with 100mM sodium acetate and adjusted to pH 5.5. Association (120s) and dissociation (300s) were performed at 30°C and shaking at l OOOrpm. ForteBio software was used for data evaluation and calculation of KD values as well as association and dissociation constants which were calculated using the HSA concentrations determined by OD280nm measurement. Table 24: Binding affinity of albumin variants to shFcRn
Figure imgf000086_0001
SEQ ID Variant KD Average μΜ Fold difference
No: relative to WT
HSA
264 HSA E492D+K573P+K574H+Q580K 0.03 5.3
265 HSA D550K+K573P+K574H+Q580K 0.02 6.1
266 HSA E492D+D550K+K573P+K574H+Q580K 0.02 6.5
267 HSA E492G+D550K+K573P+K574H+Q580K 0.02 6.9
2 WT HSA 0.15 1 .0
Example 5. Further analysis of selected combination variants.
A subset of the variants (Table 25) described in Table 19 were selected for further analysis. 3 μΙ of the PCR reactions were digested with 7 μΙ of a reaction mix containing 10 μ I of buffer 4 (New England Biolabs), 5 μΙ Dpn\ and 55 μΙ of water. Reaction mixtures were incubated at 37°C for 1 .5 hours and were then purified using a Qiagen QiaQuick PCR purification kit according to manufacturer's instructions. 2 μΙ of the prepared plasmids were used to transform competent £. coli 10-beta cells (New England Biolabs). Plasmid DNA was prepared utilising a Qiagen Plus Maxiprep kit, according to manufacturer's instructions. The resulting plasmids were sequenced to ensure the desired mutations had been introd uced . Plasmid preparation and yeast transformations were performed as described in example 1 , using strain BXP10 cir°. 24 hour yeast stocks were produced as described in example 1 .
Table 25: Variant and plasmid number
Figure imgf000087_0001
The variants listed in Table 26 and 27 were analyzed by SPR using a Biacore 3000 instrument as described in Example 2 with the exception that, prior to use, the His-tagged shFcRn was purified using IgG affinity chromatography. More specifically, the His-tagged shFcRn was captured on a Ni-HiTrap column and eluted with imidazole. The shFcRn was captured on the resin in 50mM Na-acetate, 150mM NaCI pH 5.5 and eluted with PBS pH 7.4. The eluted shFcRn was concentrated to 2-5mg/ml_ and stored at -20°C until use. using shFcRn-HIS
Table 26: SPR analysis of binding affinity of HSA variants to shFcRn
Figure imgf000088_0001
The data of Table 26 and 27 show an improvement in affinity over WT HSA in variants containing a combination of substitutions. Generally, variants containing three or four substitutions show improved binding characteristics over those containing two substitutions. Generally, inclusion of Q580K contributes substantially to an improved binding affinity.
The invention described and claimed herein is not to be limited in scope by the specific aspects herein disclosed, since these aspects are intended as illustrations of several aspects of the invention. Any equivalent aspects are intended to be within the scope of this invention. Indeed, various modifications of the invention in addition to those shown and described herein wil l become apparent to those skil led in the art from the foregoing description . Such modifications are also intended to fall within the scope of the appended claims. In the case of conflict, the present disclosure including definitions will control.

Claims

CLAIMS What is claimed is:
1 . A polypeptide which is a variant of albumin, fragment thereof or fusion polypeptide comprising said variant albumin or a fragment thereof having an altered binding affinity to FcRn compared with the binding affinity of a parent albumin, reference albumin, fragment thereof or fusion polypeptide comprising said parent albumin, reference albumin or fragment or fusion thereof to FcRn , wherein the polypeptide comprises alterations at two or more positions selected from positions corresponding to positions (a) 492 and 580; (b) 492 and 574; (c) 492 and 550; (d) 550 and 573; (e) 550 and 574; (f) 550 and 580 in SEQ ID NO: 2.
2. The polypeptide according to claim 1 comprising alterations at positions corresponding to positions 492 and 580 of SEQ ID NO: 1 , further comprising one or more (several) alterations at positions corresponding to positions selected from the group consisting of 550, 573 and 574 of SEQ ID NO: 2.
3. The polypeptide according to claim 1 comprising alterations at positions corresponding to positions 492 and 574 of SEQ ID NO: 1 , further comprising one or more (several) alterations at positions corresponding to positions selected from the group consisting 550, 573 and 580 of SEQ ID NO:
4. The polypeptide according to claim 1 comprising alterations at positions corresponding to positions 492 and 550 of SEQ ID NO: 1 , further comprising one or more (several) alterations at positions corresponding to positions selected from the group consisting 573, 574 and 580 and SEQ ID NO: 2.
5. A polypeptide according to claim 1 comprising alterations at positions corresponding to positions 550 and 573 of SEQ ID NO: 1 , further comprising one or more (several) alterations at positions corresponding to positions selected from the group consisting 492, 574, and 580 of SEQ ID NO: 2.
6. The polypeptide according to claim 1 comprising alterations at positions corresponding to positions 550 and 574 of SEQ ID NO: 1 , further comprising one or more (several) alterations at positions corresponding to positions selected from the group consisting 492, 573 and 580 of SEQ ID NO: 2.
7. The polypeptide according to claim 1 comprising alterations at positions corresponding to positions 550 and 580 of SEQ I D NO: 1 , further comprising an alteration at a position corresponding to position 492, 573 and 574 of SEQ ID NO: 2.
8. A polypeptide which is a variant of albumin, fragment thereof or fusion polypeptide comprising said variant albumin or a fragment thereof having an altered binding affinity to FcRn compared with the binding affinity of a parent albumin, reference albumin, fragment thereof or fusion polypeptide comprising said parent albumin, reference albumin or fragment or fusion thereof to FcRn, wherein:
(a) the polypeptide comprises alterations at two or more positions, in which at a position corresponding to position 492 of SEQ I D NO: 2 there is an alteration to generate at a position corresponding to position an amino acid from the group consisting of A, C, D, F,
H, I, K, L, M, N, P, Q, R, S, T, V, W, Y, preferably D and at a position corresponding to position 573 of S EQ I D N O : 2 th ere i s an alteration to generate at a position corresponding to position to an amino acid from the group consisting of C, D, E, F, G, H,
I, L, M, N, Q, R, S, T, V, W, Y, preferably Y, W or H, or
(b) at a position corresponding to position 492 of SEQ I D NO: 2 there is an alteration to generate G and at a position corresponding to position 573 there is an alteration to generate A or P and the polypeptide comprises an additional alteration at a position selected from the group consisting of 550, 574 and 580.
9. A polypeptide which is a variant of albumin, fragment thereof or fusion polypeptide comprising said variant albumin or a fragment thereof having an altered binding affinity to FcRn compared with the binding affinity of a parent albumin, reference albumin, fragment thereof or fusion polypeptide comprising said parent albumin, reference albumin or fragment or fusion thereof to FcRn, wherein:
(a) the polypeptide comprises alterations at two or more positions, in which at a position corresponding to position 573 of SEQ I D NO: 2 there is an alteration to generate an amino acid from the group consisting of A, C, D, E, F, G, H, I, L, M, N, Q, R, S, T, V, W, Y, preferably Y, W or H and at a position corresponding to position 574 of SEQ ID NO: 2 there is an alteration to generate an amino acid from the group consisting of A, C, D, E,
F, G, H, I, L, M, P, Q, R, S, T, V, W, Y, H, D, F, G, N, S or Y, more preferably H, D, F or
G, most preferably H, or
(b) at a position corresponding to position 573 of SEQ I D NO: 2 there is a P and at a position corresponding to position 574 of S EQ I D N O: 2 there is an N and the polypeptide comprises an additional alteration at a position selected from the group consisting of 492, 550, and 580.
10. A polypeptide which is a variant of albumin, fragment thereof or fusion polypeptide comprising said variant albumin or a fragment thereof having an altered binding affinity to FcRn compared with the binding affinity of a parent albumin, reference albumin, fragment thereof or fusion polypeptide comprising said parent albumin, reference albumin or fragment or fusion thereof to FcRn, wherein
(a) the polypeptide comprises alterations at two or more positions, in which at a position corresponding to position 573 of SEQ I D NO: 2 there is an alteration to generate an amino acid from the group consisting of A, C, D, E, F, G, H, I, L, M, N, Q, R, S, T, V, W, Y, preferably Y, W or H and at a position corresponding to position 580 of SEQ ID NO: 2 there is an alteration to generate an amino acid from the group consisting of C, D, E, F, G, H, I, L, M, N, P, R, S, T, V, W, Y, or
(b) at a position corresponding to position 573 of SEQ I D NO: 2 there is an alteration to generate a P and at a position corresponding to position 580 of SEQ I D NO: 2 there is an alteration to generate a K and the polypeptide comprises an additional alteration at a position selected from the group consisting of 492, 550, and 574.
1 1 . A polypeptide which is a variant of albumin, fragment thereof or fusion polypeptide comprising said variant albumin or a fragment thereof having an altered binding affinity to FcRn compared with the binding affinity of a parent albumin, reference albumin, fragment thereof or fusion polypeptide comprising said parent albumin, reference albumin or fragment or fusion thereof to FcRn, wherein:
(a) the polypeptide comprises alterations at two or more positions, in which at a position corresponding to position 574 of SEQ ID NO: 2 there is an alteration to generate an amino acid from the group consisting of A, C, D, E, F, G, H, I, L, M, P, Q, R, S, T, V, W, Y, H, D, F, G, N, S or Y, more preferably H, D, F or G, most preferably H and at a position corresponding to position 580 of SEQ I D NO: 2 there is an alteration to generate at a position corresponding to position to an amino acid from the group consisting of A, C, D, E, F, G, H, I, L, M, N, P, R, S, T, V, W, Y, or
(b) (at a position corresponding to position 574 of SEQ ID NO: 2 there is an alteration to generate an N and at a position corresponding to position 580 of SEQ ID NO: 2 there is an alteration to generate a K and the polypeptide comprises an additional alteration at a position selected from the group consisting of 492, 550, and 573.
12. The polypeptide according to any preceding claim, wherein the polypeptide comprises alterations at three or more positions selected from positions corresponding to positions 492, 550, 573, 574 and 580 in SEQ ID NO: 2.
13. The polypeptide according to any preceding claimwherein the alteration at the position corresponding to position 492, 550, 573, 574 and/or 580 is a substitution.
14. The polypeptide of claim 13 wherein the substitution at the position corresponding to position 492 is to G, D, F, H, M or R.
15. The polypeptide of claim 13 or 14 wherein the substitution at the position corresponding to position 492 is to G or D.
16. The polypeptide of any of claims 13 to 1 5 wherein the substitution at the position corresponding to position 492 is to G.
17. The polypeptide of any of claims 1 3 to 1 5 wherein the substitution at the position corresponding to position 492 is to D.
18. The polypeptide of any of claims 13 to 17 wherein the substitution at the position corresponding to position 550 is to K, L, M E, or R.
19. The polypeptide of any of claims 13 to 1 8 wherein the substitution at the position corresponding to position 550 is to K, L or M.
20. The polypeptide of any of claims 13 to 19 wherein the substitution at the position corresponding to position 550 is to K.
21 . The polypeptide of any of claims 1 3 to 20 wherein the alteration at the position corresponding to position 573 is a substitution to P, Y, W, H, F, T, I or V.
22. The polypeptide of any of claims 1 3 to 21 wherein the substitution at the position corresponding to position 573 is to P, Y or W.
23. The polypeptide of any of claims 13 to 22 wherein the substitution at the position corresponding to position 573 is to a P.
24. The polypeptide of any of claims 1 3 to 23 wherein the alteration at the position corresponding to position 574 is a substitution to H, G, D, F, N, S or Y.
25. The polypeptide of any of claims 13 to 24 wherein the substitution at the position corresponding to position 574 is to D, F, G or H.
26. The polypeptide of any of claims 13 to 25 wherein the substitution at the position corresponding to position 574 is to H.
27. The polypeptide of any of claims 1 3 to 26 wherein the substitution at the position corresponding to position 580 is to K or R.
28. The polypeptide of any of claims 13 to 27 wherein the substitution at the position corresponding to position 580 is to K.
29. The polypeptide according to any preceding claim comprising alterations corresponding to the following positions in SEQ I D NO: 2: 492G+573P+574H+580K (e.g. SEQ I D NO: 1 15); 492G+550K+573P+574H (e.g. SEQ ID NO: 257); 492D+550K+573P+574H (e.g. SEQ ID NO: 256); 492G+550K+573P (e.g. SEQ ID NO: 254); 492D+550K+573P (e.g. SEQ ID NO: 253); 550M+573P+580K (e.g . SEQ I D NO : 252); 550L+573 P+580 K (e.g. SEQ I D NO: 248); 550L+573P+574H (e.g. SEQ I D NO: 246); 492G+573P+580K (e.g. SEQ ID NO: 242); 492D+573P+580K (e.g. S EQ I D N O : 234 ); 573P+574H+580K (e.g. SEQ ID NO: 135); 550K+573P+580K (e.g. S EQ I D NO: 1 33); 550K+573P+574H (e.g. SEQ ID NO: 131 ); or 492D+573P+574H+580K (SEQ ID NO: 1 14).
30. The polypeptide of any of claims 1 to 29 wherein the reference albumin is HSA (SEQ ID No: 2) or a fragment thereof, or a fusion polypeptide comprising HSA or a fragment thereof, most preferably SEQ ID NO: 2.
31 . The polypeptide according to any of claims 1 to 30, having a stronger binding affinity to FcRn and/or longer plasma half-life than a parent albumin, reference albumin, fragment thereof or fusion polypeptide comprising said parent albumin, reference albumin or fragment or fusion thereof.
32. The polypeptide according any of claims 1 to 31 , wherein the sequence identity of the polypeptide to SEQ I D NO: 2 is more than 80%, preferably more than 90%, more preferred more than 95%, more preferred more than 96%, even more preferred more than 97%, more preferred more than 98% and most preferred more than 99%.
33. A fusion polypeptide comprising a polypeptide according to any of claims 1 to 32 and a fusion partner polypeptide selected from a therapeutic, prophylactic, diagnostic, imaging or other beneficial moiety.
34. A method for preparing a polypeptide which is a variant of albumin, fragment thereof or fusion polypeptide comprising said variant albumin or fragment thereof having a binding affinity to FcRn which is altered compared to the binding affinity of a reference albumin, fragment or fusion thereof to FcRn, comprising: (a) Providing a nucleic acid encoding a parent albumin having at least 80% sequence identity to SEQ ID NO: 2;
(b) Modifying the sequence of step (a), to encode a polypeptide which is a variant albumin, fragment thereof or fusion polypeptide comprising said variant albumin or fragment thereof comprising alterations at two or more positions corresponding to positions selected from 492, 550, 573, 574 and 580 in SEQ ID NO: 2, preferably as described in any of claims 1 to 1 1 ;
(c) Introducing the modified sequence of step (b) in a suitable host cell;
(d) Growing the cells in a suitable growth medium u nder condition lead ing to expression of the polypeptide; and
(e) Recovering the polypeptide from the growth medium; wherein the polypeptide has an altered binding affinity to FcRn and/or an altered plasma half-life compared with the half-life of a parent albumin, reference albumin, fragment thereof or fusion polypeptide comprising said parent albumin, reference albumin or fragment or fusion thereof.
35. The method of claim 34 wherein the substitution at the position corresponding to position 492 is a substitution to G, D, F, H, M or R, preferably G or D
36. The method of claim 35 wherein the substitution at the position corresponding to position 492 is to G.
37. The method of claim 35 wherein the substitution at the position corresponding to position 492 is to D.
38. The method of any of claims 34 to 37 wherein the substitution at the position corresponding to position 550 is to K, L, M, E or R, preferably K, L or M, most preferably K.
39. The method of any of claims 34 to 38 wherein the substitution at the position corresponding to position 573 is a substitution to P, Y, W, H, F, T, I or V, preferably P, Y, or W, most preferably P.
40. The method of any of claims 34 to 39 wherein the substitution at the position corresponding to position 574 is a substitution to H, D, F, G, N, S or Y, preferably H, D, F,or G, most preferably H.
41 . The method of any of claims any of claims 34 to 40 wherein the substitution at the position corresponding to position 580 is a substitution to K or R, most preferably R.
42. The method according to any of claims 34 to 41 comprising alterations at positions corresponding to the following positions in SEQ ID NO: 2: 492G+573P+574H+580K (e.g. SEQ ID NO: 1 15); 492G+550K+573P+574H (e.g. SEQ ID NO: 257); 492D+550K+573P+574H (e.g. SEQ ID NO: 256); 492G+550K+573P (e.g. SEQ ID NO: 254); 492D+550K+573P (e.g. SEQ ID NO: 253); 550M+573P+580K (e.g. SEQ ID NO: 252); 550L+573P+580K (e.g. SEQ ID NO: 248); 550L+573P+574 H (e.g . SEQ I D NO : 246); 492G+573 P+580 K (e.g. SEQ I D NO: 242); 492D+573 P+580 K (e.g. S EQ I D NO : 234); 573P+574H +580 K (e.g. S EQ I D NO : 1 35); 550K+573P+580K (e.g. SEQ I D NO: 1 33); 550K+573P+574H (e.g. SEQ I D NO: 1 31 ); or 492D+573P+574H+580K (SEQ ID NO: 1 14).
43. The method any of claims 34 to 42 wherein the reference albumin is HSA (SEQ ID No: 2) or a fragment thereof, or a fusion polypeptide comprising HSA or a fragment thereof, most preferably SEQ ID NO: 2.
44. The method according any of claims 34 to 43, wherein the sequence identity of the polypeptide to SEQ I D NO: 2 is more than 80%, preferably more than 90%, more preferred more than 95%, more preferred more than 96%, even more preferred more than 97%, more preferred more than 98% and most preferred more than 99%.
45. A conjugate comprising a polypeptide according to any of claims 1 to 33 or obtainable by a method according to any of claims 34 to 44 and a conjugation partner.
46. The conjugate according to claim 45 wherein the conjugation partner is a therapeutic, prophylactic, diagnostic, imaging or other beneficial moiety.
47. An associate comprising a polypeptide according to any of claims 1 to 33 or obtainable by a method according to any of claims 34 to 44 and a therapeutic, prophylactic, diagnostic, imaging or other beneficial moiety.
48. A nanoparticle or microparticle comprising a polypeptide according to any of claims 1 to 33 or obtainable by a method according to any of claims 34 to 44, a conjugate according to claim 45 or 46 or an associate according to claim 47.
49. A composition comprising a polypeptide according to any of claims 1 to 33 or obtainable by a method according to any of claims 34 to 44, a conjugate according to claim 45 or 46, an associate according to claim 47 or a nanoparticle or microparticle according claim 48, wherein the binding affinity of the polypeptide, fusion polypeptide, conjugate, associate or nanoparticle or microparticle to FcRn is stronger than the binding affinity of a composition comprising the corresponding parent albumin, reference albumin, fragment thereof or fusion polypeptide, conjugate, associate or nanoparticle or microparticle comprising said parent albumin, reference albumin or fragment or fusion thereof to FcRn.
50. A composition according to claim 49 where the binding affinity of the polypeptide, fusion polypeptide, conjugate, associate or nanoparticle or microparticle to FcRn is stronger than the binding affinity of HSA to FcRn.
51 . A composition according to claim 49 or 50, wherein the binding coefficient of the variant of the polypeptide, fusion polypeptide, conjugate, associate or nanoparticle or microparticle to FcRn is less than 0.9X KD of HSA to FcRn, more preferred less than 0.5X KD of HSA to FcRn, more preferred less than 0.1 X KD of HSA to FcRn, even more preferred less than 0.05X KD of HSA to FcRn, even more preferred less than 0.02X KD of HSA to FcRn and most preferred less than 0.01X KD of HSA to FcRn.
52. The composition according to any of claims 49 to 51 , comprising a polypeptide according to any of claims 1 to 33 or obtainable by a method according to any of claims 34 to 44, a conjugate according to claim 45 or 46, an associate according to claim 47 or a nanoparticle or microparticle according claim 48, further comprising a compound comprising an antibody binding domain (ABD) and a therapeutic, prophylactic, diagnostic, imaging or other beneficial moiety.
53. The composition according to any of claims 49 to 52, comprising a pharmaceutically acceptable carrier or excipient.
54. Use of a polypeptide according to any of claims 1 to 33 or obtainable by a method according to any of claims 34 to 46, a conjugate according to claim 45 or 46, an associate according to claim 47 or a nanoparticle or microparticle according claim 48 or a composition according to any of claims 49 to 53 to alter the binding affinity to FcRn or half-life, preferably in plasma, of a therapeutic, prophylactic, diagnostic, imaging or other beneficial moiety.
55. The use according to claim 54 wherein the binding affinity to FcRn is increased relative to the binding affinity of a reference comprising or consisting of HSA (SEQ I D NO: 2) or a fragment, fusion, conjugate, associate, nanoparticle or microparticle thereof to FcRn.
56. The use according to claim 54 wherein the binding affinity to FcRn is decreased relative to the binding affinity of a reference comprising or consisting of HSA (SEQ I D NO: 2) or a fragment, fusion, conjugate, associate, nanoparticle or microparticle thereof to FcRn.
57. A method for altering the binding affinity to FcRn or half-life preferably in plasma, of a molecule comprising:
(a) where the molecule is a polypeptide, fusing or conjugating the molecule to a polypeptide according to any of claims 1 to 33 or obtainable by a method of claims 34 to 44, or to a conjugate according to claim 45 or 46; associating the molecule to a polypeptide according to any of claims 1 to 33 or obtainable by a method of claims 34 to 44 or to a conjugate according to claim 45 or 46; incorporating the molecule in an associate according to claim 47, in nanoparticle or microparticle according to claim 48 or a composition according to any of claims 49 to 53;
(b) where the molecule is not a polypeptide, conjugating the molecule to a polypeptide according to any of claims 1 to 33 or obtainable by a method of claims 34 to 44, or to a conjugate according to claim 45 or 46; associating the molecule to a polypeptide according to any of claims 1 to 33 or obtainable by a method of claims 34 to 44 or to a conjugate according to claim 45 or 46; incorporating the molecule in an associate according to claim 47, i n nanoparticle or microparticle according to claim 48 or a composition according to any of claims 49 to 53.
58. A method according to claim 57 wherein the molecule is a therapeutic, prophylactic, diagnostic, imaging or other beneficial moiety.
59. A polypeptide, fusion polypeptide, conjugate, associate, nanoparticle or microparticle or composition thereof according to any of claims 1 to 33 or 45 to 53 or obtainable by the method of claims 34 to 44 wherein the polypeptide, fusion polypeptide, conjugate, associate, nanoparticle or microparticle or composition comprises one or more (several) moiety selected from those described herein.
60. A nucleic acid encoding the polypeptide or fusion polypeptide of any of claims 1 to 33.
61 . A vector comprising a nucleic acid according to claim 60.
62. A host cell comprising a nucleic acid according to claim 60 or a vector according to claim 61 .
63. A host cell according to claim 62 wherein the host cell is a eukaryote, preferably a yeast (such as Saccharomyces cerevisiae) or a mammalian cell (such as CHO or HEK) or a plant cell (such as rice).
64. A method of prophylaxis, treatment or diagnosis comprising administering a polypeptide, fusion polypeptide, conjugate, composition, associate, nanoparticle or microparticle or polynucleotide according to any of claims 1 to 33 or 45 to 53 or obtainable by the method of any of claims 34 to 44 to a subject.
PCT/EP2013/073426 2012-11-08 2013-11-08 Albumin variants WO2014072481A1 (en)

Priority Applications (11)

Application Number Priority Date Filing Date Title
EP13789000.0A EP2917233A1 (en) 2012-11-08 2013-11-08 Albumin variants
CA2890766A CA2890766A1 (en) 2012-11-08 2013-11-08 Albumin variants
CN201380069331.4A CN105452290A (en) 2012-11-08 2013-11-08 Albumin variants
RU2015121693A RU2670063C2 (en) 2012-11-08 2013-11-08 Albumin variants
KR1020157014474A KR20150082422A (en) 2012-11-08 2013-11-08 Albumin variants
JP2015541165A JP6487328B2 (en) 2012-11-08 2013-11-08 Albumin variant
AU2013343503A AU2013343503B2 (en) 2012-11-08 2013-11-08 Albumin variants
MX2015005363A MX2015005363A (en) 2012-11-08 2013-11-08 Albumin variants.
BR112015010318A BR112015010318A2 (en) 2012-11-08 2013-11-08 ALBUMIN VARIANTS
IL238185A IL238185B (en) 2012-11-08 2015-04-12 Albumin variants
AU2018201136A AU2018201136A1 (en) 2012-11-08 2018-02-16 Albumin variants

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP12191856 2012-11-08
EP12191856.9 2012-11-08

Publications (1)

Publication Number Publication Date
WO2014072481A1 true WO2014072481A1 (en) 2014-05-15

Family

ID=47137625

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2013/073426 WO2014072481A1 (en) 2012-11-08 2013-11-08 Albumin variants

Country Status (13)

Country Link
US (3) US20140128326A1 (en)
EP (1) EP2917233A1 (en)
JP (2) JP6487328B2 (en)
KR (1) KR20150082422A (en)
CN (1) CN105452290A (en)
AU (2) AU2013343503B2 (en)
BR (1) BR112015010318A2 (en)
CA (1) CA2890766A1 (en)
GB (1) GB2512156A (en)
IL (1) IL238185B (en)
MX (1) MX2015005363A (en)
RU (1) RU2670063C2 (en)
WO (1) WO2014072481A1 (en)

Cited By (63)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB2512156A (en) * 2012-11-08 2014-09-24 Novozymes Biopharma Dk As Albumin variants
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
US8999380B2 (en) 2012-04-02 2015-04-07 Moderna Therapeutics, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
US9107886B2 (en) 2012-04-02 2015-08-18 Moderna Therapeutics, Inc. Modified polynucleotides encoding basic helix-loop-helix family member E41
US9181319B2 (en) 2010-08-06 2015-11-10 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9186372B2 (en) 2011-12-16 2015-11-17 Moderna Therapeutics, Inc. Split dose administration
WO2016016859A1 (en) 2014-07-31 2016-02-04 Amgen Research (Munich) Gmbh Optimized cross-species specific bispecific single chain antibody constructs
WO2016016412A1 (en) 2014-07-31 2016-02-04 Amgen Research (Munich) Gmbh Antibody constructs for cdh19 and cd3
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
US9334328B2 (en) 2010-10-01 2016-05-10 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9428535B2 (en) 2011-10-03 2016-08-30 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
WO2016166360A1 (en) 2015-04-17 2016-10-20 Bayer Pharma Aktiengesellschaft Bispecific antibody constructs for cdh3 and cd3
US9533047B2 (en) 2011-03-31 2017-01-03 Modernatx, Inc. Delivery and formulation of engineered nucleic acids
WO2017021356A1 (en) 2015-07-31 2017-02-09 Amgen Research (Munich) Gmbh Bispecific antibody constructs binding mesothelin and cd3
WO2017021354A1 (en) 2015-07-31 2017-02-09 Amgen Research (Munich) Gmbh Antibody constructs for cd70 and cd3
WO2017021370A1 (en) 2015-07-31 2017-02-09 Amgen Research (Munich) Gmbh Bispecific antibody constructs binding egfrviii and cd3
WO2017021349A1 (en) 2015-07-31 2017-02-09 Amgen Research (Munich) Gmbh Bispecific antibody constructs binding dll3 and cd3
WO2017021362A1 (en) 2015-07-31 2017-02-09 Amgen Research (Munich) Gmbh Antibody constructs for flt3 and cd3
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9597380B2 (en) 2012-11-26 2017-03-21 Modernatx, Inc. Terminally modified RNA
WO2017112847A1 (en) 2015-12-22 2017-06-29 Albumedix A/S Improved protein expression strains
WO2017117631A1 (en) 2016-01-07 2017-07-13 Csl Limited Mutated truncated von willebrand factor
WO2017151957A1 (en) 2016-03-02 2017-09-08 Novozymes A/S Cellobiohydrolase variants and polynucleotides encoding same
WO2017165760A1 (en) 2016-03-24 2017-09-28 Novozymes A/S Cellobiohydrolase variants and polynucleotides encoding same
WO2017182427A1 (en) 2016-04-19 2017-10-26 Amgen Research (Munich) Gmbh Administration of a bispecific construct binding to cd33 and cd3 for use in a method for the treatment of myeloid leukemia
US9944691B2 (en) 2012-03-16 2018-04-17 Albumedix A/S Albumin variants
WO2018082758A1 (en) 2016-11-04 2018-05-11 Aarhus Universitet Identification and treatment of tumors characterized by an overexpression of the neonatal fc receptor
WO2018094181A1 (en) 2016-11-21 2018-05-24 Novozymes A/S Yeast cell extract assisted construction of dna molecules
JP2018527923A (en) * 2015-08-20 2018-09-27 ノボザイムス バイオファーマ デーコー アクティーゼルスカブ Albumin variants and conjugates
WO2018234518A1 (en) 2017-06-22 2018-12-27 CSL Behring Lengnau AG Modulation of fviii immunogenicity by truncated vwf
WO2018234349A1 (en) 2017-06-20 2018-12-27 Albumedix Ltd Improved protein expression strains
US10233228B2 (en) 2010-04-09 2019-03-19 Albumedix Ltd Albumin derivatives and variants
WO2019104385A1 (en) 2017-11-29 2019-06-06 Csl Limited Method of treating or preventing ischemia-reperfusion injury
US10323076B2 (en) 2013-10-03 2019-06-18 Modernatx, Inc. Polynucleotides encoding low density lipoprotein receptor
WO2019140196A1 (en) 2018-01-12 2019-07-18 Amgen Inc. Anti-pd-1 antibodies and methods of treatment
WO2019218009A1 (en) 2018-05-16 2019-11-21 Csl Limited Soluble complement receptor type 1 variants and uses thereof
WO2020025532A1 (en) 2018-07-30 2020-02-06 Amgen Research (Munich) Gmbh Prolonged administration of a bispecific antibody construct binding to cd33 and cd3
WO2020025792A1 (en) 2018-08-03 2020-02-06 Amgen Research (Munich) Gmbh Antibody constructs for cldn18.2 and cd3
WO2020049151A1 (en) 2018-09-06 2020-03-12 Bavarian Nordic A/S Storage improved poxvirus compositions
US10640504B2 (en) 2017-09-08 2020-05-05 Amgen Inc. Inhibitors of KRAS G12C and methods of using the same
US10696732B2 (en) 2009-10-30 2020-06-30 Albumedix, Ltd Albumin variants
US10711050B2 (en) 2011-11-18 2020-07-14 Albumedix Ltd Variant serum albumin with improved half-life and other properties
US10815291B2 (en) 2013-09-30 2020-10-27 Modernatx, Inc. Polynucleotides encoding immune modulating polypeptides
WO2021044362A1 (en) 2019-09-06 2021-03-11 Novartis Ag Therapeutic fusion proteins
EP3819007A1 (en) 2019-11-11 2021-05-12 Amgen Research (Munich) GmbH Dosing regimen for anti-bcma agents
WO2021111143A1 (en) 2019-12-04 2021-06-10 Albumedix Limited Methods and compositions produced thereby
EP3842451A1 (en) * 2015-03-12 2021-06-30 MedImmune, LLC Method of purifying albumin-fusion proteins
WO2021150824A1 (en) 2020-01-22 2021-07-29 Amgen Research (Munich) Gmbh Combinations of antibody constructs and inhibitors of cytokine release syndrome and uses thereof
WO2021180943A1 (en) 2020-03-12 2021-09-16 Bavarian Nordic A/S Compositions improving poxvirus stability
WO2021183861A1 (en) 2020-03-12 2021-09-16 Amgen Inc. Method for treatment and prophylaxis of crs in patients comprising a combination of bispecifc antibodies binding to cds x cancer cell and tnfalpha or il-6 inhibitor
WO2021236638A1 (en) 2020-05-19 2021-11-25 Amgen Inc. Mageb2 binding constructs
WO2021243320A2 (en) 2020-05-29 2021-12-02 Amgen Inc. Adverse effects-mitigating administration of a bispecific antibody construct binding to cd33 and cd3
WO2022060901A1 (en) 2020-09-16 2022-03-24 Amgen Inc. Methods for administering therapeutic doses of bispecific t-cell engaging molecules for the treatment of cancer
WO2022096698A1 (en) 2020-11-06 2022-05-12 Amgen Inc. Polypeptide constructs binding to cd3
WO2022096700A1 (en) 2020-11-06 2022-05-12 Amgen Research (Munich) Gmbh Polypeptide constructs selectively binding to cldn6 and cd3
WO2022096704A1 (en) 2020-11-06 2022-05-12 Amgen Inc. Antigen binding domain with reduced clipping rate
WO2022103781A1 (en) 2020-11-10 2022-05-19 Amgen Inc. Methods for administering a bcma x cd3 binding molecule
US11434302B2 (en) 2016-02-03 2022-09-06 Amgen Research (Munich) Gmbh Bispecific T cell engaging antibody constructs
US11541103B2 (en) 2017-08-03 2023-01-03 Amgen Inc. Interleukin-21 mutein/ anti-PD-1 antibody conjugates
US11555062B2 (en) 2011-10-11 2023-01-17 Viela Bio, Inc. Methods of administering a composition comprising a Tn3 scaffold and a CD40L-specific monomer
US11555061B2 (en) 2009-02-11 2023-01-17 Albumedix, Ltd Albumin variants and conjugates
WO2024077044A1 (en) 2022-10-05 2024-04-11 Amgen Inc. Combination therapies comprising t-cell redirecting therapies and agonistic anti-il-2r antibodies or fragments thereof

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014138371A1 (en) * 2013-03-06 2014-09-12 Glaxosmithkline Llc Host cells and methods of use
WO2017210684A1 (en) * 2016-06-03 2017-12-07 New York University Methods and reagents for modulating macrophage phenotype
CN110573529A (en) * 2017-04-05 2019-12-13 韩国生命工学研究院 Fusion protein activating NK cells, NK cells and pharmaceutical composition comprising the same
CN109553684A (en) * 2017-09-25 2019-04-02 中国科学院过程工程研究所 A kind of nano-carrier albumen and its preparation method and application
GB202004514D0 (en) 2020-03-27 2020-05-13 Inst De Medicina Molecular Joaeo Lobo Antunes Treatment of Immunosuppressive Cancer
CN113717873B (en) * 2021-09-27 2023-04-14 四川大学 Multiple-tolerance saccharomyces cerevisiae strain and construction method and application thereof
CN113912730B (en) * 2021-12-14 2022-03-04 北京科诺信诚科技有限公司 Sustained-release anti-FcRn antibody or antigen-binding fragment and application thereof

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011051489A2 (en) * 2009-10-30 2011-05-05 Novozymes Biopharma Dk A/S Albumin variants
WO2011124718A1 (en) * 2010-04-09 2011-10-13 Novozymes A/S Albumin derivatives and variants
WO2012059486A1 (en) * 2010-11-01 2012-05-10 Novozymes Biopharma Dk A/S Albumin variants
WO2012150319A1 (en) * 2011-05-05 2012-11-08 Novozymes Biopharma Dk A/S Albumin variants

Family Cites Families (162)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5625041A (en) 1990-09-12 1997-04-29 Delta Biotechnology Limited Purification of proteins
GB9019919D0 (en) 1990-09-12 1990-10-24 Delta Biotechnology Ltd Purification of proteins
US2714586A (en) 1951-06-25 1955-08-02 Phillips Petroleum Co Washing urea and thiourea containing adducts
JPS4983148A (en) 1972-12-16 1974-08-09
US4302386A (en) 1978-08-25 1981-11-24 The Ohio State University Antigenic modification of polypeptides
US4741900A (en) 1982-11-16 1988-05-03 Cytogen Corporation Antibody-metal ion complexes
US4757006A (en) 1983-10-28 1988-07-12 Genetics Institute, Inc. Human factor VIII:C gene and recombinant methods for production
JPS60169498A (en) 1984-02-10 1985-09-02 Kyowa Hakko Kogyo Co Ltd Adult t cell leukemic viral antigen peptide derivative
DE3683980D1 (en) 1985-04-12 1992-04-02 Genetics Inst NEW PROCOAGULATION PROTEINS.
GR860984B (en) 1985-04-17 1986-08-18 Zymogenetics Inc Expression of factor vii and ix activities in mammalian cells
HU213344B (en) 1987-04-09 1997-05-28 Delta Biotechnology Ltd Process for producing of stable dezintegrating plasmid-vectors
EP0305216B1 (en) 1987-08-28 1995-08-02 Novo Nordisk A/S Recombinant Humicola lipase and process for the production of recombinant humicola lipases
GB8725529D0 (en) 1987-10-30 1987-12-02 Delta Biotechnology Ltd Polypeptides
IL88326A (en) 1987-11-18 1993-03-15 Gist Brocades Nv Purification of serum albumin
US5075222A (en) 1988-05-27 1991-12-24 Synergen, Inc. Interleukin-1 inhibitors
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
GB8909916D0 (en) 1989-04-29 1989-06-14 Delta Biotechnology Ltd Polypeptides
US5766883A (en) 1989-04-29 1998-06-16 Delta Biotechnology Limited Polypeptides
FR2650598B1 (en) 1989-08-03 1994-06-03 Rhone Poulenc Sante DERIVATIVES OF ALBUMIN WITH THERAPEUTIC FUNCTION
US5073627A (en) 1989-08-22 1991-12-17 Immunex Corporation Fusion proteins comprising GM-CSF and IL-3
US5208020A (en) 1989-10-25 1993-05-04 Immunogen Inc. Cytotoxic agents comprising maytansinoids and their therapeutic use
GB8927722D0 (en) 1989-12-07 1990-02-07 British Bio Technology Proteins and nucleic acids
DE4000939A1 (en) 1990-01-15 1991-07-18 Brem Gottfried Prof Dr Dr METHOD FOR OBTAINING ANTIBODIES
JP3230091B2 (en) 1990-06-25 2001-11-19 ウェルファイド株式会社 Method for suppressing coloration of human serum albumin
IL99552A0 (en) 1990-09-28 1992-08-18 Ixsys Inc Compositions containing procaryotic cells,a kit for the preparation of vectors useful for the coexpression of two or more dna sequences and methods for the use thereof
US5698426A (en) 1990-09-28 1997-12-16 Ixsys, Incorporated Surface expression libraries of heteromeric receptors
CA2058820C (en) 1991-04-25 2003-07-15 Kotikanyad Sreekrishna Expression cassettes and vectors for the secretion of human serum albumin in pichia pastoris cells
US5264586A (en) 1991-07-17 1993-11-23 The Scripps Research Institute Analogs of calicheamicin gamma1I, method of making and using the same
FR2686899B1 (en) 1992-01-31 1995-09-01 Rhone Poulenc Rorer Sa NOVEL BIOLOGICALLY ACTIVE POLYPEPTIDES, THEIR PREPARATION AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM.
US5663060A (en) 1992-04-07 1997-09-02 Emory University Hybrid human/animal factor VIII
ZA932522B (en) 1992-04-10 1993-12-20 Res Dev Foundation Immunotoxins directed against c-erbB-2(HER/neu) related surface antigens
DE4226971C2 (en) 1992-08-14 1997-01-16 Widmar Prof Dr Tanner Modified fungal cells and processes for the production of recombinant products
US5728553A (en) 1992-09-23 1998-03-17 Delta Biotechnology Limited High purity albumin and method of producing
CA2149329C (en) 1992-11-13 2008-07-15 Darrell R. Anderson Therapeutic application of chimeric and radiolabeled antibodies to human b lymphocyte restricted differentiation antigen for treatment of b cell lymphoma
EP0710283A4 (en) 1993-07-22 1997-07-02 Merck & Co Inc EXPRESSION OF HUMAN INTERLEUKIN-1-g(b) IN A TRANSGENIC ANIMAL
DE4343591A1 (en) 1993-12-21 1995-06-22 Evotec Biosystems Gmbh Process for the evolutionary design and synthesis of functional polymers based on shape elements and shape codes
US6811773B1 (en) 1993-12-22 2004-11-02 Human Genome Sciences, Inc. Human monocyte colony inhibitory factor (M-CIF) polypeptides
US5605793A (en) 1994-02-17 1997-02-25 Affymax Technologies N.V. Methods for in vitro recombination
GB9404270D0 (en) 1994-03-05 1994-04-20 Delta Biotechnology Ltd Yeast strains and modified albumins
US5773001A (en) 1994-06-03 1998-06-30 American Cyanamid Company Conjugates of methyltrithio antitumor agents and intermediates for their synthesis
US7597886B2 (en) 1994-11-07 2009-10-06 Human Genome Sciences, Inc. Tumor necrosis factor-gamma
US5712374A (en) 1995-06-07 1998-01-27 American Cyanamid Company Method for the preparation of substantiallly monomeric calicheamicin derivative/carrier conjugates
US5714586A (en) 1995-06-07 1998-02-03 American Cyanamid Company Methods for the preparation of monomeric calicheamicin derivative/carrier conjugates
US5955349A (en) 1996-08-26 1999-09-21 Zymogenetics, Inc. Compositions and methods for producing heterologous polypeptides in Pichia methanolica
US5716808A (en) 1995-11-09 1998-02-10 Zymogenetics, Inc. Genetic engineering of pichia methanolica
GB9526733D0 (en) 1995-12-30 1996-02-28 Delta Biotechnology Ltd Fusion proteins
US6509313B1 (en) 1996-02-28 2003-01-21 Cornell Research Foundation, Inc. Stimulation of immune response with low doses of cytokines
US5854039A (en) 1996-07-17 1998-12-29 Zymogenetics, Inc. Transformation of pichia methanolica
US5736383A (en) 1996-08-26 1998-04-07 Zymogenetics, Inc. Preparation of Pichia methanolica auxotrophic mutants
US6274305B1 (en) 1996-12-19 2001-08-14 Tufts University Inhibiting proliferation of cancer cells
US6605699B1 (en) 1997-01-21 2003-08-12 Human Genome Sciences, Inc. Galectin-11 polypeptides
US7196164B2 (en) 1997-07-08 2007-03-27 Human Genome Sciences, Inc. Secreted protein HHTLF25
US7053190B2 (en) 1997-03-07 2006-05-30 Human Genome Sciences, Inc. Secreted protein HRGDF73
US6506569B1 (en) 1997-05-30 2003-01-14 Human Genome Sciences, Inc. Antibodies to human tumor necrosis factor receptor TR10
US5948609A (en) 1997-12-03 1999-09-07 Carter; Daniel C. Oxygen-transporting albumin-based blood replacement composition and blood volume expander
CA2306554A1 (en) 1998-08-06 2000-02-17 Jin Po Lee Uric acid assay device with stabilized uricase reagent composition
AU3128000A (en) 1998-12-23 2000-07-31 Human Genome Sciences, Inc. Peptidoglycan recognition proteins
GB9902000D0 (en) 1999-01-30 1999-03-17 Delta Biotechnology Ltd Process
ATE443714T1 (en) 1999-05-17 2009-10-15 Conjuchem Biotechnologies Inc LONG-ACTING PEPTIDE INHIBITORS OF VIRUS FUSION WITH BODY CELLS IN VIRAL INFECTIONS
WO2000071079A2 (en) 1999-05-21 2000-11-30 American Bioscience, Inc. Protein stabilized pharmacologically active agents, methods for the preparation thereof and methods for the use thereof
US7572619B2 (en) 2000-03-22 2009-08-11 Octagene Gmbh Recombinant blood clotting factors
EP1276849A4 (en) 2000-04-12 2004-06-09 Human Genome Sciences Inc Albumin fusion proteins
US20030091565A1 (en) 2000-08-18 2003-05-15 Beltzer James P. Binding polypeptides and methods based thereon
WO2002022809A2 (en) 2000-09-15 2002-03-21 Coley Pharmaceutical Gmbh PROCESS FOR HIGH THROUGHPUT SCREENING OF CpG-BASED IMMUNO-AGONIST/ANTAGONIST
US7615537B2 (en) 2000-10-25 2009-11-10 Genzyme Corporation Methods for treating blood coagulation disorders
WO2002043658A2 (en) 2000-11-06 2002-06-06 The Jackson Laboratory Fcrn-based therapeutics for the treatment of auto-immune disorders
SI1724284T1 (en) 2000-12-07 2009-12-31 Lilly Co Eli GLP-1 fusion proteins
US7175988B2 (en) 2001-02-09 2007-02-13 Human Genome Sciences, Inc. Human G-protein Chemokine Receptor (CCR5) HDGNR10
US7507413B2 (en) 2001-04-12 2009-03-24 Human Genome Sciences, Inc. Albumin fusion proteins
DE60236646D1 (en) 2001-04-13 2010-07-22 Human Genome Sciences Inc Anti-VEGF-2 antibodies
AUPR446701A0 (en) 2001-04-18 2001-05-17 Gene Stream Pty Ltd Transgenic mammals for pharmacological and toxicological studies
EP1406917A4 (en) 2001-06-15 2007-11-14 New Century Pharmaceuticals Human albumin animal models for drug evaluation, toxicology and immunogenicity studies
EP1421187B1 (en) 2001-07-27 2007-10-10 THE GOVERNMENT OF THE UNITED STATES OF AMERICA, as represented by THE SECRETARY, DEPARTMENT OF HEALTH AND HUMAN SERVICES Systems for in vivo site-directed mutagenesis using oligonucleotides
CN1405182A (en) 2001-08-10 2003-03-26 中国人民解放军军事医学科学院生物工程研究所 Serum albumin and granulocyte colony stimulating factor fusion protein
EP2277889B1 (en) 2001-12-21 2014-07-09 Human Genome Sciences, Inc. Fusion proteins of albumin and interferon beta
US20080167238A1 (en) 2001-12-21 2008-07-10 Human Genome Sciences, Inc. Albumin Fusion Proteins
EP1463752A4 (en) 2001-12-21 2005-07-13 Human Genome Sciences Inc Albumin fusion proteins
KR20040089608A (en) 2002-02-07 2004-10-21 델타 바이오테크놀로지 리미티드 HIV Inhibiting Proteins
AU2003210806A1 (en) 2002-03-05 2003-09-22 Eli Lilly And Company Heterologous g-csf fusion proteins
US6995933B1 (en) 2002-04-30 2006-02-07 Western Digital Technologies, Inc. Disk drive for optimizing write current settings relative to drive operating characteristics and ambient temperature readings
AU2003240822A1 (en) 2002-05-30 2003-12-19 Human Genome Sciences, Inc. Antibodies that specifically bind to neurokinin b
CN1241946C (en) 2002-07-01 2006-02-15 美国福源集团 Human serum albumins recombined merge protein having hyperplasia stimulation function to multiple cells
GB0217347D0 (en) 2002-07-26 2002-09-04 Univ Edinburgh Novel albumins
EP1572955A2 (en) 2002-08-02 2005-09-14 Human Genome Sciences, Inc. Antibodies against c3a receptor
US7749713B2 (en) 2002-11-19 2010-07-06 Hasan Kulaksiz Diagnostic method for diseases by screening for hepcidin in human or animal tissues, blood or body fluids and therapeutic uses therefor
CA2513213C (en) 2003-01-22 2013-07-30 Human Genome Sciences, Inc. Albumin fusion proteins
DE602004017290D1 (en) 2003-02-17 2008-12-04 Novozymes Biopharma Uk Ltd CONJUGATES FOR MEDICAL FIGURES INCLUDING CARRIER, TARGETING UNITS AND CONTRAST
GB0305989D0 (en) 2003-03-15 2003-04-23 Delta Biotechnology Ltd Agent
US20070041987A1 (en) 2003-03-19 2007-02-22 Daniel Carter Fragments or polymers of albumin with tunable vascular residence time for use in therapeutic delivery and vaccine development
CA2522680A1 (en) 2003-04-15 2004-10-28 Xenon Pharmaceuticals Inc. Juvenile hemochromatosis gene (hfe2a), expression products and uses thereof
WO2004101620A2 (en) 2003-05-01 2004-11-25 Compound Therapeutics, Inc. Serum albumin scaffold-based proteins and uses thereof
WO2005007121A2 (en) 2003-07-18 2005-01-27 Massachusetts Institute Of Technology Mutant interleukin-2(il-2) polypeptides
WO2005082423A2 (en) 2003-11-18 2005-09-09 Beth Israel Deaconess Medical Center Serum albumin conjugated to fluorescent substances for imaging
GB0329722D0 (en) 2003-12-23 2004-01-28 Delta Biotechnology Ltd Modified plasmid and use thereof
GB0329681D0 (en) 2003-12-23 2004-01-28 Delta Biotechnology Ltd Gene expression technique
CA2486245C (en) 2003-12-26 2013-01-08 Nipro Corporation Albumin having enhanced antimicrobial activity
JP4649954B2 (en) 2003-12-26 2011-03-16 ニプロ株式会社 Albumin with enhanced antibacterial activity
KR100671005B1 (en) 2004-01-15 2007-01-18 고려대학교 산학협력단 Biomarker proteins for diagnosing the exposure to PAH
BRPI0507026A (en) 2004-02-09 2007-04-17 Human Genome Sciences Inc albumin fusion proteins
RU2369404C2 (en) * 2004-02-09 2009-10-10 Хьюман Дженом Сайенсиз, Инк. Fused proteins of albumine
JP4492156B2 (en) 2004-03-03 2010-06-30 ニプロ株式会社 Protein containing serum albumin domain
US20060018859A1 (en) 2004-07-16 2006-01-26 Carter Daniel C Modified human serum albumin with reduced or eliminated affinity to chemical or biological contaminants at Cys 34
US20060051859A1 (en) 2004-09-09 2006-03-09 Yan Fu Long acting human interferon analogs
EP1791565B1 (en) 2004-09-23 2016-04-20 Genentech, Inc. Cysteine engineered antibodies and conjugates
WO2006066595A2 (en) 2004-12-22 2006-06-29 Novozymes A/S Recombinant production of serum albumin
AU2005317828A1 (en) 2004-12-23 2006-06-29 Novozymes Delta Limited Gene expression technique
WO2006073195A1 (en) 2005-01-07 2006-07-13 Biomarker Science Co., Ltd Method for predicting or diagnosing diabetes and kit for predicting or diagnosing diabetes
US20060178301A1 (en) 2005-02-04 2006-08-10 Mathias Jurs Albumin-fused ciliary neurotrophic factor
WO2006118772A2 (en) 2005-04-29 2006-11-09 The Jackson Laboratory Fcrn antibodies and uses thereof
GB0512707D0 (en) 2005-06-22 2005-07-27 Delta Biotechnology Ltd Gene expression technique
KR20080071119A (en) 2005-08-12 2008-08-01 휴먼 게놈 사이언시즈, 인코포레이티드 Albumin fusion proteins
US8008257B2 (en) 2005-10-20 2011-08-30 University Of Ottawa Heart Institute ANF fusion proteins
AU2006329215A1 (en) 2005-12-22 2007-06-28 Conjuchem Biotechnologies Inc. Process for the production of preformed conjugates of albumin and a therapeutic agent
EP1816201A1 (en) 2006-02-06 2007-08-08 CSL Behring GmbH Modified coagulation factor VIIa with extended half-life
WO2007112940A2 (en) 2006-03-31 2007-10-11 Ablynx N.V. Albumin-derived amino acid sequence, use thereof for increasing the half-life of therapeutic proteins and of other therapeutic compounds and entities, and constructs comprising the same
AU2007258609B2 (en) 2006-06-07 2013-01-24 Human Genome Sciences, Inc. Albumin fusion proteins
EP3896090B1 (en) 2006-06-14 2022-01-12 CSL Behring GmbH Proteolytically cleavable fusion protein comprising a blood coagulation factor
CA2657277C (en) 2006-07-13 2015-11-24 Upperton Limited Process for preparing particles of proteinaceous material
JP4983148B2 (en) 2006-08-18 2012-07-25 ニプロ株式会社 Glycan-containing albumin, method for producing the same, and use thereof
EP2615108B1 (en) 2006-09-08 2016-10-26 Ambrx, Inc. Modified human plasma polypeptide or fc scaffolds and thier uses
US20100129846A1 (en) 2006-12-07 2010-05-27 Power3 Medical Products, Inc. Isoform specificities of blood serum proteins and their use as differentially expressed protein biomarkers for diagnosis of breast cancer
EP2604623A3 (en) 2007-08-08 2013-10-02 Novozymes Biopharma DK A/S Transferrin variants and conjugates
US8906356B2 (en) 2007-11-05 2014-12-09 Massachusetts Institute Of Technology Mutant interleukin-2 (IL-2) polypeptides
CA2611540C (en) 2007-11-09 2017-05-30 Nipro Corporation Sugar chain-containing albumin as a drug carrier to the liver
RU2490278C2 (en) 2007-12-21 2013-08-20 Медиммун Лимитед ELEMENT BOUND WITH INTERLEUKIN-4 RECEPTOR α (IL-4Rα)-173
EP2288715B1 (en) 2008-04-11 2014-09-24 Merrimack Pharmaceuticals, Inc. Human serum albumin linkers and conjugates thereof
AU2009243187C1 (en) 2008-04-28 2015-12-24 President And Fellows Of Harvard College Supercharged proteins for cell penetration
CA2731216A1 (en) 2008-07-18 2010-01-21 Oragenics, Inc. Compositions for the detection and treatment of colorectal cancer
WO2010059315A1 (en) 2008-11-18 2010-05-27 Merrimack Pharmaceuticals, Inc. Human serum albumin linkers and conjugates thereof
WO2010065950A2 (en) 2008-12-05 2010-06-10 Abraxis Bioscience, Llc Albumin binding peptide-mediated disease targeting
WO2010068278A2 (en) 2008-12-10 2010-06-17 The Scripps Research Institute Production of carrier-peptide conjugates using chemically reactive unnatural amino acids
JP5936112B2 (en) 2009-02-11 2016-06-15 アルブミディクス アクティーゼルスカブ Albumin variants and complexes
US20120076728A1 (en) 2009-04-08 2012-03-29 The Regents Of The University Of California Human protein scaffold with controlled serum pharmacokinetics
US9221886B2 (en) 2009-04-28 2015-12-29 President And Fellows Of Harvard College Supercharged proteins for cell penetration
WO2010138814A2 (en) 2009-05-29 2010-12-02 The Brigham And Women's Hospital, Inc. Disrupting fcrn-albumin interactions
WO2010141329A1 (en) 2009-06-01 2010-12-09 Medimmune, Llc Molecules with extended half-lives and uses thereof
KR101286721B1 (en) 2009-06-05 2013-07-16 한국과학기술연구원 Recombinant albumins fused with poly-cysteine peptide and the methods for preparing the same
NZ597580A (en) 2009-07-20 2013-11-29 Univ Nat Taiwan Polypeptides selective for alpha.v.beta.3 integrin conjugated with a variant of human serum albumin (hsa) and pharmaceutical uses thereof
EP2456787A4 (en) 2009-07-24 2013-01-30 Univ Leland Stanford Junior Cytokine compositions and methods of use thereof
CA2770617C (en) 2009-08-10 2018-02-20 Mark Smith Reversible covalent linkage of functional molecules
UY32920A (en) 2009-10-02 2011-04-29 Boehringer Ingelheim Int BISPECIFIC UNION MOLECULES FOR ANTI-ANGIOGENESIS THERAPY
US20130064788A1 (en) 2009-10-10 2013-03-14 Eleven Biotherapeutics, Inc. Il-17 family cytokine compositions and uses
RU2012131251A (en) 2009-12-23 2014-01-27 Нэйшнл Ченг Кунг Юниверсити COMPOSITIONS AND METHODS FOR THE TREATMENT OF EYE DISEASES RELATED TO ANGIOGENESIS
CN101875693B (en) 2010-01-22 2012-07-18 成都正能生物技术有限责任公司 Albumin variant having anti-angiogenesis activity and preparation method thereof
CN104610454A (en) 2010-02-16 2015-05-13 米迪缪尼有限公司 HSA-related compositions and methods of use
US9238080B2 (en) 2010-05-21 2016-01-19 Merrimack Pharmaceuticals, Inc. Bi-specific fusion proteins
WO2011161127A1 (en) 2010-06-21 2011-12-29 Medimmune, Llc Protease variants of human neprilysin
EP2603522A1 (en) 2010-08-13 2013-06-19 GlaxoSmithKline Intellectual Property Development Limited Improved anti-serum albumin binding variants
US9045564B2 (en) 2011-02-15 2015-06-02 Medimmune, Llc HSA-related compositions and methods of use
EP2675471A4 (en) * 2011-02-15 2015-01-28 Medimmune Llc Hsa-related compositions and methods of use
JP5641536B2 (en) * 2011-03-15 2014-12-17 日本パーカライジング株式会社 Electrodeposition solution for fixed abrasive saw wire
KR20140053991A (en) 2011-07-18 2014-05-08 아츠 바이올로직스 에이/에스 Long acting luteinizing hormone (lh) compound
WO2013075066A2 (en) 2011-11-18 2013-05-23 Eleven Biotherapeutics, Inc. Proteins with improved half-life and other properties
BR112014018679A2 (en) * 2012-03-16 2017-07-04 Novozymes Biopharma Dk As albumin variants
WO2014005596A1 (en) 2012-07-03 2014-01-09 Aarhus Universitet Modified payload molecules and their interactions and uses
US8757798B2 (en) 2012-08-31 2014-06-24 Cody Air LLC Eyewear frame
MX2015005363A (en) * 2012-11-08 2015-11-06 Novozymes Biopharma Dk As Albumin variants.
EP3318124A3 (en) 2013-02-16 2018-05-30 Albumedix A/S Pharmacokinetic animal model
EP2992329A1 (en) 2013-05-03 2016-03-09 Eleven Biotherapeutics, Inc. Albumin variants binding to fcrn
WO2015036579A1 (en) 2013-09-13 2015-03-19 Novozymes Biopharma Dk A/S Albumin variants
JP6306700B2 (en) 2013-11-01 2018-04-04 ユニバーシティ オブ オスロUniversity of Oslo Modified albumin and use thereof
WO2017029407A1 (en) 2015-08-20 2017-02-23 Albumedix A/S Albumin variants and conjugates

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011051489A2 (en) * 2009-10-30 2011-05-05 Novozymes Biopharma Dk A/S Albumin variants
WO2011124718A1 (en) * 2010-04-09 2011-10-13 Novozymes A/S Albumin derivatives and variants
WO2012059486A1 (en) * 2010-11-01 2012-05-10 Novozymes Biopharma Dk A/S Albumin variants
WO2012150319A1 (en) * 2011-05-05 2012-11-08 Novozymes Biopharma Dk A/S Albumin variants

Non-Patent Citations (9)

* Cited by examiner, † Cited by third party
Title
CHAITY CHAUDHURY ET AL: "Albumin Binding to FcRn: Distinct from the FcRn-IgG Interaction", BIOCHEMISTRY, AMERICAN CHEMICAL SOCIETY, US, vol. 45, no. 15, 18 April 2006 (2006-04-18), pages 4983 - 4990, XP002629973, ISSN: 0006-2960, [retrieved on 20060322], DOI: 10.1021/BI052628Y *
IWAO ET AL: "Changes of net charge and alpha-helical content affect the pharmacokinetic properties of human serum albumin", BIOCHIMICA ET BIOPHYSICA ACTA (BBA) - PROTEINS & PROTEOMICS, ELSEVIER, NETHERLANDS, vol. 1774, no. 12, 19 September 2007 (2007-09-19) - 1 December 2007 (2007-12-01), pages 1582 - 1590, XP022385158, ISSN: 1570-9639, DOI: 10.1016/J.BBAPAP.2007.09.001 *
J. TERJE ANDERSEN ET AL: "Cross-species Binding Analyses of Mouse and Human Neonatal Fc Receptor Show Dramatic Differences in Immunoglobulin G and Albumin Binding", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 285, no. 7, 12 February 2010 (2010-02-12), pages 4826 - 4836, XP055022597, ISSN: 0021-9258, DOI: 10.1074/jbc.M109.081828 *
JAN TERJE ANDERSEN ET AL, NATURE COMMUNICATIONS, 3 January 2012 (2012-01-03), XP055097637, Retrieved from the Internet <URL:http://www.nature.com/ncomms/journal/v3/n1/extref/ncomms1607-s1.pdf> [retrieved on 20140121] *
JAN TERJE ANDERSEN ET AL: "Structure-based mutagenesis reveals the albumin-binding site of the neonatal Fc receptor", NATURE COMMUNICATIONS, vol. 3, 3 January 2012 (2012-01-03), pages 610, XP055097625, DOI: 10.1038/ncomms1607 *
MASAKI OTAGIRI ET AL: "Pharmaceutically Important Pre- and Posttranslational Modifications on Human Serum Albumin", BIOLOGICAL & PHARMACEUTICAL BULLETIN, vol. 32, no. 4, 1 January 2009 (2009-01-01), pages 527 - 534, XP055054558, ISSN: 0918-6158, DOI: 10.1248/bpb.32.527 *
MICHAEL M SCHMIDT ET AL: "Supplemental Information to: Crystal Structure of an HSA/FcRn Complex Reveals Recycling by Competitive Mimicry of HSA Ligands at a pH-Dependent Hydrophobic Interface", STRUCTURE, VOLUME 21, 1966-1978, 10 October 2013 (2013-10-10), XP055097774, Retrieved from the Internet <URL:http://dx.doi.org/10.1016/j.str.2013.08.022> [retrieved on 20140122] *
MICHAEL M. SCHMIDT ET AL: "Crystal Structure of an HSA/FcRn Complex Reveals Recycling by Competitive Mimicry of HSA Ligands at a pH-Dependent Hydrophobic Interface", STRUCTURE, vol. 21, no. 11, 10 October 2013 (2013-10-10) - 1 November 2013 (2013-11-01), pages 1966 - 1978, XP055097728, ISSN: 0969-2126, DOI: 10.1016/j.str.2013.08.022 *
See also references of EP2917233A1 *

Cited By (129)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11555061B2 (en) 2009-02-11 2023-01-17 Albumedix, Ltd Albumin variants and conjugates
US10696732B2 (en) 2009-10-30 2020-06-30 Albumedix, Ltd Albumin variants
US10233228B2 (en) 2010-04-09 2019-03-19 Albumedix Ltd Albumin derivatives and variants
US9181319B2 (en) 2010-08-06 2015-11-10 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9937233B2 (en) 2010-08-06 2018-04-10 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US9447164B2 (en) 2010-08-06 2016-09-20 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9657295B2 (en) 2010-10-01 2017-05-23 Modernatx, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9334328B2 (en) 2010-10-01 2016-05-10 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US10064959B2 (en) 2010-10-01 2018-09-04 Modernatx, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9701965B2 (en) 2010-10-01 2017-07-11 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US9950068B2 (en) 2011-03-31 2018-04-24 Modernatx, Inc. Delivery and formulation of engineered nucleic acids
US9533047B2 (en) 2011-03-31 2017-01-03 Modernatx, Inc. Delivery and formulation of engineered nucleic acids
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US10751386B2 (en) 2011-09-12 2020-08-25 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US10022425B2 (en) 2011-09-12 2018-07-17 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US9428535B2 (en) 2011-10-03 2016-08-30 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US11555062B2 (en) 2011-10-11 2023-01-17 Viela Bio, Inc. Methods of administering a composition comprising a Tn3 scaffold and a CD40L-specific monomer
US10711050B2 (en) 2011-11-18 2020-07-14 Albumedix Ltd Variant serum albumin with improved half-life and other properties
US9186372B2 (en) 2011-12-16 2015-11-17 Moderna Therapeutics, Inc. Split dose administration
US9295689B2 (en) 2011-12-16 2016-03-29 Moderna Therapeutics, Inc. Formulation and delivery of PLGA microspheres
US9271996B2 (en) 2011-12-16 2016-03-01 Moderna Therapeutics, Inc. Formulation and delivery of PLGA microspheres
US10329340B2 (en) 2012-03-16 2019-06-25 Albumedix Ltd Albumin variants
US9944691B2 (en) 2012-03-16 2018-04-17 Albumedix A/S Albumin variants
US9192651B2 (en) 2012-04-02 2015-11-24 Moderna Therapeutics, Inc. Modified polynucleotides for the production of secreted proteins
US9149506B2 (en) 2012-04-02 2015-10-06 Moderna Therapeutics, Inc. Modified polynucleotides encoding septin-4
US9301993B2 (en) 2012-04-02 2016-04-05 Moderna Therapeutics, Inc. Modified polynucleotides encoding apoptosis inducing factor 1
US9303079B2 (en) 2012-04-02 2016-04-05 Moderna Therapeutics, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9254311B2 (en) 2012-04-02 2016-02-09 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins
US9255129B2 (en) 2012-04-02 2016-02-09 Moderna Therapeutics, Inc. Modified polynucleotides encoding SIAH E3 ubiquitin protein ligase 1
US9233141B2 (en) 2012-04-02 2016-01-12 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins associated with blood and lymphatic disorders
US9221891B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. In vivo production of proteins
US9220755B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins associated with blood and lymphatic disorders
US9220792B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. Modified polynucleotides encoding aquaporin-5
US9216205B2 (en) 2012-04-02 2015-12-22 Moderna Therapeutics, Inc. Modified polynucleotides encoding granulysin
US10501512B2 (en) 2012-04-02 2019-12-10 Modernatx, Inc. Modified polynucleotides
US9828416B2 (en) 2012-04-02 2017-11-28 Modernatx, Inc. Modified polynucleotides for the production of secreted proteins
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
US9114113B2 (en) 2012-04-02 2015-08-25 Moderna Therapeutics, Inc. Modified polynucleotides encoding citeD4
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9587003B2 (en) 2012-04-02 2017-03-07 Modernatx, Inc. Modified polynucleotides for the production of oncology-related proteins and peptides
US9107886B2 (en) 2012-04-02 2015-08-18 Moderna Therapeutics, Inc. Modified polynucleotides encoding basic helix-loop-helix family member E41
US9095552B2 (en) 2012-04-02 2015-08-04 Moderna Therapeutics, Inc. Modified polynucleotides encoding copper metabolism (MURR1) domain containing 1
US9675668B2 (en) 2012-04-02 2017-06-13 Moderna Therapeutics, Inc. Modified polynucleotides encoding hepatitis A virus cellular receptor 2
US9089604B2 (en) 2012-04-02 2015-07-28 Moderna Therapeutics, Inc. Modified polynucleotides for treating galactosylceramidase protein deficiency
US9061059B2 (en) 2012-04-02 2015-06-23 Moderna Therapeutics, Inc. Modified polynucleotides for treating protein deficiency
US9050297B2 (en) 2012-04-02 2015-06-09 Moderna Therapeutics, Inc. Modified polynucleotides encoding aryl hydrocarbon receptor nuclear translocator
US8999380B2 (en) 2012-04-02 2015-04-07 Moderna Therapeutics, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
US9878056B2 (en) 2012-04-02 2018-01-30 Modernatx, Inc. Modified polynucleotides for the production of cosmetic proteins and peptides
US9782462B2 (en) 2012-04-02 2017-10-10 Modernatx, Inc. Modified polynucleotides for the production of proteins associated with human disease
US9827332B2 (en) 2012-04-02 2017-11-28 Modernatx, Inc. Modified polynucleotides for the production of proteins
US9814760B2 (en) 2012-04-02 2017-11-14 Modernatx, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
US10501524B2 (en) 2012-11-08 2019-12-10 Albumedix Ltd Albumin variants
GB2512156A (en) * 2012-11-08 2014-09-24 Novozymes Biopharma Dk As Albumin variants
US10934341B2 (en) 2012-11-08 2021-03-02 Albumedix, Ltd. Albumin variants
US9597380B2 (en) 2012-11-26 2017-03-21 Modernatx, Inc. Terminally modified RNA
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
US10815291B2 (en) 2013-09-30 2020-10-27 Modernatx, Inc. Polynucleotides encoding immune modulating polypeptides
US10323076B2 (en) 2013-10-03 2019-06-18 Modernatx, Inc. Polynucleotides encoding low density lipoprotein receptor
WO2016016859A1 (en) 2014-07-31 2016-02-04 Amgen Research (Munich) Gmbh Optimized cross-species specific bispecific single chain antibody constructs
WO2016016412A1 (en) 2014-07-31 2016-02-04 Amgen Research (Munich) Gmbh Antibody constructs for cdh19 and cd3
US11661462B2 (en) 2014-07-31 2023-05-30 Amgen Research (Munich) Gmbh Optimized cross-species specific bispecific single chain antibody contructs
EP3842451A1 (en) * 2015-03-12 2021-06-30 MedImmune, LLC Method of purifying albumin-fusion proteins
US11548933B2 (en) 2015-03-12 2023-01-10 Medlmmune, Llc Method of purifying albumin-fusion proteins
US11926666B2 (en) 2015-04-17 2024-03-12 Amgen Research (Munich) Gmbh Bispecific antibody constructs for CDH3 and CD3
EP4276116A2 (en) 2015-04-17 2023-11-15 Amgen Research (Munich) GmbH Bispecific antibody constructs for cdh3 and cd3
US11028171B2 (en) 2015-04-17 2021-06-08 Amgen Research (Munich) Gmbh Bispecific antibody constructs for CDH3 and CD3
WO2016166360A1 (en) 2015-04-17 2016-10-20 Bayer Pharma Aktiengesellschaft Bispecific antibody constructs for cdh3 and cd3
US10683351B2 (en) 2015-07-31 2020-06-16 Amgen Research (Munich) Gmbh Antibody constructs for DLL3 and CD3
EP3912999A1 (en) 2015-07-31 2021-11-24 Amgen Research (Munich) GmbH Bispecific antibody constructs binding egfrviii and cd3
US10294300B2 (en) 2015-07-31 2019-05-21 Amgen Research (Munich) Gmbh Antibody constructs for DLL3 and CD3
US11884720B2 (en) 2015-07-31 2024-01-30 Amgen Research (Munich) Gmbh Antibody constructs for MSLN and CD3
EP4327885A2 (en) 2015-07-31 2024-02-28 Amgen Research (Munich) GmbH Antibody constructs for msln and cd3
US11155629B2 (en) 2015-07-31 2021-10-26 Amgen Research (Munich) Gmbh Method for treating glioblastoma or glioma with antibody constructs for EGFRVIII and CD3
WO2017021354A1 (en) 2015-07-31 2017-02-09 Amgen Research (Munich) Gmbh Antibody constructs for cd70 and cd3
US10519241B2 (en) 2015-07-31 2019-12-31 Amgen Research (Munich) Gmbh Antibody constructs for EGFRVIII and CD3
EP4219562A2 (en) 2015-07-31 2023-08-02 Amgen Research (Munich) GmbH Antibody constructs for flt3 and cd3
WO2017021362A1 (en) 2015-07-31 2017-02-09 Amgen Research (Munich) Gmbh Antibody constructs for flt3 and cd3
US11591396B2 (en) 2015-07-31 2023-02-28 Amgen Research (Munich) Gmbh Antibody constructs for DLL3 and CD3
EP3865514A1 (en) 2015-07-31 2021-08-18 Amgen Research (Munich) GmbH Bispecific antibody constructs binding dll3 and cd3
WO2017021356A1 (en) 2015-07-31 2017-02-09 Amgen Research (Munich) Gmbh Bispecific antibody constructs binding mesothelin and cd3
WO2017021349A1 (en) 2015-07-31 2017-02-09 Amgen Research (Munich) Gmbh Bispecific antibody constructs binding dll3 and cd3
WO2017021370A1 (en) 2015-07-31 2017-02-09 Amgen Research (Munich) Gmbh Bispecific antibody constructs binding egfrviii and cd3
US11447567B2 (en) 2015-07-31 2022-09-20 Amgen Research (Munich) Gmbh Antibody constructs for FLT3 and CD3
US10851170B2 (en) 2015-07-31 2020-12-01 Amgen Research (Munich) Gmbh Antibody constructs for CD70 and CD3
JP7007261B2 (en) 2015-08-20 2022-01-24 アルブミディクス リミティド Albumin variants and conjugates
JP2018527923A (en) * 2015-08-20 2018-09-27 ノボザイムス バイオファーマ デーコー アクティーゼルスカブ Albumin variants and conjugates
US10633428B2 (en) 2015-08-20 2020-04-28 Albumedix Ltd Albumin variants and conjugates
US10023618B2 (en) 2015-12-22 2018-07-17 Albumedix A/S Protein expression strains
WO2017112847A1 (en) 2015-12-22 2017-06-29 Albumedix A/S Improved protein expression strains
WO2017117631A1 (en) 2016-01-07 2017-07-13 Csl Limited Mutated truncated von willebrand factor
US10806774B2 (en) 2016-01-07 2020-10-20 CSL Behring Lengnau AG Mutated truncated von Willebrand Factor
US11434302B2 (en) 2016-02-03 2022-09-06 Amgen Research (Munich) Gmbh Bispecific T cell engaging antibody constructs
WO2017151957A1 (en) 2016-03-02 2017-09-08 Novozymes A/S Cellobiohydrolase variants and polynucleotides encoding same
WO2017165760A1 (en) 2016-03-24 2017-09-28 Novozymes A/S Cellobiohydrolase variants and polynucleotides encoding same
US11053318B2 (en) 2016-04-19 2021-07-06 Amgen Research (Munich) Gmbh Administration of a bispecific construct binding to CD33 and CD3 for use in a method for the treatment of myeloid leukemia
WO2017182427A1 (en) 2016-04-19 2017-10-26 Amgen Research (Munich) Gmbh Administration of a bispecific construct binding to cd33 and cd3 for use in a method for the treatment of myeloid leukemia
WO2018082758A1 (en) 2016-11-04 2018-05-11 Aarhus Universitet Identification and treatment of tumors characterized by an overexpression of the neonatal fc receptor
WO2018094181A1 (en) 2016-11-21 2018-05-24 Novozymes A/S Yeast cell extract assisted construction of dna molecules
WO2018234349A1 (en) 2017-06-20 2018-12-27 Albumedix Ltd Improved protein expression strains
US11130979B2 (en) 2017-06-20 2021-09-28 Albumedix Ltd Protein expression strains
US11396670B2 (en) 2017-06-20 2022-07-26 Albumedix Limited Protein expression strains
EP4026908A1 (en) 2017-06-20 2022-07-13 Albumedix Ltd Improved protein expression strains
US11141466B2 (en) 2017-06-22 2021-10-12 CSL Behring Lengnau AG Modulation of FVIII immunogenicity by truncated VWF
WO2018234518A1 (en) 2017-06-22 2018-12-27 CSL Behring Lengnau AG Modulation of fviii immunogenicity by truncated vwf
US11541103B2 (en) 2017-08-03 2023-01-03 Amgen Inc. Interleukin-21 mutein/ anti-PD-1 antibody conjugates
US10640504B2 (en) 2017-09-08 2020-05-05 Amgen Inc. Inhibitors of KRAS G12C and methods of using the same
WO2019104385A1 (en) 2017-11-29 2019-06-06 Csl Limited Method of treating or preventing ischemia-reperfusion injury
WO2019140196A1 (en) 2018-01-12 2019-07-18 Amgen Inc. Anti-pd-1 antibodies and methods of treatment
US11518808B2 (en) 2018-01-12 2022-12-06 Amgen Inc. Anti-PD-1 antibodies and methods of treatment
WO2019218009A1 (en) 2018-05-16 2019-11-21 Csl Limited Soluble complement receptor type 1 variants and uses thereof
WO2020025532A1 (en) 2018-07-30 2020-02-06 Amgen Research (Munich) Gmbh Prolonged administration of a bispecific antibody construct binding to cd33 and cd3
WO2020025792A1 (en) 2018-08-03 2020-02-06 Amgen Research (Munich) Gmbh Antibody constructs for cldn18.2 and cd3
US11692031B2 (en) 2018-08-03 2023-07-04 Amgen Research (Munich) Gmbh Antibody constructs for CLDN18.2 and CD3
WO2020049151A1 (en) 2018-09-06 2020-03-12 Bavarian Nordic A/S Storage improved poxvirus compositions
WO2021044362A1 (en) 2019-09-06 2021-03-11 Novartis Ag Therapeutic fusion proteins
WO2021094000A1 (en) 2019-11-11 2021-05-20 Amgen Research (Munich) Gmbh Dosing regimen for anti-bcma agents
EP3819007A1 (en) 2019-11-11 2021-05-12 Amgen Research (Munich) GmbH Dosing regimen for anti-bcma agents
WO2021111143A1 (en) 2019-12-04 2021-06-10 Albumedix Limited Methods and compositions produced thereby
WO2021150824A1 (en) 2020-01-22 2021-07-29 Amgen Research (Munich) Gmbh Combinations of antibody constructs and inhibitors of cytokine release syndrome and uses thereof
WO2021180943A1 (en) 2020-03-12 2021-09-16 Bavarian Nordic A/S Compositions improving poxvirus stability
WO2021183861A1 (en) 2020-03-12 2021-09-16 Amgen Inc. Method for treatment and prophylaxis of crs in patients comprising a combination of bispecifc antibodies binding to cds x cancer cell and tnfalpha or il-6 inhibitor
WO2021236638A1 (en) 2020-05-19 2021-11-25 Amgen Inc. Mageb2 binding constructs
WO2021243320A2 (en) 2020-05-29 2021-12-02 Amgen Inc. Adverse effects-mitigating administration of a bispecific antibody construct binding to cd33 and cd3
WO2022060901A1 (en) 2020-09-16 2022-03-24 Amgen Inc. Methods for administering therapeutic doses of bispecific t-cell engaging molecules for the treatment of cancer
WO2022096704A1 (en) 2020-11-06 2022-05-12 Amgen Inc. Antigen binding domain with reduced clipping rate
WO2022096700A1 (en) 2020-11-06 2022-05-12 Amgen Research (Munich) Gmbh Polypeptide constructs selectively binding to cldn6 and cd3
WO2022096698A1 (en) 2020-11-06 2022-05-12 Amgen Inc. Polypeptide constructs binding to cd3
WO2022103781A1 (en) 2020-11-10 2022-05-19 Amgen Inc. Methods for administering a bcma x cd3 binding molecule
WO2024077044A1 (en) 2022-10-05 2024-04-11 Amgen Inc. Combination therapies comprising t-cell redirecting therapies and agonistic anti-il-2r antibodies or fragments thereof

Also Published As

Publication number Publication date
IL238185A0 (en) 2015-05-31
RU2670063C2 (en) 2018-10-17
JP6487328B2 (en) 2019-03-20
IL238185B (en) 2019-09-26
CN105452290A (en) 2016-03-30
AU2013343503A1 (en) 2015-04-30
AU2018201136A1 (en) 2018-03-08
US20200102367A1 (en) 2020-04-02
BR112015010318A2 (en) 2017-08-22
KR20150082422A (en) 2015-07-15
US20140128326A1 (en) 2014-05-08
US10501524B2 (en) 2019-12-10
JP2019048821A (en) 2019-03-28
JP2015535300A (en) 2015-12-10
AU2013343503B2 (en) 2017-12-14
US20150210752A1 (en) 2015-07-30
GB2512156A (en) 2014-09-24
EP2917233A1 (en) 2015-09-16
CA2890766A1 (en) 2014-05-15
US10934341B2 (en) 2021-03-02
GB201319754D0 (en) 2013-12-25
MX2015005363A (en) 2015-11-06
RU2015121693A (en) 2016-12-27

Similar Documents

Publication Publication Date Title
US10934341B2 (en) Albumin variants
US10329340B2 (en) Albumin variants
US20200102368A1 (en) Albumin variants
US20130225496A1 (en) Albumin Variants
US20160222087A1 (en) Albumin variants

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 201380069331.4

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 13789000

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 238185

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: MX/A/2015/005363

Country of ref document: MX

ENP Entry into the national phase

Ref document number: 2013343503

Country of ref document: AU

Date of ref document: 20131108

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2015541165

Country of ref document: JP

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2890766

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2013789000

Country of ref document: EP

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112015010318

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 20157014474

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2015121693

Country of ref document: RU

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 112015010318

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20150506