US20140128326A1 - Albumin variants - Google Patents

Albumin variants Download PDF

Info

Publication number
US20140128326A1
US20140128326A1 US14/075,104 US201314075104A US2014128326A1 US 20140128326 A1 US20140128326 A1 US 20140128326A1 US 201314075104 A US201314075104 A US 201314075104A US 2014128326 A1 US2014128326 A1 US 2014128326A1
Authority
US
United States
Prior art keywords
seq
albumin
polypeptide
fragment
hsa
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US14/075,104
Other languages
English (en)
Inventor
Jason Cameron
Karen Ann Delahay
Jens Erik Nielsen
Andrew Plumridge
Jan Terje Andersen
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Novozymes AS
Novozymes Biopharma DK AS
Albumedix Ltd
Universitetet i Oslo
Original Assignee
Novozymes AS
Novozymes Biopharma DK AS
Novozymes Biopharma UK Ltd
Universitetet i 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
Application filed by Novozymes AS, Novozymes Biopharma DK AS, Novozymes Biopharma UK Ltd, Universitetet i Oslo filed Critical Novozymes AS
Priority to US14/075,104 priority Critical patent/US20140128326A1/en
Assigned to NOVOZYMES BIOPHARMA DK A/S reassignment NOVOZYMES BIOPHARMA DK A/S ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ANDERSEN, JAN TERJE, NIELSEN, JENS ERIK, CAMERON, JASON, DELAHAY, Karen Ann, PLUMRIDGE, ANDREW
Publication of US20140128326A1 publication Critical patent/US20140128326A1/en
Priority to US14/685,112 priority patent/US10501524B2/en
Priority to US16/598,939 priority patent/US10934341B2/en
Abandoned legal-status Critical Current

Links

Images

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
  • 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 DIII (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 and H536 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 in drug delivery.
  • Albumin 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 et al (1990) discloses a natural variant K536E.
  • Minchiotti et al (1987) Biochim. Biophys. Acta 916, 411-418 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.
  • WO2011/051489 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.
  • WO2011/103076 discloses albumin variants, each containing a substitution in Domain III of HSA.
  • WO2012/112188 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; WO2010/118169) 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.).
  • the studies of Kenanova et al are limited to domain III of HSA and therefore do not consider HSA in its native intact configuration.
  • 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.
  • 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 DIIIa and subdomain DIIIb.
  • Positions 550, 573, 574 and 580 are located in subdomain DIIIb.
  • Subdomain IIIb is located proximal to the connector loop between subdomain DIIIa and subdomain DIIIb.
  • 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 albumin or fragment thereof, according 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.
  • FIG. 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 III 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 FIG. 1 , Domains I, II and III are referred to as 1, 2 and 3 (respectively).
  • FIG. 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.
  • FIG. 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; DI (CBI)), while the area distally localized from the interface is shown as DII (orange) and DIII is split into sub-domains DIIIa (in colour, this is cyan) and DIIIb (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;
  • 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 number 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 X M, Carter D C (July 1992). “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 three 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.
  • 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 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 ID NO: 2 (mature), SEQ ID NO: 4 (immature)), primate serum albumin, (such as chimpanzee serum albumin (e.g. predicted sequence XP — 517233.2 SEQ ID NO: 5), gorilla serum albumin or macaque serum albumin (e.g. NP — 001182578, SEQ ID NO: 6), rodent serum albumin (such as hamster serum albumin (e.g.
  • human serum albumin e.g. AAA98797 or P02768-1, SEQ ID NO: 2 (mature), SEQ ID NO: 4 (immature)
  • primate serum albumin such as chimpanzee serum albumin (e.g. predicted sequence XP — 517233.2 SEQ
  • P08835-1 Version 2 SEQ ID NO: 19
  • 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 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 ID 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 ID 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 ID 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 ID 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; O73860.pro: turkey ovalbumin).
  • HSA as disclosed in SEQ ID 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 ID 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 ID 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 III (e.g. SEQ ID NO: 27), a molecule consisting of or comprising domain II and domain III (e.g. SEQ ID 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 ID 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 ID 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 ID 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, N.Y.).
  • the polynucleotide of SEQ ID NO: 1 or a subsequence thereof, as well as the amino acid sequence of SEQ ID 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.
  • 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 100 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 ID 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 5 ⁇ 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 2 ⁇ 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 n , using the calculation according to Bolton and McCarthy (1962 , Proc. Natl. Acad. Sci. USA 48: 1390) in 0.9 M NaCl, 0.09 M Tris-HCl pH 7.6, 6 mM EDTA, 0.5% NP-40, 1 ⁇ Denhardt's solution, 1 mM 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 6 ⁇ SCC plus 0.1% SDS for 15 minutes and twice each for 15 minutes using 6 ⁇ 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 ID NO: 30 (truncated heavy chain of the major histocompatibility complex class I-like Fc receptor (FCGRT)) and SEQ ID NO: 31 (beta-2-microglobulin). Together, SEQ ID 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 following 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 1758 of SEQ ID NO: 1e.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:
  • sequence identity between two deoxyribonucleotide sequences is determined using the Needleman-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” 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 100 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 DI+DII, DI+DIII, DII+DIII, DIII+DIII, DI+DIII+DIII, DIII+DIII+DIII, or fragments of such domains or combinations of domains.
  • HSA domains I, II and III may be defined with reference to HSA (SEQ ID NO: 2).
  • HSA domain I may consist of or comprise amino acids 1 to 194 ( ⁇ 1 to 15 amino acids) of SEQ ID NO: 2
  • HSA domain II 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 III 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, 11, 12, 13, 14, or 15 amino acids to the C-terminus and/or to the N-terminus of the stated amino acid position.
  • 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, II and III 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 BLOSUM62) substitution matrix.
  • Other suitable software includes MUSCLE ((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. 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 means a configuration in which a control sequence 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 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.
  • 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 ID 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 ID 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.
  • FIG. 1 was generated by MUSCLE using the default parameters including output in ClustalW 1.81 format.
  • the raw output data was shaded using BoxShade 3.21 (http://www.ch.embnet.orq/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.
  • 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 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 ID 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 ID 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 10, 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: 511-518; 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
  • 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).
  • the distance alignment matrix Holm and Sander, 1998 , Proteins 33: 88-96
  • combinatorial extension Shindyalov and Bourne, 1998 , Protein Engineering 11: 739-747
  • albumin variants of the present invention the nomenclature described below is adapted for ease of reference.
  • the accepted IUPAC 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.
  • “Arg170Tyr,Glu” represents a substitution of arginine at position 170 with tyrosine or glutamic acid.
  • “Tyr167Gly,Ala+Arg170Gly,Ala” designates the following variants: “Tyr167Gly+Arg170Gly”, “Tyr167Gly+Arg170Ala”, “Tyr167Ala+Arg170Gly”, and “Tyr167Ala+Arg170Ala”.
  • 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 SEQ ID NO: 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 (j).
  • 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 ID 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 ID NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492D+550K (e.g. SEQ ID 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 ID 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: 110), 492H+573P (e.g. SEQ ID NO: 111) or 492R+573P (e.g. SEQ ID NO: 113) 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+573A, 492G+573A, 492G+N503K+573A, 492G+N503H-F573A, 492G+573P, 492G+N503K+573P or 492G+N503H+573P.
  • a variant albumin may comprise alterations at positions corresponding to positions 492+574 of SEQ ID NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492D+574H (e.g. SEQ ID NO: 232), 492G+574H
  • SEQ ID NO: 241 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 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 550+573 of SEQ ID NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 550K+573P (e.g. SEQ ID NO: 117).
  • a variant albumin may comprise alterations at positions corresponding to positions 550+574 of SEQ ID 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. SEQ ID NO: 245).
  • a variant albumin may comprise alterations at positions corresponding to positions 550+580 of SEQ ID 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 ID 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. SEQ ID NO: 124), 574N+573P (e.g. SEQ ID NO: 125) or 5745+573P (e.g. SEQ ID NO: 126). It is preferred that the variant does not consist of SEQ ID 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 ID NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 580K+573P (e.g. SEQ ID NO: 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 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+550+573 of SEQ ID 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 ID 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 ID 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 ID NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492D+573P+574H (e.g. SEQ ID NO: 233).
  • a variant albumin may comprise alterations at positions corresponding to positions 492+573+580 of SEQ ID 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 ID NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492D+574H+580K (SEQ ID 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. 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) 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 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) 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 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) or 492G+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 ID 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 ID NO: 266) or 492G+550K+573P+574H+580K (e.g. SEQ ID 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 574 in SEQ ID 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 ID 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 ID 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 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 574;
  • variant albumin 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.
  • 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;
  • 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;
  • 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 ID 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 ID NO: 2, 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
  • positions corresponding to positions 492, 573, 574 and 580 in SEQ ID 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: 114;
  • 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: 115.
  • 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 WO2011/051489.
  • the variant albumin does not consist of SEQ ID NO: 2 with amino acids 573 to 585 replaced with (a) KKLVAASQAALGL, (b) PKFVAASQAALA, (c) PNLVTRCKDALA, (d) PKLVESSKATLG or (e) PKLVASTQAALA.
  • 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 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.
  • 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.
  • 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.
  • 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; introduction of reactive groups of the surface 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.
  • 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.
  • 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.
  • WO 2001/79271A (particularly page 9 and/or Table 1)
  • WO 2003/59934 (particularly Table 1)
  • WO03/060071 (particularly Table 1)
  • 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 nucleic 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), 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 (CUP1), and Saccharomyces cerevisiae 3-phosphoglycerate kinase.
  • ENO-1 Saccharo
  • useful promoters for use in rice and mammalian cells, such as CHO or HEK.
  • 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.
  • Preferred terminators for yeast host cells are obtained from the genes for Saccharomyces cerevisiae enolase, Saccharomyces cerevisiae cytochrome C(CYC1), Saccharomyces cerevisiae alcohol dehydrogenase (ADH1) and Saccharomyces cerevisiae glyceraldehyde-3-phosphate dehydrogenase.
  • Other useful terminators for yeast host cells are described by Romanos et al., 1992, supra.
  • useful terminators for use in rice and mammalian cells, such as CHO or HEK 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 (ENO-1), Saccharomyces cerevisiae 3-phosphoglycerate kinase, Saccharomyces cerevisiae alpha-factor, and Saccharomyces cerevisiae alcohol dehydrogenase/glyceraldehyde-3-phosphate dehydrogenase (ADH2/GAP).
  • ENO-1 Saccharomyces cerevisiae enolase
  • Saccharomyces cerevisiae 3-phosphoglycerate kinase Saccharomyces cerevisiae alpha-factor
  • 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.
  • yeast host cells 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.
  • 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.
  • Useful signal peptides for yeast host cells are obtained 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.
  • 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:
  • 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 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: 2004/0171154; 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: 1050-1054) 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. Pat. No. 5,223,409; WO 92/06204) and region-directed mutagenesis (Derbyshire et al., 1986 , Gene 46: 145; Ner et al., 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 within
  • 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 suitable 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.
  • 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.
  • 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 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 compound 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”.
  • 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.
  • 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).
  • a taxane e.g. paclitaxel
  • 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’.
  • 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, 100, 130, 150, 200, 300, 400, 500, 600, 700, 800, 900, or 1000 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 ⁇ m (micron) filter.
  • the average diameter of a micro-particle is from 1000 nm (1 ⁇ m (micron)) to 100 ⁇ m (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 administration 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 albumin 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.
  • albumin component e.g. albumin, variant, fragment, fusion, conjugate or associate
  • HSA wild-type albumin
  • a variant, fragment, fusion, conjugate or associate is wild-type albumin (e.g. HSA) or a variant, fragment, fusion, conjugate or associate.
  • 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:
  • ‘molecule’ examples 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 0.9 ⁇ KD for HSA to FcRn, more preferred less than 0.5 ⁇ KD for HSA to FcRn, more preferred less than 0.1 ⁇ KD for HSA to FcRn, even more preferred less than 0.05 ⁇ KD for HSA to FcRn, even more preferred less than 0.02 ⁇ KD for HSA to FcRn and most preferred less than 0.01 ⁇ KD for HSA to FcRn (where ⁇ 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 composition 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 2 ⁇ KD for HSA to FcRn, more preferred more than 5 ⁇ KD for HSA to FcRn, more preferred more than 10 ⁇ KD for HSA to FcRn, even more preferred more than 25 ⁇ KD for HSA to FcRn, most preferred more than 50 ⁇ KD, more than 60 ⁇ , more than 70 ⁇ KD, more than 80 ⁇ , more than 90 ⁇ or more than 100 ⁇ 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
  • 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).
  • Coupling method The coupling may be performed by injecting 20 ⁇ g/ml of the protein in 10 mM sodium acetate pH 5.0 (GE Healthcare). Phosphate buffer (67 mM phosphate buffer, 0.15 M NaCl, 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 NaCl, 3 mM EDTA, 0.005% surfactant P20) at pH 7.4 (Biacore AB).
  • test molecule e.g. HSA or variant
  • Flow rate of injection constant, e.g. 30 ⁇ l/ml
  • Example 2 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 albumin, 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.
  • 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 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.
  • 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 ID NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492D+550K (e.g. SEQ ID 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 ID 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: 110), 492H+573P (e.g. SEQ ID NO: 111) or 492R+573P (e.g. SEQ ID NO: 113) 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 ID NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492D+574H (e.g. SEQ ID NO: 232), 492G+574H (e.g. SEQ ID NO: 241) or 492D+574H (e.g. SEQ ID NO: 232).
  • 492D+574H e.g. SEQ ID 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 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 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: 117).
  • a variant albumin may comprise alterations at positions corresponding to positions 550+574 of SEQ ID 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. SEQ ID NO: 245).
  • a variant albumin may comprise alterations at positions corresponding to positions 550+580 of SEQ ID 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 ID 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. SEQ ID NO: 124), 574N+573P (e.g. SEQ ID NO: 125) or 5745+573P (e.g. SEQ ID NO: 126). It is preferred that the variant does not consist of SEQ ID 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 ID NO: 2 (or equivalent position of other albumins or variants or fragments thereof).
  • Such alterations may comprise 580K+573P (e.g. SEQ ID NO: 128) or 580R+573P (e.g. SEQ ID NO: 129).
  • 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 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+550+573 of SEQ ID 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 ID 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 ID 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 ID NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492D+573P+574H (e.g. SEQ ID NO: 233).
  • a variant albumin may comprise alterations at positions corresponding to positions 492+573+580 of SEQ ID 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 ID NO: 2 (or equivalent position of other albumins or variants or fragments thereof). Such alterations may comprise 492D+574H+580K (SEQ ID 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 SEQ ID 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).
  • 550K+574H+573P e.g. SEQ ID NO: 131
  • 550L+573P+574H e.g. SEQ ID NO: 246
  • 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) 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 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) 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 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) or 492G+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 ID 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 ID NO: 266) or 492G+550K+573P+574H+580K (e.g. SEQ ID 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 ID 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 ID 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 ID 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 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 574.
  • a variant albumin with alterations e.g. comprising alterations
  • alterations e.g. comprising alterations
  • 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 (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;
  • 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 ID 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 ID NO: 2, 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;
  • variant albumin comprising alterations at positions corresponding to positions 492, 573, 574 and 580 in SEQ ID 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: 114;
  • 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: 115.
  • 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 II are maintained (i.e. 242, 247, 288, 338, 367).
  • One or more (several), preferably all, of the His residues in domain III 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, 11, 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: 4-1BB 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 (MIG), A partial CXCR4B protein, A platelet basic protein (PBP), a1-antitrypsin, ACRP-30 Homologue; Complement Component Clq 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 III, APM-1; ACRP-30; Famoxin, apo-lipoprotein species, Arylsulfatase B, b57
  • 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 (GCP-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, HER2Receptor Mab, hirudin, HIV
  • SMIP Small Modular ImmunoPharmaceuticalTM
  • dAb Fab′ fragments, F(ab′)2, scAb, scFv or scFv fragment
  • plasminogen Influenza Vaccine
  • Inhibin alpha Inhibin beta
  • insulin insulin-like growth factor
  • Integrin Mab 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 sub-species
  • Interferons such as interferon alpha species and sub-species, interferon beta species and sub-species, interferon gamma species and sub-species
  • 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®, All-transretinoic Acid, Alpha Interferon, Altretamine, Amethopterin, Amifostine, Aminoglutethimide, Anagrelide, Anandron®, Anastrozole, Arabinosylcytosine, Ara-C, Aranesp®, Aredia®, Arimidex®, Aromasin®,
  • Paclitaxel or Paclitaxel Protein-bound Pamidronate, Panitumumab, Panretin®, Paraplatin®, Pediapred®, PEG Interferon, Pegaspargase, Pegfilgrastim, PEG-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), Rubex®, Rubidomycin hydrochloride, Sandostatin®, Sandostatin LAR®, Sargramostim, Solu-Cortef®, Sol
  • 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-1b 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 VIII, Helixate FS, Kogenate FS, Factor IX, BeneFIX, Factor Vila, 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-Intron
  • drugs for the treatment of HIV 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
  • MS multiple sclerosis
  • 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.
  • the polypeptide according to embodiment 1 or 2 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. 4.
  • polypeptide according to embodiment 1, 2 or 3 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: 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 580 of SEQ ID NO: 2. 6.
  • 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.
  • polypeptide according to any preceding embodiment comprising alterations at positions corresponding to positions 550 and 580 of SEQ ID NO: 1, further comprising an alteration at a position corresponding to position 492, 573 and 574 of SEQ ID NO: 2.
  • the polypeptide according to any preceding embodiment in which: (a) at a position corresponding to position 492 of SEQ ID 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 ID NO: 2 there is an alteration
  • 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 ID 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
  • 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,
  • polypeptide according to any preceding embodiment in which: (a) 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 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 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
  • 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.
  • 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.
  • the polypeptide of embodiment 14 wherein the substitution at the position corresponding to position 492 is to G, D, F, H, M or R.
  • the polypeptide of embodiment 14 or 15 wherein the substitution at the position corresponding to position 492 is to G or D.
  • 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 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.
  • 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 ID NO: 124); 574N+573P (e.g. SEQ ID NO: 125); 574S+573P (e.g. SEQ ID NO: 126); 550K+580K (e.g. SEQ ID NO: 132); 550K+574H (e.g.
  • SEQ ID NO: 130 550K+573P (e.g. SEQ ID NO: 117); 492D+573P (e.g. SEQ ID NO: 108); 492F+573P (e.g. SEQ ID NO: 109); 492H+573P (e.g. SEQ ID NO: 111); 492R+573P (e.g. SEQ ID NO: 112); 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.
  • 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.
  • 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.
  • 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 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); 36.
  • 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.
  • 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: 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 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.
  • 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. 43.
  • 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 45.
  • substitution at the position corresponding to position 492 is to G. 46.
  • 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.
  • 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.
  • 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.
  • substitution at the position corresponding to position 580 is a substitution to K or R, most preferably R. 51.
  • 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 ID NO: 132); 550K+574H (e.g. SEQ ID NO: 130); 550K+573P (e.g. SEQ ID NO: 117); 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: 111); 492R+573P (e.g. SEQ ID NO: 112); 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 SEQ ID NO: 232
  • 492G+550K e.g. SEQ ID NO: 240
  • 550M+574H e.g. SEQ ID NO: 249
  • 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.
  • 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.
  • 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
  • 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.
  • 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: 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.
  • 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).
  • 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.
  • 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.
  • a nanoparticle or microparticle 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 or an associate according to embodiment 64.
  • 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.
  • 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 or a nanoparticle 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
  • 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.
  • 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.
  • 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, 3r d ed. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y).
  • 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 ⁇ l of reaction product was assessed by agarose gel electrophoresis and the remainder treated with 5 ⁇ l of 10 ⁇ buffer 4 (New England Biolabs—50 mM potassium acetate, 20 mM Tris-acetate, 10 mM magnesium acetate, 1 mM DTT pH 7.9 at 25° C.) and 1 ⁇ l DpnI (NEB) for 1 hour at 37° C.
  • 10 ⁇ buffer 4 New England Biolabs—50 mM potassium acetate, 20 mM Tris-acetate, 10 mM magnesium acetate, 1 mM DTT pH 7.9 at 25° C.
  • the reactions were purified by addition of 50 ⁇ l water, prior to application of a vacuum for 20 minutes. A further 50 ⁇ l of water was added and a vacuum applied until the well was dry. Plasmid DNA was recovered by addition of 30 ⁇ l of water and incubation for 1 minute. 2 ⁇ l of this purified product was transformed into 15 ⁇ l 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, Bsal/SphI 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 ADH1 terminator) into HindIII/SphI-digested pDB4005.
  • pDB4005 is described in WO 2011/051489 (incorporated herein by reference).
  • 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 SalI and Bsu361 restriction sites and inserted into similarly digested parent plasmid, as indicated in Table 5.
  • pDB4673 HSA K573P
  • the fragment encoding K573P+K574H+Q580K was removed from pDB5032 using the SalI and Bsu36I restriction sites and inserted into similarly digested parent plasmid, as indicated in Table 6.
  • Plasmid and amino acid substitution Relevant Variant SEQ Parent Plasmid Variant ID No. Plasmid name HSA E492D + K573P + 114 pDB4770 pDB5091 K574H + Q580K HSA E492G + K573P + 115 pDB4772 pDB5092 K574H + Q580K
  • 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 11 were used to produce pDB5027-29 and pDB5032.
  • the mutated PCR products were digested using SalI and Bsu361 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 WO2011/051489.
  • Chip surface was left to stabilize with a constant flow (5 ⁇ L/min) of running buffer—Di-basic/Mono-basic phosphate buffer pH5.5 ((67 mM phosphate buffer, 0.15 M NaCl, 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 ⁇ 45 ⁇ L Di-basic/Mono-basic phosphate buffer at 30 ⁇ L/min followed by HBS_EP (0.01 M HEPES, 0.15 M NaCl, 3 mM EDTA, 0.005% surfactant P20) at pH 7.4 (GE Healthcare)) regeneration steps (12s) in between each injection.
  • 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-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.
  • HSA-D550K+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 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.
  • 3 ⁇ l of the prepared plasmid, 3 ⁇ l of Acc651/BamHI-digested pDB3936 and 1 ⁇ l 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 al 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 ⁇ l 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 10 mL 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 ⁇ L custom packed Atoll columns with 8 run in parallel in a 96-well format using the same procedure as described in WO 2011/051489 (incorporated herein by reference), with volumes scaled down appropriately.
  • albumin affinity matrix Albumin affinity matrix
  • 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.
  • shFcRn was captured on a GSTrap column and eluted with reduced glutathione.
  • shFcRn was dialyzed into PBS into pH 7.4 and further purified using IgG SepharoseTM 6 Fast Flow (GE Healthcare).
  • the shFcRn was captured on the resin in 50 mM Na-acetate, 150 mM NaCl pH 5.5 and eluted with PBS pH 7.4.
  • the eluted shFcRn was concentrated to 2-5 mg/mL and stored at ⁇ 20° C. until use.
  • Immobilised level of shFcRn-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.
  • the His-tagged shFcRn was captured on a Ni-HiTrap column and eluted with imidazole.
  • the shFcRn was captured on the resin in 50 mM Na-acetate, 150 mM NaCl pH 5.5 and eluted with PBS pH 7.4.
  • the eluted shFcRn was concentrated to 2-5 mg/mL and stored at ⁇ 20° C. until use. using shFcRn-HIS

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Epidemiology (AREA)
  • Toxicology (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Biomedical Technology (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Physics & Mathematics (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Wood Science & Technology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Optics & Photonics (AREA)
  • Radiology & Medical Imaging (AREA)
  • Medicinal Preparation (AREA)
US14/075,104 2012-11-08 2013-11-08 Albumin variants Abandoned US20140128326A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US14/075,104 US20140128326A1 (en) 2012-11-08 2013-11-08 Albumin variants
US14/685,112 US10501524B2 (en) 2012-11-08 2015-04-13 Albumin variants
US16/598,939 US10934341B2 (en) 2012-11-08 2019-10-10 Albumin variants

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
EP12191856 2012-11-08
EP12191856.9 2012-11-08
US201261724669P 2012-11-09 2012-11-09
US14/075,104 US20140128326A1 (en) 2012-11-08 2013-11-08 Albumin variants

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/685,112 Division US10501524B2 (en) 2012-11-08 2015-04-13 Albumin variants

Publications (1)

Publication Number Publication Date
US20140128326A1 true US20140128326A1 (en) 2014-05-08

Family

ID=47137625

Family Applications (3)

Application Number Title Priority Date Filing Date
US14/075,104 Abandoned US20140128326A1 (en) 2012-11-08 2013-11-08 Albumin variants
US14/685,112 Active 2035-12-09 US10501524B2 (en) 2012-11-08 2015-04-13 Albumin variants
US16/598,939 Active US10934341B2 (en) 2012-11-08 2019-10-10 Albumin variants

Family Applications After (2)

Application Number Title Priority Date Filing Date
US14/685,112 Active 2035-12-09 US10501524B2 (en) 2012-11-08 2015-04-13 Albumin variants
US16/598,939 Active US10934341B2 (en) 2012-11-08 2019-10-10 Albumin variants

Country Status (13)

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

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160017343A1 (en) * 2013-03-06 2016-01-21 Glaxosmithkline Llc Host cells and methods of use
US20160075762A1 (en) * 2009-10-30 2016-03-17 Novozymes Biopharma Dk A/S Albumin variants
US9944691B2 (en) 2012-03-16 2018-04-17 Albumedix A/S Albumin variants
US10233228B2 (en) 2010-04-09 2019-03-19 Albumedix Ltd Albumin derivatives and variants
CN109553684A (zh) * 2017-09-25 2019-04-02 中国科学院过程工程研究所 一种纳米载体蛋白及其制备方法和应用
US10501524B2 (en) 2012-11-08 2019-12-10 Albumedix Ltd Albumin variants
US10633428B2 (en) 2015-08-20 2020-04-28 Albumedix Ltd Albumin variants and conjugates
US10711050B2 (en) 2011-11-18 2020-07-14 Albumedix Ltd Variant serum albumin with improved half-life and other properties
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
CN113912730A (zh) * 2021-12-14 2022-01-11 北京科诺信诚科技有限公司 缓释的抗FcRn抗体或抗原结合片段及其应用
US20220031741A1 (en) * 2016-06-03 2022-02-03 New York University Methods and reagents for modulating macrophage phenotype
US11434302B2 (en) 2016-02-03 2022-09-06 Amgen Research (Munich) Gmbh Bispecific T cell engaging antibody constructs
US11447567B2 (en) 2015-07-31 2022-09-20 Amgen Research (Munich) Gmbh Antibody constructs for FLT3 and CD3
US11555061B2 (en) 2009-02-11 2023-01-17 Albumedix, Ltd Albumin variants and conjugates
US11661462B2 (en) * 2014-07-31 2023-05-30 Amgen Research (Munich) Gmbh Optimized cross-species specific bispecific single chain antibody contructs
US11884720B2 (en) 2015-07-31 2024-01-30 Amgen Research (Munich) Gmbh Antibody constructs for MSLN and CD3

Families Citing this family (55)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2600901B1 (fr) 2010-08-06 2019-03-27 ModernaTX, Inc. Compositions pharmaceutiques a base d'acides nucléiques modifiés et leur utilisation medicale
CN104531671A (zh) 2010-10-01 2015-04-22 现代治疗公司 设计核酸及其使用方法
CA2831613A1 (fr) 2011-03-31 2012-10-04 Moderna Therapeutics, Inc. Administration et formulation d'acides nucleiques genetiquement modifies
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
EP3682905B1 (fr) 2011-10-03 2021-12-01 ModernaTX, Inc. Nucléosides, nucléotides et acides nucléiques modifiés et leurs utilisations
SI2771022T1 (sl) 2011-10-11 2020-12-31 Viela Bio, Inc. CD40L-specifična ogrodja pridobljena iz TN3 in postopki njihove uporabe
ES2923757T3 (es) 2011-12-16 2022-09-30 Modernatx Inc Composiciones de ARNm modificado
US9303079B2 (en) 2012-04-02 2016-04-05 Moderna Therapeutics, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
AU2013243953A1 (en) 2012-04-02 2014-10-30 Modernatx, Inc. Modified polynucleotides for the production of nuclear proteins
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
DK2922554T3 (en) 2012-11-26 2022-05-23 Modernatx Inc Terminalt modificeret rna
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
WO2015048744A2 (fr) 2013-09-30 2015-04-02 Moderna Therapeutics, Inc. Polynucléotides codant des polypeptides de modulation immunitaire
WO2015051214A1 (fr) 2013-10-03 2015-04-09 Moderna Therapeutics, Inc. Polynucléotides codant pour un récepteur de lipoprotéines de faible densité
AR101669A1 (es) 2014-07-31 2017-01-04 Amgen Res (Munich) Gmbh Constructos de anticuerpos para cdh19 y cd3
JP6862348B2 (ja) 2015-03-12 2021-04-21 メディミューン,エルエルシー アルブミン融合タンパク質を精製する方法
EP4276116A3 (fr) 2015-04-17 2024-01-17 Amgen Research (Munich) GmbH Constructions d'anticorps bispécifiques pour cdh3 et cd3
TWI793062B (zh) 2015-07-31 2023-02-21 德商安美基研究(慕尼黑)公司 Dll3及cd3抗體構築體
TWI717375B (zh) 2015-07-31 2021-02-01 德商安美基研究(慕尼黑)公司 Cd70及cd3抗體構築體
WO2017112847A1 (fr) 2015-12-22 2017-06-29 Albumedix A/S Souches améliorées pour l'expression de protéines
KR20180094114A (ko) 2016-01-07 2018-08-22 체에스엘 베링 리컴비넌트 퍼실리티 아게 돌연변이된 절단된 폰 빌레브란트 인자
CN109415712A (zh) 2016-03-02 2019-03-01 诺维信公司 纤维二糖水解酶变体和编码它们的多核苷酸
US11965189B2 (en) 2016-03-24 2024-04-23 Novozymes A/S Cellobiohydrolase variants and polynucleotides encoding same
JOP20170091B1 (ar) 2016-04-19 2021-08-17 Amgen Res Munich Gmbh إعطاء تركيبة ثنائية النوعية ترتبط بـ cd33 وcd3 للاستخدام في طريقة لعلاج اللوكيميا النخاعية
WO2018082758A1 (fr) 2016-11-04 2018-05-11 Aarhus Universitet Identification et traitement de tumeurs caractérisées par une surexpression du récepteur fc néonatal
US20210284991A1 (en) 2016-11-21 2021-09-16 Novozymes A/S Yeast Cell Extract Assisted Construction of DNA Molecules
CN110573529A (zh) * 2017-04-05 2019-12-13 韩国生命工学研究院 激活nk细胞的融合蛋白、nk细胞和包含其的药物组合物
CA3064037A1 (fr) 2017-06-20 2018-12-27 Albumedix Ltd Souches ameliorees pour l'expression de proteines
AU2018287215A1 (en) 2017-06-22 2020-02-06 CSL Behring Lengnau AG Modulation of FVIII immunogenicity by truncated VWF
CN111164100B (zh) 2017-08-03 2024-03-12 美国安进公司 白介素-21突变蛋白和治疗方法
JP7150823B2 (ja) 2017-09-08 2022-10-11 アムジエン・インコーポレーテツド KRas G12Cの阻害剤及びそれを使用する方法
KR20200093562A (ko) 2017-11-29 2020-08-05 씨에스엘 리미티드 허혈-재관류 손상을 치료 또는 예방하는 방법
CR20210319A (es) 2018-01-12 2021-07-27 Amgen Inc ANTICUERPOS ANTI-PD-1 Y MÉTODOS DE TRATAMIENTO (Div. 2020-330)
KR20210013091A (ko) 2018-05-16 2021-02-03 시에스엘 리미티드 가용성 보체 수용체 1형 변이체 및 이의 용도
CN112771075A (zh) 2018-07-30 2021-05-07 安进研发(慕尼黑)股份有限公司 结合至cd33和cd3的双特异性抗体构建体的延长施用
UY38326A (es) 2018-08-03 2020-01-31 Amgen Inc Constructos de anticuerpos para cldn18.2 y cd3
WO2020049151A1 (fr) 2018-09-06 2020-03-12 Bavarian Nordic A/S Compositions de poxvirus améliorées en matière de stockage
CR20220096A (es) 2019-09-06 2022-05-11 Novartis Ag Proteínas de fusión terapéuticas
EP3819007A1 (fr) 2019-11-11 2021-05-12 Amgen Research (Munich) GmbH Régime de dosage pour agents anti-bcma
WO2021111143A1 (fr) 2019-12-04 2021-06-10 Albumedix Limited Procédés et compositions produites par ceux-ci
EP4093771A1 (fr) 2020-01-22 2022-11-30 Amgen Research (Munich) GmbH Combinaisons de constructions d'anticorps et d'inhibiteurs du syndrome de libération de cytokine et leurs utilisations
EP4118113A1 (fr) 2020-03-12 2023-01-18 Amgen Inc. Méthodes de traitement et de prophylaxie du crs chez des patients, comprenant une association d'anticorps bispécifiques se liant à une cellule tumorale du cds x et d'un inhibiteur du tnf alpha ou de l'il-6
BR112022017438A2 (pt) 2020-03-12 2022-10-18 Bavarian Nordic As Composições que melhoram a estabilidade do poxvírus
GB202004514D0 (en) 2020-03-27 2020-05-13 Inst De Medicina Molecular Joaeo Lobo Antunes Treatment of Immunosuppressive Cancer
EP4153633A1 (fr) 2020-05-19 2023-03-29 Amgen Inc. Constructions de liaison à mageb2
EP4157874A2 (fr) 2020-05-29 2023-04-05 Amgen Inc. Administration atténuant des effets indésirables d'une construction d'anticorps bispécifique de liaison à cd33 et cd3
WO2022060901A1 (fr) 2020-09-16 2022-03-24 Amgen Inc. Méthodes d'administration de doses thérapeutiques de molécules bispécifiques activant les lymphocytes t pour le traitement du cancer
MX2023005343A (es) 2020-11-06 2023-05-22 Amgen Res Munich Gmbh Construcciones polipeptidicas que se unen selectivamente a cldn6 y cd3.
KR20230104229A (ko) 2020-11-06 2023-07-07 암젠 인크 Cd3에 결합하는 폴리펩티드 구축물
CA3198064A1 (fr) 2020-11-06 2022-05-12 Amgen Inc. Domaine de liaison a l'antigene a taux de coupure reduit
TW202233682A (zh) 2020-11-10 2022-09-01 美商安進公司 用於投與BCMAxCD3結合分子之方法
CN113717873B (zh) * 2021-09-27 2023-04-14 四川大学 一种多重耐受性酿酒酵母菌株及其构建方法和应用
WO2024077044A1 (fr) 2022-10-05 2024-04-11 Amgen Inc. Polythérapies comprenant des thérapies de redirection de lymphocytes t et des anticorps agonistes anti-il-2r ou des fragments de ceux-ci
WO2024089258A1 (fr) 2022-10-28 2024-05-02 Aarhus Universitet Albumine conjuguée à des oligodésoxynucléotides cpg en tant que super composés adjuvants de la réponse immunitaire

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011051489A2 (fr) * 2009-10-30 2011-05-05 Novozymes Biopharma Dk A/S Variants d'albumine
US20120322739A1 (en) * 2011-05-05 2012-12-20 Novozymes Biopharma Uk Limited Albumin Variants
US20130225496A1 (en) * 2010-11-01 2013-08-29 Novozymes Biopharma Dk A/S Albumin Variants

Family Cites Families (163)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB9019919D0 (en) 1990-09-12 1990-10-24 Delta Biotechnology Ltd Purification of proteins
US5625041A (en) 1990-09-12 1997-04-29 Delta Biotechnology Limited Purification of proteins
US2714586A (en) 1951-06-25 1955-08-02 Phillips Petroleum Co Washing urea and thiourea containing adducts
JPS4983148A (fr) 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 (ja) 1984-02-10 1985-09-02 Kyowa Hakko Kogyo Co Ltd 成人t細胞白血病ウイルス抗原ペプチド誘導体
AU5772886A (en) 1985-04-12 1986-11-05 Genetics Institute Inc. Novel procoagulant proteins
GR860984B (en) 1985-04-17 1986-08-18 Zymogenetics Inc Expression of factor vii and ix activities in mammalian cells
IE63424B1 (en) 1987-04-09 1995-04-19 Delta Biotechnology Ltd Yeast vector
ATE125865T1 (de) 1987-08-28 1995-08-15 Novo Nordisk As Rekombinante humicola-lipase und verfahren zur herstellung von rekombinanten humicola-lipasen.
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
US5766883A (en) 1989-04-29 1998-06-16 Delta Biotechnology Limited Polypeptides
GB8909916D0 (en) 1989-04-29 1989-06-14 Delta Biotechnology Ltd Polypeptides
FR2650598B1 (fr) 1989-08-03 1994-06-03 Rhone Poulenc Sante Derives de l'albumine a fonction therapeutique
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 (de) 1990-01-15 1991-07-18 Brem Gottfried Prof Dr Dr Verfahren zur antikoerpergewinnung
JP3230091B2 (ja) 1990-06-25 2001-11-19 ウェルファイド株式会社 ヒト血清アルブミンの着色抑制方法
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 (fr) 1991-04-25 2003-07-15 Kotikanyad Sreekrishna Cassettes d'expression et vecteurs pour la secretion de serum albumine humaine dans des cellules pichia pastoris
US5264586A (en) 1991-07-17 1993-11-23 The Scripps Research Institute Analogs of calicheamicin gamma1I, method of making and using the same
FR2686899B1 (fr) 1992-01-31 1995-09-01 Rhone Poulenc Rorer Sa Nouveaux polypeptides biologiquement actifs, leur preparation et compositions pharmaceutiques les contenant.
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
DE4244915C2 (de) 1992-08-14 1998-12-03 Widmar Prof Dr Tanner DNA-Moleküle codierend Proteine, die an der O-Glykosylierung von Proteinen beteiligt sind
US5728553A (en) 1992-09-23 1998-03-17 Delta Biotechnology Limited High purity albumin and method of producing
DK0669836T3 (da) 1992-11-13 1996-10-14 Idec Pharma Corp Terapeutisk anvendelse af kimære og radioaktivt mærkede antistoffer og humant B-lymfocytbegrænset differentieringsantigen til behandling af B-cellelymfom
WO1995003402A1 (fr) 1993-07-22 1995-02-02 Merck & Co., Inc. EXPRESSION DE L'INTERLEUKINE-β HUMAINE DANS UN ANIMAL TRANSGENIQUE
DE4343591A1 (de) 1993-12-21 1995-06-22 Evotec Biosystems Gmbh Verfahren zum evolutiven Design und Synthese funktionaler Polymere auf der Basis von Formenelementen und Formencodes
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
EP1030931A4 (fr) 1998-08-06 2004-05-26 Syntron Biores Inc Dispositif de test d'acide urique presentant une composition reactive d'uricase stabilisee
US6444790B1 (en) 1998-12-23 2002-09-03 Human Genome Sciences, Inc. Peptidoglycan recognition proteins
GB9902000D0 (en) 1999-01-30 1999-03-17 Delta Biotechnology Ltd Process
ES2334106T3 (es) 1999-05-17 2010-03-05 Conjuchem Biotechnologies Inc. Inhibidores peptidos de fusion de larga vida contra infecciones virales.
CA2684454A1 (fr) 1999-05-21 2000-11-18 American Bioscience, Llc. Agents a stabilisation proteinique actifs pharmacologiquement; procedes de fabrication et methodes d'utilisation
IL151857A0 (en) 2000-03-22 2003-04-10 Octagene Gmbh Production of recombinant blood clotting factors in human cell lines
AU2001266557A1 (en) 2000-04-12 2001-10-23 Human Genome Sciences, Inc. Albumin fusion proteins
US20030091565A1 (en) 2000-08-18 2003-05-15 Beltzer James P. Binding polypeptides and methods based thereon
ATE510850T1 (de) 2000-09-15 2011-06-15 Coley Pharm Gmbh Verfahren zum hochdurchsatzscreening von auf cpg basierenden immunagonisten und -antagonisten
AU2002245204A1 (en) 2000-10-25 2002-07-24 Genzyme Corporation Methods for treating blood coagulation disorders
WO2002043658A2 (fr) 2000-11-06 2002-06-06 The Jackson Laboratory Traitement a base de fcrn pour troubles autoimmuns
CZ308214B6 (cs) 2000-12-07 2020-03-04 Eli Lilly And Company GLP-1 fúzní proteiny
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
EP2228389B1 (fr) 2001-04-13 2015-07-08 Human Genome Sciences, Inc. Anticorps contre facteur de croissance endothéliale vasculaire 2
AUPR446701A0 (en) 2001-04-18 2001-05-17 Gene Stream Pty Ltd Transgenic mammals for pharmacological and toxicological studies
EP1406917A4 (fr) 2001-06-15 2007-11-14 New Century Pharmaceuticals Modeles animaux exprimant l'albumine humaine pour evaluation de medicaments, etudes toxicologiques et d'immunogenicite
JP2005500841A (ja) 2001-07-27 2005-01-13 アメリカ合衆国 オリゴヌクレオチドを用いるインビボ部位指定変異誘発のためのシステム
CN1405182A (zh) 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 血清白蛋白与粒细胞集落刺激因子的融合蛋白
WO2003059934A2 (fr) 2001-12-21 2003-07-24 Human Genome Sciences, Inc. Proteines de fusion d'albumine
EP2261250B1 (fr) 2001-12-21 2015-07-01 Human Genome Sciences, Inc. Protéines de fusion d'albumine et GCSF
US20080167238A1 (en) 2001-12-21 2008-07-10 Human Genome Sciences, Inc. Albumin Fusion Proteins
WO2003066824A2 (fr) 2002-02-07 2003-08-14 Aventis Behring Gmbh Peptides du domaine kunitz fusionnes a l'albumine
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 (zh) 2002-07-01 2006-02-15 美国福源集团 对多种细胞具刺激增生作用的人血清白蛋白重组融合蛋白
GB0217347D0 (en) 2002-07-26 2002-09-04 Univ Edinburgh Novel albumins
EP1572955A2 (fr) 2002-08-02 2005-09-14 Human Genome Sciences, Inc. Anticorps diriges contre le recepteur c3a
RU2359268C2 (ru) 2002-11-19 2009-06-20 ДиАрДжи ИНТЕРНЭШНЛ, ИНК. Способ диагностики заболеваний путем скрининга тканей, крови или жидкостей организма животных или человека на предмет обнаружения нефизиологических уровней гепсидина и его терапевтическое применение
CA2513213C (fr) 2003-01-22 2013-07-30 Human Genome Sciences, Inc. Proteines hybrides d'albumine
AU2004212263B2 (en) 2003-02-17 2009-12-03 Novozymes Biopharma Dk A/S Conjugates for medical imaging comprising carrier, targeting moiety and a contrast agent
GB0305989D0 (en) 2003-03-15 2003-04-23 Delta Biotechnology Ltd Agent
WO2004082640A2 (fr) 2003-03-19 2004-09-30 New Century Pharmaceuticals, Inc. Fragments ou polymeres d'albumine a temps de residence vasculaire modulable utilisables pour la mise au point de therapies d'administration et de vaccins
WO2004092405A2 (fr) 2003-04-15 2004-10-28 Xenon Pharmaceuticals Inc. Gene d'hemochromatose juvenile (hfe2a), produits d'expression et leurs utilisations
WO2004101620A2 (fr) 2003-05-01 2004-11-25 Compound Therapeutics, Inc. Proteines a base d'echafaudage de serum albumine et utilisations desdites proteines
US7569215B2 (en) 2003-07-18 2009-08-04 Massachusetts Institute Of Technology Mutant interleukin-2 (IL-2) polypeptides
US20080308744A1 (en) 2003-11-18 2008-12-18 Beth Israel Deaconess Medical Center Serum Albumin Conjugated to Fluorescent Substances for Imaging
US9057061B2 (en) 2003-12-23 2015-06-16 Novozymes Biopharma Dk A/S Gene expression technique
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 (fr) 2003-12-26 2013-01-08 Nipro Corporation Albumine possedant une activite antimicrobienne accrue
JP4649954B2 (ja) 2003-12-26 2011-03-16 ニプロ株式会社 抗菌作用が増強されたアルブミン
KR100671005B1 (ko) 2004-01-15 2007-01-18 고려대학교 산학협력단 Pah 노출 여부 진단용 바이오 마커 단백질
HUE027902T2 (en) 2004-02-09 2016-11-28 Human Genome Sciences Inc Corp Service Company Albumin fusion proteins
RU2369404C2 (ru) * 2004-02-09 2009-10-10 Хьюман Дженом Сайенсиз, Инк. Слитые белки альбумина
JP4492156B2 (ja) 2004-03-03 2010-06-30 ニプロ株式会社 血清アルブミンドメインを含む蛋白質
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
ES2579805T3 (es) 2004-09-23 2016-08-16 Genentech, Inc. Anticuerpos y conjugados modificados por ingeniería genética con cisteína
AU2005318697A1 (en) 2004-12-22 2006-06-29 Novozymes A/S Recombinant production of serum albumin
JPWO2006073195A1 (ja) 2005-01-07 2008-06-12 敏一 吉川 糖尿病の予知・診断方法および糖尿病予知・診断用キット
US20060178301A1 (en) 2005-02-04 2006-08-10 Mathias Jurs Albumin-fused ciliary neurotrophic factor
CA2606378A1 (fr) 2005-04-29 2006-11-09 The Jackson Laboratory Anticorops de fcrn et utilisations
GB0512707D0 (en) 2005-06-22 2005-07-27 Delta Biotechnology Ltd Gene expression technique
MX2008001865A (es) 2005-08-12 2008-04-15 Human Genome Sciences Inc Proteinas de fusion de albumina.
CA2562249A1 (fr) 2005-10-20 2007-04-20 University Of Ottawa Heart Institute Analogue du facteur natriuretique auriculaire
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 (fr) 2006-02-06 2007-08-08 CSL Behring GmbH Facteur de coagulation VIIa modifié ayant une stabilité 'half-life' améliorée
WO2007112940A2 (fr) 2006-03-31 2007-10-11 Ablynx N.V. Séquence d'acides aminés dérivée de l'albumine, son utilisation pour augmenter la demi-vie de protéines thérapeutiques et d'autres composés et entités thérapeutiques, et produits de recombinaison qui la comprennent
EP2474318A1 (fr) 2006-06-07 2012-07-11 Human Genome Sciences, Inc. Protéines de fusion d'albumine
ES2889920T3 (es) 2006-06-14 2022-01-14 Csl Behring Gmbh Proteínas de fusión escindibles proteolíticamente con alta actividad específica molar
CA2657277C (fr) 2006-07-13 2015-11-24 Upperton Limited Procede de fabrication de particules d'un materiau proteique
JP4983148B2 (ja) 2006-08-18 2012-07-25 ニプロ株式会社 糖鎖含有アルブミン、その製造方法およびその用途
KR20090060294A (ko) 2006-09-08 2009-06-11 암브룩스, 인코포레이티드 변형된 인간 혈장 폴리펩티드 또는 Fc 스캐폴드 및 그의 용도
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
CN101835801B (zh) 2007-08-08 2014-09-10 诺维信生物制药丹麦公司 转铁蛋白变体和偶联物
WO2009061853A2 (fr) 2007-11-05 2009-05-14 Massachusetts Institute Of Technology Polypeptides d'interleukine-2 (il-2) mutants
CA2611540C (fr) 2007-11-09 2017-05-30 Nipro Corporation Albumine renfermant une chaine de sucre comme porteur de medicament vers le foie
DK2245064T3 (da) 2007-12-21 2014-10-27 Medimmune Ltd BINDINGSELEMENTER TIL INTERLEUKIN-4-RECEPTOR-ALFA (IL-4Ralfa)
JP5639039B2 (ja) 2008-04-11 2014-12-10 メリマック ファーマシューティカルズ インコーポレーティッド ヒト血清アルブミンリンカーおよびそのコンジュゲート
CN102066405B (zh) 2008-04-28 2015-09-30 哈佛大学校长及研究员协会 用于细胞穿透的超荷电蛋白
EP2310526A4 (fr) 2008-07-18 2011-11-02 Oragenics Inc Compositions pour la détection et le traitement du cancer colorectal
WO2010059315A1 (fr) 2008-11-18 2010-05-27 Merrimack Pharmaceuticals, Inc. Lieurs de sérum-albumine humaine et conjugués de ceux-ci
EP2373331A4 (fr) 2008-12-05 2015-11-18 Abraxis Bioscience Llc Ciblage d une maladie à médiation par peptide de liaison à l albumine
US8637306B2 (en) 2008-12-10 2014-01-28 The Scripps Research Institute Production of carrier-peptide conjugates using chemically reactive unnatural amino acids
SG2014012918A (en) 2009-02-11 2014-04-28 Novozymes Biopharma Dk As Albumin variants and conjugates
JP5841046B2 (ja) 2009-04-08 2016-01-06 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア 制御された血清中薬物動態を有するヒトタンパク質スキャフォールド
JP2012525146A (ja) 2009-04-28 2012-10-22 プレジデント アンド フェロウズ オブ ハーバード カレッジ 細胞透過のための過剰に荷電されたタンパク質
US8232067B2 (en) 2009-05-29 2012-07-31 Brigham & Women's Hospital, Inc. Disrupting FCRN-albumin interactions
ES2548030T3 (es) 2009-06-01 2015-10-13 Medimmune, Llc Moléculas con semividas prolongadas y usos de las mismas
KR101286721B1 (ko) 2009-06-05 2013-07-16 한국과학기술연구원 폴리-시스테인 펩티드 융합 재조합 알부민 및 이의 제조방법
US20110015130A1 (en) 2009-07-20 2011-01-20 Woei-Jer Chuang Polypeptides Selective for alphavbeta3 Integrin Conjugated With a Variant Of Human Serum Albumin (HSA) And Pharmaceutical Uses Thereof
WO2011011797A2 (fr) 2009-07-24 2011-01-27 The Board Of Trustees Of The Leland Stanford Junior University Compositions de cytokine et leurs méthodes d’utilisation
BR112012003089A2 (pt) 2009-08-10 2016-08-16 Ucl Business Plc ligação covalente reversível de moléculas funcionais
UY32920A (es) 2009-10-02 2011-04-29 Boehringer Ingelheim Int Moleculas de unión biespecíficas para la terapia anti-angiogénesis
CA2777222A1 (fr) 2009-10-10 2011-04-14 Eleven Biotherapeutics, Inc. Compositions de cytokines de la famille il-17 et utilisations
WO2011079175A1 (fr) 2009-12-23 2011-06-30 National Cheng Kung University Compositions et méthodes pour le traitement de maladies ophtalmiques liées à une angiogenèse
CN101875693B (zh) 2010-01-22 2012-07-18 成都正能生物技术有限责任公司 具备抗血管生成活性的白蛋白变异体及其制备方法
JP2013519392A (ja) 2010-02-16 2013-05-30 メディミューン,エルエルシー Hsa関連組成物および使用方法
WO2011124718A1 (fr) * 2010-04-09 2011-10-13 Novozymes A/S Dérivés et variants d'albumine
ES2674567T3 (es) 2010-05-21 2018-07-02 Merrimack Pharmaceuticals, Inc. Proteínas de fusión biespecíficas
WO2011161127A1 (fr) 2010-06-21 2011-12-29 Medimmune, Llc Variants de protéase
EP2603522A1 (fr) 2010-08-13 2013-06-19 GlaxoSmithKline Intellectual Property Development Limited Variants de liaison anti-sérum-albumine améliorés
JP2014510518A (ja) * 2011-02-15 2014-05-01 メディミューン,エルエルシー Hsa関連組成物および使用方法
US9045564B2 (en) 2011-02-15 2015-06-02 Medimmune, Llc HSA-related compositions and methods of use
JP5641536B2 (ja) * 2011-03-15 2014-12-17 日本パーカライジング株式会社 固定砥粒ソーワイヤー用電着液
KR20140053991A (ko) 2011-07-18 2014-05-08 아츠 바이올로직스 에이/에스 장기간 작용하는 황체 형성 호르몬 (lh) 화합물
US20140315817A1 (en) 2011-11-18 2014-10-23 Eleven Biotherapeutics, Inc. Variant serum albumin with improved half-life and other properties
PL2825556T3 (pl) 2012-03-16 2018-10-31 Albumedix A/S Warianty albuminy
WO2014005596A1 (fr) 2012-07-03 2014-01-09 Aarhus Universitet Molécules cargo modifiées, leurs interactions et leurs utilisations
US8757798B2 (en) 2012-08-31 2014-06-24 Cody Air LLC Eyewear frame
CA2890766A1 (fr) 2012-11-08 2014-05-15 Novozymes Biopharma Dk A/S Variants d'albumine
CN105007722A (zh) 2013-02-16 2015-10-28 诺维信生物制药丹麦公司 药代动力学动物模型
WO2014179657A1 (fr) 2013-05-03 2014-11-06 Eleven Biotherapeutics, Inc. Variantes d'albumine se liant à fcrn
US20160222087A1 (en) 2013-09-13 2016-08-04 Novozymes Biopharma Dk A/S Albumin variants
EP3063171B1 (fr) 2013-11-01 2019-07-24 University Of Oslo Variants d'albumine et utilisations de ceux-ci
CA2989966C (fr) 2015-08-20 2024-04-30 Albumedix A/S Variants de l'albumine et leurs conjugues

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011051489A2 (fr) * 2009-10-30 2011-05-05 Novozymes Biopharma Dk A/S Variants d'albumine
US20120220530A1 (en) * 2009-10-30 2012-08-30 Novozymes Biopharma Dk A/S Albumin variants
US20130225496A1 (en) * 2010-11-01 2013-08-29 Novozymes Biopharma Dk A/S Albumin Variants
US20120322739A1 (en) * 2011-05-05 2012-12-20 Novozymes Biopharma Uk Limited Albumin Variants

Cited By (20)

* 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
US20160075762A1 (en) * 2009-10-30 2016-03-17 Novozymes Biopharma Dk A/S Albumin variants
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
US10711050B2 (en) 2011-11-18 2020-07-14 Albumedix Ltd Variant serum albumin with improved half-life and other properties
US9944691B2 (en) 2012-03-16 2018-04-17 Albumedix A/S Albumin variants
US10329340B2 (en) 2012-03-16 2019-06-25 Albumedix Ltd Albumin variants
US10501524B2 (en) 2012-11-08 2019-12-10 Albumedix Ltd Albumin variants
US10934341B2 (en) 2012-11-08 2021-03-02 Albumedix, Ltd. Albumin variants
US9926570B2 (en) * 2013-03-06 2018-03-27 Glaxosmithkline Llc Host cells and methods of use
US20160017343A1 (en) * 2013-03-06 2016-01-21 Glaxosmithkline Llc Host cells and methods of use
US11661462B2 (en) * 2014-07-31 2023-05-30 Amgen Research (Munich) Gmbh Optimized cross-species specific bispecific single chain antibody contructs
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
US11447567B2 (en) 2015-07-31 2022-09-20 Amgen Research (Munich) Gmbh Antibody constructs for FLT3 and CD3
US11884720B2 (en) 2015-07-31 2024-01-30 Amgen Research (Munich) Gmbh Antibody constructs for MSLN and CD3
US10633428B2 (en) 2015-08-20 2020-04-28 Albumedix Ltd Albumin variants and conjugates
US11434302B2 (en) 2016-02-03 2022-09-06 Amgen Research (Munich) Gmbh Bispecific T cell engaging antibody constructs
US20220031741A1 (en) * 2016-06-03 2022-02-03 New York University Methods and reagents for modulating macrophage phenotype
CN109553684A (zh) * 2017-09-25 2019-04-02 中国科学院过程工程研究所 一种纳米载体蛋白及其制备方法和应用
CN113912730A (zh) * 2021-12-14 2022-01-11 北京科诺信诚科技有限公司 缓释的抗FcRn抗体或抗原结合片段及其应用

Also Published As

Publication number Publication date
US10934341B2 (en) 2021-03-02
GB2512156A (en) 2014-09-24
AU2013343503B2 (en) 2017-12-14
IL238185B (en) 2019-09-26
BR112015010318A2 (pt) 2017-08-22
MX2015005363A (es) 2015-11-06
GB201319754D0 (en) 2013-12-25
CN105452290A (zh) 2016-03-30
AU2013343503A1 (en) 2015-04-30
US20150210752A1 (en) 2015-07-30
KR20150082422A (ko) 2015-07-15
JP6487328B2 (ja) 2019-03-20
RU2015121693A (ru) 2016-12-27
IL238185A0 (en) 2015-05-31
EP2917233A1 (fr) 2015-09-16
JP2019048821A (ja) 2019-03-28
RU2670063C2 (ru) 2018-10-17
CA2890766A1 (fr) 2014-05-15
AU2018201136A1 (en) 2018-03-08
WO2014072481A1 (fr) 2014-05-15
US10501524B2 (en) 2019-12-10
US20200102367A1 (en) 2020-04-02
JP2015535300A (ja) 2015-12-10

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
AS Assignment

Owner name: NOVOZYMES BIOPHARMA DK A/S, DENMARK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:CAMERON, JASON;DELAHAY, KAREN ANN;PLUMRIDGE, ANDREW;AND OTHERS;SIGNING DATES FROM 20131210 TO 20131218;REEL/FRAME:032461/0595

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION