EP1924596A2 - Protéines de fusion avec l'albumine - Google Patents

Protéines de fusion avec l'albumine

Info

Publication number
EP1924596A2
EP1924596A2 EP06813242A EP06813242A EP1924596A2 EP 1924596 A2 EP1924596 A2 EP 1924596A2 EP 06813242 A EP06813242 A EP 06813242A EP 06813242 A EP06813242 A EP 06813242A EP 1924596 A2 EP1924596 A2 EP 1924596A2
Authority
EP
European Patent Office
Prior art keywords
albumin
therapeutic
variant
fragment
protein
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.)
Withdrawn
Application number
EP06813242A
Other languages
German (de)
English (en)
Other versions
EP1924596A4 (fr
Inventor
Craig Rosen
Adam Bell
Paul Moore
Yanggu Shi
David Lafleur
Michael Laird
William Haseltine
Douglas Woods
Jason Bock
Mani Subramanian
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.)
Human Genome Sciences Inc
Original Assignee
Human Genome Sciences Inc
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 Human Genome Sciences Inc filed Critical Human Genome Sciences Inc
Publication of EP1924596A2 publication Critical patent/EP1924596A2/fr
Publication of EP1924596A4 publication Critical patent/EP1924596A4/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • 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
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/643Albumins, e.g. HSA, BSA, ovalbumin or a Keyhole Limpet Hemocyanin [KHL]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • the invention relates generally to Therapeutic proteins (including, but not limited to, at least one polypeptide, antibody, peptide, or fragment and variant thereof) fused to albumin or fragments or variants of albumin.
  • the invention encompasses polynucleotides encoding therapeutic albumin fusion proteins, therapeutic albumin fusion proteins, compositions, pharmaceutical compositions, formulations and kits.
  • Host cells transformed with the polynucleotides encoding therapeutic albumin fusion proteins are also encompassed by the invention, as are methods of making the albumin fusion proteins of the invention using these polynucleotides, and/or host cells.
  • HSA Human serum albumin
  • HA a protein of 585 amino acids in its mature form (as shown in Figure 1 (SEQ ID N ⁇ :l)), is responsible for a significant proportion of the osmotic pressure of serum and also functions as a carrier of endogenous and exogenous ligands.
  • HSA Human serum albumin
  • rHA recombinant HA
  • increasing " the dosing schedule for administration of the therapeutic protein often results in an increase in injection site reactions, side-effects, and toxicity in the patient.
  • administration of the therapeutic protein at a higher dose also commonly results in an increase in toxicity and side-effects in the patient.
  • the present invention encompasses albumin fusion proteins comprising a Therapeutic protein (e.g., a polypeptide, antibody, or peptide, or fragment or variant thereof) fused to albumin or a fragment (portion) or variant of albumin.
  • a Therapeutic protein e.g., a polypeptide, antibody, or peptide, or fragment or variant thereof
  • the present invention also encompasses polynucleotides comprising, or alternatively consisting of, nucleic acid molecules encoding a Therapeutic protein (e.g., a polypeptide, antibody, or peptide, or fragment or variant thereof) fused to albumin or a fragment (portion) or variant of albumin.
  • the present invention also encompasses polynucleotides, comprising, or alternatively consisting of, nucleic acid molecules encoding proteins comprising a Therapeutic protein (e.g., a polypeptide, antibody, or peptide, or fragment or variant thereof) fused to albumin or a fragment (portion) or variant of albumin, that is sufficient to prolong the shelf life of the Therapeutic protein, to increase the plasma stability of the Therapeutic protein compared to its unfused state, and/or stabilize the Therapeutic protein and/or its activity in solution (or in a pharmaceutical composition) in vitro and/or in vivo.
  • a Therapeutic protein e.g., a polypeptide, antibody, or peptide, or fragment or variant thereof
  • Albumin fusion proteins encoded by a polynucleotide of the invention are also encompassed by the invention, as are host cells transformed with polynucleotides of the invention, and methods of making the albumin fusion proteins of the invention and using these polynucleotides of the invention, and/or host cells.
  • albumin fusion proteins include, but are not limited to, those described in Table 2 and the polynucleotides encoding such proteins.
  • the invention also encompasses pharmaceutical formulations comprising an albumin fusion protein of the invention and a pharmaceutically acceptable diluent or carrier.
  • Such formulations may be in a kit or container.
  • kit or container may be packaged with instructions pertaining to the extended shelf life of the Therapeutic protein.
  • Such formulations may be used in methods of treating, preventing, ameliorating or diagnosing a disease or disease symptom in a patient, preferably a mammal, most preferably a human, comprising the step of administering the pharmaceutical formulation to the patient.
  • the present invention encompasses methods of preventing, treating, or ameliorating a disease or disorder
  • the present invention encompasses a method of treating a disease or disorder listed in the "Preferred Indication: Y" column of Table 1 comprising administering to a patient in which such treatment, prevention or amelioration is desired an albumin fusion protein of the invention that comprises a Therapeutic protein or portion corresponding to a Therapeutic protein (or fragment or variant thereof) disclosed in the
  • Therapeutic Protein: X column of Table 1 (in the same row as the disease or disorder to be treated as listed in the "Preferred Indication: Y” column of Table 1) in an amount effective to treat, prevent or ameliorate the disease or disorder.
  • an albumin fusion protein described in Table 1 or 2 has extended shelf life.
  • an albumin fusion protein described in Table 1 or 2 is more stable than the corresponding unfused Therapeutic molecule described in Table 1.
  • the present invention further includes transgenic organisms modified to contain the nucleic acid molecules of the invention (including, but not limited to, the polynucleotides described in Tables 1 and 2), preferably modified to express an albumin fusion protein of the invention.
  • Figure IA-D shows the amino acid sequence of the mature form of human albumin (SEQ ID NO:1) and a polynucleotide encoding it (SEQ ID N ⁇ :2). Nucleotides 1 to 17 55 of SEQ ID N ⁇ :2 encode the mature form of human albumin (SEQ ID NO:1).
  • Figure 2 shows the restriction map of the pPPC000 5 cloning vector ATCC deposit PTA-3278.
  • Figure 3 shows the restriction map of the pSAC3 5 yeast S. cerevisiae expression vector (Sleep et ah, BioTechnology 8:42 (1990)).
  • Figure 4 compares the anti-proliferative activity of IFN albumin fusion protein encoded by CID 3165 (CID 3165 protein) and recombinant IFNa (rJFNa) on Hs294T melanoma cells. The cells were cultured with varying concentrations of either CID 3165 protein or rIFNa and proliferation was measured by BrdU incorporation after 3 days of culture. CID 3165 protein caused measurable inhibition of cell proliferation at concentrations above 10 ng/ml with 50% inhibition achieved at approximately 200 ng/ml.
  • FIG. 5 shows the effect of various dilutions of IFNa albumin fusion proteins on SEAP activity in the ISRE-SEAP/293F reporter cells.
  • One preparation of EFNa fused upstream of albumin ( ⁇ ) was tested, as well as two different preparations of IFNa fused downstream of albumin (•) and ( ⁇ ).
  • FIG. 6 shows the effect of time and-dose of IFNa albumin fusion protein encoded -by DNA comprised in construct 2249 (CID 2249 protein) on the mRNA level of OAS (p41) in treated monkeys (see Example 76).
  • first bar Vehicle control
  • 2 nd bar 30 ug/kg CID 2249 protein day 1 iv
  • third bar 30 ug/kg CID 2249 protein day 1 sc
  • 4 th bar 300 ug/kg CID 2249 protein day 1 sc
  • 5 01 bar 40 ug/kg recombinant IFNa day 1, 3 and 5 sc.
  • FIG. 7 shows the dose-response relationship of BNP albumin fusion proteins encoded by DNA comprised in constructs CID 3691 and 3618 (CID 3691 and 3618 protein) on activating cGMP formation in NPR-A/293F reporter cells (see Examples 78 and 79). Both BNP peptide ( ⁇ ), as well as, two different preparations of BNP fused upstream of albumin (D) and (•) were tested.
  • Figure 8 shows the effect of BNP albumin fusion protein on mean arterial pressure in spontaneously hypertensive rats (see Example 78).
  • Vehicle (D), BNP peptide (•), or BNP albumin fusion protein (O) were delivered via tail vein injection. Systolic and diastolic blood pressures were recorded by cuff-tail method.
  • Figure 9 shows the plasma cGMP levels in eleven- to 12-week-old male C57/BL6 mice after intravenous injection of recombinant BNP peptide (•) or BNP albumin fusion protein (O (see Example 78). cGMP levels were determined from plasma prepared from tail bleeds collected at several time points after intravenous injection.
  • Figure 10 shows the dose-response relationship of BNP peptide and BNP albumin fusion proteins encoded by DNA comprised in constructs CID 3796 and 3959 on activating cGMP formation in NPR-A/293F reporter cells (see Example 80). Both BNP peptide ( ⁇ ), as well as, two different preparations comprising BNP fused downstream of albumin, (D) and (O) were tested.
  • Figure 1 IA shows the dose-response relationship of BNP and ANP peptides with or without treatment of neprilysin for 24 hours on activating cGMP formation in NPR-A/293F reporter cells (see Example 81).
  • Figure HB shows the dose-reponse relationship of ANP peptide on activating cGMP formation in NPR-A/293F reporter cells following treatment of neprilysin or control MES buffer for 20 minutes, 1 hour, or 24 hours (see Example 81).
  • Figure HC shows the dose-reponse relationship of ANP albumin fusion protein comprising ANP fused upstream of albumin and encoded by DNA comprised in construct CID3484 on activating cGMP formation in NPR-A/293F reporter cells following treatment of neprilysin or control MES buffer for 20 minutes, 1 hour, or 24 hours (see Example 81).
  • Figure 1 ID shows the percentage of intact natriuretic peptides following treatment with neprilysin for the specified time.
  • ANP and BNP peptides as well as, two albumin fusion proteins comprising BNP fused upstream of albumin via tripartite glycines (CID 3809) and ANP fused upstream to albumin (CID 3484) were tested (see Example 81).
  • Figure 12 shows the reduction in HCV RNA titer, as measured by median HCV RNA change (logio IU/ml), in patients infected with chronic hepatitis C genotype 1 and who have previously failed to respond to at least one treatment regimen of pegylated interferon alpha in combination with ribavirin (PEG-RBV) (nonresponders) following treatment with HSA-IFN ⁇ 2b in combination with ribavirin for 0 to 24 weeks.
  • FIGs 15A-H show the hemodynamic effects of HSA-BNP (Construct ID #3959) administered via a single intravenous bolus at 0.5 mg/kg or 5 mg/kg in a normal dog model.
  • Asterisks indicate statistically significant changes from baseline (p ⁇ 0.05).
  • FIGs 16A-H show the renal effects of HSA-BNP (Construct ID #3959) administered via a single intravenous bolus at 0.5 mg/kg or 5 mg/kg in a normal dog model.
  • Urine flow rate/30 minute collection
  • sodium excretion sodium excretion
  • Asterisks indicate statistically significant changes from baseline (p ⁇ 0.05).
  • FIGS 17A-F show the hormonal effects of HSA-BNP (Construct ID #3959) administered via a single intravenous bolus at 0. 5 mg/kg or 5 mg/kg in a normal dog model.
  • - Asterisks indicate statistically significant changes from baseline (p ⁇ 0.05).
  • Figures 18A-C show the effect of a single intravenous bolus of 5 mg/kg HSA-BNP (Construct ID #3959) on systolic and mean arterial blood pressure in normal, healthy, awake beagles surgically implanted with a Data Sciences International radiotelemetry transmitter, which had systemic arterial blood pressure, heart rate and ECG data collection capabilities.
  • Change from baseline of systolic blood pressure ( Figure 18A), difference in mean systolic blood pressure (Figure 18B), and change from baseline in mean arterial pressures (Figure 18C) over 48 hours of continuous data recording following infusion are presented.
  • Asterisks indicate a statistically significant difference in baseline-adjusted mean values for 5 mg/kg HSA-BNP (Construct ID #3959) compared to vehicle (p ⁇ 0.05).
  • Figures 19A and B show a comparison of the effect of an intravenous bolus of 0.02 mg/kg unfused BNP peptide and a subcutaneous injection of 10 mg/kg HSA-BNP (Construct ID #3959) on systemic blood pressure in normal, healthy, awake beagles surgically implanted with a Data Sciences International radiotelemetry transmitter, which had systemic arterial blood pressure, heart rate and ECG data collection capabilities. Change from baseline of systolic blood pressure over 48 hours of continuous data recording following administration of BNP ( Figure 19A) and HSA-BNP (Construct ID #3959) are presented. Asterisks indicate a statistically significant difference in baseline-adjusted mean values for 5 mg/kg HSA-BNP (Construct ID #3959) compared to vehicle (p ⁇ 0.05).
  • polynucleotide refers to a nucleic acid molecule having a nucleotide sequence encoding a fusion protein comprising, or alternatively consisting of, at least one molecule of albumin (or a fragment or variant thereof) joined in frame to at least one Therapeutic protein X (or fragment or variant thereof); a nucleic acid molecule having a nucleotide sequence encoding a fusion protein comprising, or alternatively consisting of, the amino acid sequence of SEQ ID NO:Y (as described in column 6 of Table 2) or a fragment or variant thereof; a nucleic acid molecule having a nucleotide sequence comprising or alternatively consisting of the sequence shown in SEQ ID NO:X; a nucleic acid molecule having a nucleotide sequence encoding a fusion protein comprising, or alternatively consisting of, the amino acid sequence of SEQ
  • nucleic acid molecule having a nucleotide sequence encoding a Therapeutic albumin fusion protein of the invention examples include a nucleic acid molecule having a nucleotide sequence contained in an albumin fusion construct described in Table 2, or a nucleic acid molecule having a nucleotide sequence contained in an albumin fusion construct deposited with the ATCC (as described in Table 3).
  • albumin fusion construct refers to a nucleic acid molecule comprising, or alternatively consisting of, a polynucleotide encoding at least one molecule of albumin (or a fragment or variant thereof) joined in frame to at least one polynucleotide encoding at least one molecule of a Therapeutic protein (or fragment or variant thereof); a nucleic acid molecule comprising, or alternatively consisting of, a polynucleotide encoding at least one molecule of albumin (or a fragment or variant thereof) joined in frame to at least one polynucleotide encoding at least one molecule of a Therapeutic protein (or fragment or variant thereof) generated as described in Table 2 or in the Examples; or a nucleic acid molecule comprising, or alternatively consisting of, a polynucleotide encoding at least one molecule of albumin (or a fragment or variant thereof) joined in frame to at least one polynucleotide
  • the present invention relates generally to polynucleotides encoding albumin fusion proteins; albumin fusion proteins; and methods of treating, preventing, or ameliorating diseases or disorders using albumin fusion proteins or polynucleotides encoding albumin fusion proteins.
  • albumin fusion protein refers to a protein formed by the fusion of at least one molecule of albumin (or a fragment or variant thereof) to at least one molecule of a Therapeutic protein (or fragment or variant thereof).
  • An albumin fusion protein of the invention comprises at least a fragment or variant of a Therapeutic protein and at least a fragment or variant of human serum albumin, which are associated with one another by genetic fusion (i.e., the albumin fusion protein is generated by translation of a nucleic acid in which a polynucleotide encoding all or a portion of a Therapeutic protein is joined in-frame with a polynucleotide encoding all or a portion of albumin).
  • the Therapeutic protein and albumin protein, once part of the albumin fusion protein may each be referred to as a "portion", "region” or "moiety" of the albumin fusion protein (e.g., a "Therapeutic protein portion" or an "albumin protein portion”).
  • an albumin fusion protein of the invention comprises at least one molecule of a Therapeutic protein X or fragment or variant of thereof (including, but not limited to a mature form of the Therapeutic protein X) and at least one molecule of albumin or fragment or variant thereof (including but not limited to a mature form of albumin).
  • an albumin fusion protein of the invention is processed by a host cell and secreted into the surrounding culture medium. Processing of the nascent albumin fusion protein that occurs in.
  • the secretory pathways of the host used for expression- may include, but is not limited to signal peptide cleavage; formation of disulfide bonds; proper folding; addition and processing of carbohydrates (such as for example, N- and O- linked glycosylation); specific proteolytic cleavages; and assembly into multimeric proteins.
  • An albumin fusion protein of the invention is preferably in the processed form.
  • the "processed form of an albumin fusion protein" refers to an albumin fusion protein product which has undergone N- terminal signal peptide cleavage, herein also referred to as a "mature albumin fusion protein".
  • a representative clone containing an albumin fusion construct of the invention was deposited with the American Type Culture Collection (herein referred to as "ATCC®"). Furthermore, it is possible to retrieve a given albumin fusion construct from the deposit by techniques known in the art and described elsewhere herein.
  • the ATCC® is located at 10801 University Boulevard, Manassas, Virginia 20110- 2209, USA. The ATCC® deposits were made pursuant to the terms of the Budapest Treaty on the international recognition of the deposit of microorganisms for the purposes of patent procedure.
  • the invention provides a polynucleotide encoding an albumin fusion protein comprising, or alternatively consisting of, a Therapeutic protein and a serum albumin protein.
  • the invention provides an albumin fusion protein comprising, or alternatively consisting of, a Therapeutic protein and a serum albumin protein.
  • the invention provides an albumin fusion protein comprising, or alternatively consisting of, a Therapeutic protein and a serum albumin protein encoded by a polynucleotide described in Table 2.
  • the invention provides a polynucleotide encoding an albumin fusion protein whose sequence is shown as SEQ ID NO:Y in Table 2.
  • the invention provides an albumin fusion protein comprising, or alternatively consisting of, a biologically active and/or therapeutically active fragment of a Therapeutic protein and a serum albumin protein.
  • the invention provides an albumin fusion protein comprising, or alternatively consisting of, a biologically active and/or therapeutically active variant of a Therapeutic protein and a serum albumin protein.
  • the serum albumin protein component of the albumin fusion protein is the mature portion of serum albumin.
  • the invention further encompasses polynucleotides encoding these albumin fusion proteins.
  • the invention provides an albumin fusion protein comprising, or alternatively consisting of, a Therapeutic protein, and a biologically active and/or therapeutically active fragment of serum albumin.
  • the invention provides an albumin fusion protein comprising, or alternatively consisting of, a Therapeutic protein and a biologically active and/or therapeutically active variant of serum albumin.
  • the Therapeutic protein portion of the albumin fusion protein is the mature portion of the Therapeutic protein.
  • the Therapeutic protein portion of the albumin fusion protein is the extracellular soluble domain of the Therapeutic protein.
  • the Therapeutic protein portion of the albumin fusion protein is the active form of the Therapeutic protein.
  • the invention further encompasses polynucleotides encoding these albumin fusion proteins.
  • the invention provides an albumin fusion protein comprising, or alternatively consisting of, a biologically active and/or therapeutically active fragment or variant of a Therapeutic protein and a biologically active and/or therapeutically active fragment or variant of serum albumin.
  • the invention provides an albumin fusion protein comprising, or alternatively consisting of, the mature portion of a Therapeutic protein and the mature portion of serum albumin.
  • the invention further encompasses polynucleotides encoding these albumin fusion proteins.
  • a polynucleotide of the invention encodes a protein comprising or alternatively consisting of, at least a fragment or variant of a Therapeutic protein and at least a fragment or variant of human serum albumin, which are associated with one another, preferably by genetic fusion.
  • An additional embodiment includes a polynucleotide encoding a protein comprising or alternatively consisting of at least a fragment or variant of a Therapeutic protein and at least a fragment or variant of human serum albumin, which are linked with one another by chemical conjugation.
  • Therapeutic protein refers to proteins, polypeptides, antibodies, peptides or fragments or variants thereof, having one or more therapeutic and/or biological activities.
  • Therapeutic proteins encompassed by the invention include but are not limited to, proteins, polypeptides, peptides, antibodies, and biologies. (The terms peptides, proteins, and polypeptides are used interchangeably herein.) It is specifically contemplated that the term “Therapeutic protein” encompasses antibodies and fragments and variants thereof.
  • a protein of the invention may contain at least a fragment or variant of a Therapeutic protein, and/or at least a fragment or variant of an antibody. Additionally, the term “Therapeutic protein” may refer to the endogenous or naturally occurring correlate of a Therapeutic protein.
  • a polypeptide displaying a “therapeutic activity” or a protein that is “therapeutically active” is meant a polypeptide that possesses one or more known biological and/or therapeutic activities associated with a therapeutic protein such as one or more of the Therapeutic proteins described herein or otherwise known in the art.
  • a "Therapeutic protein” is a protein that is useful to treat, prevent or ameliorate a disease, condition or disorder.
  • a "Therapeutic protein” may be one that binds specifically to a particular cell type (normal (e.g., lymphocytes) or abnormal e.g., (cancer cells)) and therefore may be used to target a compound (drug, or cytotoxic agent) to that cell type specifically.
  • a particular cell type normal (e.g., lymphocytes) or abnormal e.g., (cancer cells)
  • a compound drug, or cytotoxic agent
  • a non-exhaustive list of "Therapeutic protein" portions which may be comprised by an albumin fusion protein of the invention includes, but is not limited to, IFN ⁇ , , ANP, BNP, LANP, VDP, KUP, CNP, DNP, HCC-I, beta defensin-2, fractalkine, oxyntomodulin, killer toxin peptide, TIMP-4, PYY, adrenomedullin, ghrelin, CGRP, IGF-I, neuraminidase, hemagglutinin, butyrylcholinesterase, endothelin, and mechano growth factor.
  • Interferon hybrids may also be fused to the amino or carboxy terminus of albumin to form an interferon hybrid albumin fusion protein.
  • Interferon hybrid albumin fusion protein may have enhanced, or alternatively, suppressed interferon activity, such as antiviral responses, regulation of cell growth, and modulation of immune response (Lebleu et al, PNAS USA, 73:3107-3111 (1976); Gresser et al., Nature, 2 5 l: 5 43- 5 4 5 (1974); and Johnson, Texas Reports Biol Med, 35:3 5 7-369 (1977)).
  • Each interferon hybrid albumin fusion protein can be used to treat, prevent, or ameliorate viral infections ⁇ e.g., hepatitis ⁇ e.g., HCV); or VSSf), multiple sclerosis, or cancer.
  • the interferon hybrid portion of the interferon hybrid albumin fusion protein comprises an interferon alpha-interferon alpha hybrid (herein referred to as an alpha-alpha hybrid).
  • the alpha-alpha hybrid portion of the interferon hybrid albumin fusion protein consists, or alternatively comprises, of interferon alpha A fused to interferon alpha D.
  • the A/D hybrid is fused at the common BgHI restriction site to interferon alpha D, wherein the N-terminal portion of the A/D hybrid corresponds to amino acids 1-62 of interferon alpha A and the C-terminal portion corresponds to amino acids 64-166 of interferon alpha D.
  • this A/D hybrid would comprise the amino acid sequence: CDLPQraSLGSRRm ⁇ LAQMRXiISLFSCLKI>R ⁇
  • the A/D hybrid is fused at the common PvuIH restriction site, wherein the N- terminal portion of the A/D hybrid corresponds to amino acids 1-91 of interferon alpha A and the C-terminal portion corresponds to amino acids 93- 166 of interferon alpha D.
  • this A/D hybrid would comprise the amino acid sequence:
  • the alpha-alpha hybrid portion of the interferon hybrid albumin fusion protein consists, or alternatively comprises, of interferon alpha A fused to interferon alpha F.
  • the A/F hybrid is fused at the common PvuIH restriction site, wherein the N-terminal portion of the A/F hybrid corresponds to amino acids 1-91 of interferon alpha A and the C-terminal portion corresponds to amino acids 93-166 of interferon alpha F.
  • this A/F hybrid would comprise the amino acid sequence:
  • the alpha-alpha hybrid portion of the interferon hybrid albumin fusion protein consists, or alternatively comprises, of interferon alpha A fused to interferon alpha B.
  • the A/B hybrid is fused at the common PvuHI restriction site, wherein the N- terminal portion of the A/B hybrid corresponds to amino acids 1-91 of interferon alpha A and the C-terminal portion corresponds to amino acids 93- 166 of interferon alpha B.
  • this A/B hybrid would comprise an amino acid sequence:
  • CDLPQTHSLGSPJITLMLLAQMRX 1 ISLFSCLKDRHDFGFPQEEFGNQFQKAETrPVLHEMIQQIFNLFSTKDSSAAWDETLLDKFYTELYQQL NDLEX 2 X 3 X 4 X 5 QEVGVmSPLMYEDSrLAVRKYFQPJTLYLTEKKYSSCAWEVVP ⁇ EIMRSFSLSlNLQKRLKSKE (SEQ ID NO: 102), wherein the X 1 is R or K and X 2 through X 5 is SCVM or VLCD.
  • the interferon hybrid portion of the interferon hybrid albumin fusion protein comprises an interferon beta- interferon alpha hybrid (herein referred to as a beta-alpha hybrid).
  • the beta-alpha hybrid portion of the interferon hybrid albumin fusion protein consists, or alternatively comprises, of interferon beta-1 fused to interferon alpha D (also referred to as interferon alpha-1).
  • the beta- I/alpha D hybrid is fused wherein the N-terminal portion corresponds to amino acids 1-73 of interferon beta-1 and the C-terminal portion corresponds to amino acids 74-167 of interferon alpha D.
  • this beta-1/alpha D hybrid would comprise an amino acid sequence:
  • the interferon hybrid portion of the interferon hybrid albumin fusion protein comprises an interferon alpha- interferon beta hybrid (herein referred to as a alpha-beta hybrid).
  • the alpha-beta hybrid portion of the interferon hybrid albumin fusion protein consists, or alternatively comprises, of interferon alpha D (alsojeferred to .as interferon alpha-1) fused to interferon beta-1.
  • the alpha D/beta-1 hybrid is fused wherein the N-terminal portion corresponds to amino acids 1-73 of interferon alpha D and the C- terminal portion corresponds to amino acids 74-166 of interferon beta-1.
  • this alpha D/beta-1 hybrid would have an amino acid sequence:
  • the interferon hybrid portion of the interferon hybrid albumin fusion proteins may comprise additional combinations of alpha-alpha interferon hybrids, alpha-beta interferon hybrids, and beta-alpha interferon hybrids.
  • the interferon hybrid portion of the interferon hybrid albumin fusion protein may be modified to include mutations, substitutions, deletions, or additions to the amino acid sequence of the interferon hybrid. Such modifications to the interferon hybrid albumin fusion proteins may be made, for example, to improve levels of production, increase stability, increase or decrease activity, or confer new biological properties.
  • interferon hybrid albumin fusion proteins are encompassed by the invention, as are host cells and vectors containing polynucleotides encoding the polypeptides.
  • a interferon hybrid albumin fusion protein encoded by a polynucleotide as described above has extended shelf life.
  • a interferon hybrid albumin fusion protein encoded by a polynucleotide described above has a longer serum half-life and/or more stabilized activity in solution (or in a pharmaceutical composition) in vitro and/or in vivo than the corresponding unfused interferon hybrid molecule.
  • a "Therapeutic protein” is a protein that has a biological activity, and in particular, a biological activity that is useful for treating, preventing or ameliorating a disease.
  • a non-inclusive list of biological activities that may be possessed by a Therapeutic protein includes, inhibition of HTV-I infection of cells, stimulation of intestinal epithelial cell proliferation, reducing intestinal epithelial cell permeability, stimulating insulin secretion, induction of bronchodilation and vasodilation, inhibition of aldosterone and renin secretion, blood pressure regulation, promoting neuronal growth, enhancing an immune response, enhancing inflammation, suppression of appetite, or any one or more of the biological activities described in the "Biological Activities" section below and/or as disclosed for a given Therapeutic protein in Table 1 (column 2).
  • IFN-alpha-HSA fusions are used to inhibit viral agents classified under Category A- FiIo (Ebola), Arena (Piohende), Category B- Toga (VEE) or Category C- Bunya (Punto toro), Flavi (Yellow fever, West Nile).
  • CPE inhibition, neutral red staining and virus yield assays were employed to evaluate the antiviral activities of INF-alpha fused downstream of HSA (CID 316S protein).
  • CID 3165 protein The pharmacokinetics and pharmacodynamic activity of CID 3165 protein in cynomolgus monkeys and human subjects were evaluated. The results indicate that antiviral activity was achieved against all the RNA viruses evaluated with a favorable safety index.
  • the IC5Q values ranged from ⁇ 0.1 ng/ml (Punta Toro A) to 19 ng/ml (VEE) in the CPE assay.
  • Punta Toro A Punta Toro A
  • VEE 19 ng/ml
  • CID 316S protein was 9Q hours and was detectable up to 14 days post-dose.
  • CID 3165 protein was safe and well tolerated.
  • C 7n ⁇ following single injection doses was dose-proportional.
  • the mean C ⁇ in the 500 ug cohort was 22 ng/ml, and the mean t ira was 150 hours. Dosing once every 2-4 weeks or more is supported by the pharmacokinetics. Antiviral response against Hepatitis C was observed in the majority of subjects in the single injection cohorts (120-500 ug).
  • IFN-alpha-HSA fusions are used to treat patients with chronic Hepatitis C infection (HCV).
  • Interferon alpha also known as interferon alfa or leukocyte interferon, is the standard of care for treatment of patients infected with HCV.
  • the term "interferon alpha" refers to a family of highly homologous related polypeptides with anti-viral activity.
  • the interferon alpha portion of the IFN-alpha-HSA fusion consists or alternatively comprises any interferon alpha or fragment thereof known in the art.
  • Non-limiting examples of the interferon alpha portion of the IFN-alpha-HSA fusion proteins of the invention include, but are not limited to, the interferon alpha proteins disclosed in the Therapeutic protein column of Table 1.
  • the interferon alpha portion consists or alternatively comprises interferon alpha- 2a, interferon alpha-2b, interferon alpha-2c, consensus interferon, interferon alfacon-1, interferon alpha-nl, interferon alpha-n3, any commercially available form of interferon alpha, such as, for example, INTRON ® A (Schering Corp., Kenilworth, N.J.), ROFERON ® A (Hoffman-La Roche, Nutley, NJ.
  • the interferon alpha portion of the IFN-alpha-HSA fusion protein consists or alternatively comprises interferon alpha modified or formulated for extended or controlled release.
  • the interferon alpha portion consists, or alternatively comprises commercially available extended release or controlled release interferon alpha, including, but not limited to interferon-alpha-XL (Flamel Technologies, France) and LOCTERONTM (BioLex Therapeutics/OctoPlus, Pittsboro, NC).
  • the interferon alpha portion of the IFN-alpha-HSA fusion protein may be modified by the attachment of chemical moieties.
  • Jhe. inteferon .alpha portion may_be modified by pegylation.
  • the interferon alpha portion of the IFN-alpha-HSA fusion protein consists or alternatively comprises pegylated forms of interferon alpha-2a, 2b, or consensus interferon and include, but are not limited to, a commercially available pegylated interferon alpha, such as, for example, PEG-INTRON ® (Schering Corp., Kenilworth, N. J.), PEGASYS ® (Hoffman-La Roche, Nutley, N. J.), PEG-OMNIFERONTM (Viragen, Inc., Plantation, FL) or a fragment thereof.
  • PEG-INTRON ® Schering Corp., Kenilworth, N. J.
  • PEGASYS ® Heoffman-La Roche, Nutley, N. J.
  • PEG-OMNIFERONTM Virtualagen, Inc., Plantation, FL
  • IFN-alpha-HSA "IFN-alpha-HSA" fusions refers to the HSA fused to any of the inter
  • Patients infected with HCV may fall within two categories based on previous exposure to an interferon regimen for treatment of the HCV infection.
  • "Treatment-naive patients” or “na ⁇ ve patients” are those patients who have never been treated with an interferon regimen.
  • "Treatment-experienced patients” or “experienced patients” are those patients who have been treated or are currently being treated with an interferon regimen.
  • “Non-responders” are experienced patients who have been previously treated with an interferon regimen but have failed to meet the primary endpoint of treatment such as an early viral load reduction (EVR) or an end-of-treamient response (ETR).
  • EMR early viral load reduction
  • ETR end-of-treamient response
  • HCV patient refers to a patient who is infected with HCV and who is either na ⁇ ve or experienced.
  • HCV patient who is “experienced” is either a non-responder or a relapser.
  • the Hepatitis C virus can be classified into numerous genotypes, with four genotypes, genotype 1, 2, 3, or 4, being the most prevalent.
  • the Hepatitis C virus that infects an HCV patient comprises a single genotype.
  • the Hepatitis virus can comprise a combination of two or more genotypes.
  • the genotype of Hepatitis C virus may also be a variant of one of the known HCV genotypes.
  • the Hepatitis C virus of the HCV patient is genotype 1 or a variant thereof.
  • HCV refers to the Hepatitis C virus of any genotype, or combination or variants thereof.
  • the standard treatment regimen for patients with HCV involves treatment with interferon alpha in combination with an antiviral agent, such as, ribavirin.
  • an antiviral agent such as, ribavirin.
  • the interferon alpha is administered daily, twice-a-week, or weekly and the ribavirin is administered daily.
  • recent studies have also used inteferon alpha in combination with other antiviral agents known in the art for the treatment of HCV.
  • the IFN-aipha-HSA fusion may be administered to the HCV patient either alone or in combination with an antiviral agent, such as, for example, ribavirin.
  • IFN-alpha-HSA fusion may be administered to the HCV patient in combination with one or more antiviral agents, such as, for example, ribavirin and an additional antiviral agent.
  • the HCV patients are treated with an IFN-alpha-HSA fusion by administration once every 2-4 weeks alone or in combination with an effective amount of an antiviral agent.
  • the HCV patients are treated with an IFN-alpha-HSA fusion by administration once every 2-4 weeks in combination with an effective amount of one or more antiviral agents.
  • the IFN-alpha-HSA fusion is administered to the HCV patient once every 4 weeks.
  • the IFN- alpha-HSA fusion is administered to the HCV patient more than once every 4 weeks.
  • the IFN-alpha-HSA fusion is adminstered once every 4 weeks or more to an HCV patient, wherein the treatment also includes administration of an effective amount of one or more antiviral agents.
  • IFN-alpha-HSA fusions may be used as a low-dose monotherapy for maintenance therapy of HCV.
  • IFN-alpha-HSA fusions may be used in combination with ribavirin and one or more other antiviral agents for the treatement of HCV.
  • IFN-alpha-HSA fusions may be used in combination with one or more antiviral agents, other than ribavirin, for the treatment of HCV.
  • IFN-alpha-HSA fusions may be used for the treatment of other viral infections.
  • IFN-alpha-HSA fusions may be used for the treatment of Hepatitis B (HBV).
  • IFN-alpha-HSA fusions may be used for the treatment of Human Papilloma Virus (HPV).
  • HPV Human Papilloma Virus
  • IFN-alpha-HSA fusions may be used in the treatment of cancer, including, but not limited to hairy cell leukemia, malignant melanoma, follicular lymphoma, chronic myelogenous leukemia, AIDS related Kaposi's Sarcoma, multiple myeloma, or renal cell cancer.
  • HSA fusions with natriuretic peptides may be used for the treatment of cardiovascular disorders.
  • HSA fusions with natriuretic peptides including but not limited to ANP-HSA fusions or BNP-HSA fusions
  • HSA fusions with natriuretic peptides including but not limited to ANP-HSA fusions or BNP-HSA fusions, may be used in the treatement of post-myocardial infarction.
  • HSA fusions with natriuretic peptides may be used to additional cardiovascular disorders, including, but not limited to hypertension, salt-sensitive hypertension, angina pectoris, peripherial artery disease, hypotension, cardiac volume overload, cardiac decompensation, cardiac failure, left ventricular dysfunction, dyspnea, myocardial reperfusion injury, or left ventricular remodeling.
  • HSA fusions with natriuretic peptides may be used in the treatment of renal diseases, including, but not limited to diabetic nephrophathy; glomerular hypertrophy, glomerular injury, renal glomerular disease, actute and/or chronic renal failure.
  • HSA fusions with natriuretic peptides may be used to treat stroke or excess fluid in tissues.
  • HSA may be fused with natriuretic peptide variants including, but not limited to, BNP-HSA fusions wherein the BNP component of the fusion protein is BNP amino acid residues 1-29.
  • the BNP component of the HSA fusion protein consists of two BNP variants (e.g., BNP amino acid residues 1-29) in tandem.
  • the BNP component of the HSA fusion protein consists of three, four, five or more BNP variants (e.g., BNP amino acid residues 1-29) in tandem.
  • HSA fusions with BNP variants may be used for the treatment of congestive heart failure.
  • HSA fusions with BNP variants e.g., BNP amino acid residues 1-29 may be used in the treatment of post-myocardial infarction.
  • HSA fusions with BNP variants may be used to treat additional cardiovascular disorders, including, but not limited to, hypertension, salt-sensitive hypertension, angina pectoris, peripheral artery disease, hypotension, cardiac volume overload, cardiac decompensation, cardiac failure, non-hemodynamic CHF, left ventricular dysfunction, dyspnea, myocardial reperfusion injury, or left ventricular remodeling, hi another embodiment, HSA fusions with BNP variants (e.g., BNP amino acid residues 1-29) may be used in the treatment for elevated aldosterone levels, which can lead to vasoconstriction, impaired cardiac output and/or hypertension, hi a preferred embodiment, HSA fusion with BNP variants (e.g., BNP amino acid residues 1-29) may be used in the treatment of renal disorders or diseases, including, but not limited to, diabetic nephropathy; glomerular hypertrophy, glomerular injury, renal glomerular disease,
  • the invention is directed to natriuretic peptide variants including, but not limited to BNP amino acid ⁇ ⁇ ⁇ ⁇ residues 1-29, wherein the peptides ' are'n ⁇ 't fused with HSA.
  • the BNP variants of the invention have the sequence of two BNP variants (e.g., BNP amino acid residues 1-29) in tandem.
  • the BNP variants of the invention have the sequence of three, four, five or more BNP variants (e.g., BNP amino acid residues 1-29) in tandem.
  • the BNP variants (e.g., BNP amino acid residues 1-29) of the invention may be used for the treatment of congestive heart failure.
  • the BNP variants (e.g., BNP amino acid residues 1-29) of the invention may be used in the treatment of post-myocardial infarction.
  • the BNP variants (e.g., BNP amino acid residues 1-29) of the invention may be used to treat additional cardiovascular disorders, including, but not limited to, hypertension, salt-sensitive hypertension, angina pectoris, peripheral artery disease, hypotension, cardiac volume overload, cardiac decompensation, cardiac failure, non-hemodynamic CHF, left ventricular dysfunction, dyspnea, myocardial reperfusion injury, or left ventricular remodeling, hi another embodiment, the BNP variants (e.g., BNP amino acid residues 1-29) of the invention may be used in the treatment for elevated aldosterone levels, which can lead to vasoconstriction, impaired cardiac output and/or hypertension.
  • the BNP variants (e.g., BNP amino acid residues 1-29) of the invention may be used in the treatment of renal disorders or diseases, including, but not limited to, diabetic nephropathy; glomerular hypertrophy, glomerular injury, renal glomerular disease, acute and/or chronic renal failure.
  • the BNP variants (e.g., BNP amino acid residues 1-29) of the invention may be used to treat stroke or excess fluid in tissues.
  • the invention is directed to natriuretic peptide variants including, but not limited to, BNP variants (e.g., BNP amino acid residues 1-29), that have been modified in order to extend half-life, biological activity, and/or to facilitate purification of the variant.
  • BNP variants e.g., BNP amino acid residues 1-29
  • the natriuretic peptide variants may be pegylated, methylated, or otherwise chemically modified or conjugated using techniques known in the art.
  • natriuretic peptide variants of the invention may be fused or conjugated to an antibody Fc region, or portion thereof.
  • the antibody portion fused to a natriuretic variants (e.g., BNP amino acid residues 1-29) of the invention may comprise the constant region, hinge region, CHl domain, CH2 domain, and CH3 domain or any combination of whole domains or portions thereof.
  • the natriuretic variants may also be fused or conjugated to the above antibody portions to form multimers.
  • Fc portions fused to the polypeptides of the present invention can form dimers through disulfide bonding between the Fc portions.
  • Higher multimeric forms can be made by fusing the variants to portions of IgA and IgM. Methods for fusing or conjugating the variants of the present invention to antibody portions are known in the art. See, e.g., U.S. Patent Nos.
  • the " m ⁇ dified BNP variants of the invention have the sequence of two BNP variants (e.g., BNP amino acid residues 1-29) in tandem.
  • the modified BNP variants of the invention have the sequence of three, four, five or more BNP variants (e.g., BNP amino acid residues 1-29) in tandem, fa a preferred embodiment, the modified BNP variants (e.g., BNP amino acid residues 1-29) of the invention may be used for the treatment of congestive heart failure.
  • the modified BNP variants (e.g., BNP amino acid residues 1-29) of the invention may be used in the treatment of post-myocardial infarction.
  • the modified BNP variants (e.g., BNP amino acid residues 1-29) of the invention may be used to treat additional cardiovascular disorders, including, but not limited to, hypertension, salt-sensitive hypertension, angina pectoris, peripheral artery disease, hypotension, cardiac volume overload, cardiac decompensation, cardiac failure, non-hemodynamic CHF, left ventricular dysfunction, dyspnea, myocardial reperfusion injury, or left ventricular remodeling.
  • the modified BNP variants (e.g., BNP amino acid residues 1-29) of the invention may be used in the treatment for elevated aldosterone levels, which can lead to vasoconstriction, impaired cardiac output and/or hypertension.
  • the modified BNP variants (e.g., BNP amino acid residues 1-29) of the invention may be used in the treatment of renal disorders or diseases, including, but not limited to, diabetic nephropathy; glomerular hypertrophy, glomerular injury, renal glomerular disease, acute and/or chronic renal failure.
  • the modified BNP variants (e.g., BNP amino acid residues 1-29) of the invention may be used to treat stroke or excess fluid in tissues.
  • CNP-HSA fusions may be used in the regulation of endochodral ossification.
  • CNP-HSA fusions may be used in the treatment of skeletal dysplasias, including, but not limited to achondroplasia, hypochondroplasia, and thanatophoric dysplasia.
  • therapeutic activity or “activity” may refer to an activity whose effect is consistent with a desirable therapeutic outcome in humans, or to desired effects in non-human mammals or in other species or organisms.
  • Therapeutic activity may be measured in vivo or in vitro. For example, a desirable effect may be assayed in cell culture.
  • in vitro or cell culture assays are commonly available for many Therapeutic proteins as described in the art. Examples of assays include, but are not limited to those described herein in the Examples section or in the "Exemplary Activity Assay” column (column 3) of Table 1.
  • Therapeutic proteins corresponding to a Therapeutic protein portion of an albumin fusion protein of the invention are often modified by the attachment of one or more oligosaccharide groups.
  • the modification referred to as glycosylation, can dramatically affect the physical properties of proteins and can be important in protein stability, secretion, and localization. Glycosylation occurs at specific locations along the polypeptide backbone.
  • glycosylation characterized by O-Iinked oligosaccharides, which are attached to serine or threonine residues
  • glycosylation characterized by N-linked oligosaccharides which are attached to asparagine residues in an Asn-X-Ser or Asn-X-Thr sequence, where X can be any amino acid except proline.
  • N-acetylneuramic acid also known as sialic acid
  • Variables such as protein structure and cell type influence the number and nature of the carbohydrate units within the chains at different glycosylation sites. Glycosylation isomers are also common at the same site within a given cell type.
  • Therapeutic proteins corresponding to a Therapeutic protein portion of an albumin fusion protein of the invention may be modified so that glycosylation at one or more sites is altered as a result of manipulation(s) of their nucleic acid sequence, by the host cell in which they are expressed, or due to other conditions of their expression.
  • glycosylation isomers may be produced by abolishing or introducing glycosylation sites, e.g., by substitution or deletion of amino acid residues, such as substitution of glutamine for asparagine, or unglycosylated recombinant proteins may be produced by expressing the proteins in host cells that will not glycosylate them, e.g. in E. coli or glycosylation-def ⁇ cient yeast.
  • Therapeutic proteins particularly those disclosed in Table 1, and their nucleic acid and amino acid sequences are well known in the art and available in public databases such as Chemical Abstracts Services Databases (e.g., the CAS Registry), GenBank, and subscription provided databases such as GenSeq (e.g., Derwent).
  • GenSeq e.g., Derwent
  • Exemplary nucleotide sequences of Therapeutic proteins which may be used to derive a polynucleotide of the invention are shown in column 7, "SEQ ID NO:X,” of Table 2.
  • Sequences shown as SEQ ID NO:X may be a wild type polynucleotide sequence encoding a given Therapeutic protein (e.g., either full length or mature), or in some instances the sequence may be a variant of said wild type polynucleotide sequence (e.g., a polynucleotide which encodes the wild type Therapeutic protein, wherein the DNA sequence of said polynucleotide has been optimized, for example, for expression in a particular species; or a polynucleotide encoding a variant of the wild type Therapeutic protein (i.e., a site directed mutant; an allelic variant)).
  • a variant of the wild type Therapeutic protein i.e., a site directed mutant; an allelic variant
  • SEQ ID NO:X it is well within the ability of the skilled artisan to use the sequence shown as SEQ ID NO:X to derive the construct described in the same row. For example, if SEQ ID N0:X corresponds to a full length protein, but only a portion of that protein is used to generate the specific CID, it is within the skill of the art to rely on molecular biology techniques, such as PCR, to amplify the specific fragment and clone it into the appropriate vector.
  • Additional Therapeutic proteins corresponding to a Therapeutic protein portion of an albumin fusion protein of the invention include, but are not limited to, one or more of the Therapeutic proteins or peptides disclosed in the "Therapeutic Protein X" column of Table 1 (column 1), or fragment or.-variant -thereof. - - - - - - - - - - - - - - ⁇ - - - ⁇ - -
  • Table 1 provides a non-exhaustive list of Therapeutic proteins that correspond to a Therapeutic protein portion of an albumin fusion protein of the invention, or an albumin fusion protein encoded by a polynucleotide of the invention.
  • the first column, "Therapeutic Protein X,” discloses Therapeutic protein molecules that may be followed by parentheses containing scientific and brand names of proteins that comprise, or alternatively consist of, that Therapeutic protein molecule or a fragment or variant thereof.
  • “Therapeutic protein X” as used herein may refer either to an individual Therapeutic protein molecule, or to the entire group of Therapeutic proteins associated with a given Therapeutic protein molecule disclosed in this column.
  • the "Biological activity” column (column 2) describes Biological activities associated with the Therapeutic protein molecule.
  • Example 3 provides references that describe assays which may be used to test the therapeutic and/or biological activity of a Therapeutic protein:X or an albumin fusion protein comprising a Therapeutic protein X (or fragment thereof) portion.
  • Each of the references cited in the "Exemplary Activity Assay” column are herein incorporated by reference in their entireties, particularly with respect to the description of the respective activity assay described in the reference (see Methods section therein, for example) for assaying the corresponding biological activity set forth in the "Biological Activity” column of Table 1.
  • the fourth column describes disease, disorders, and/or conditions that may be treated, prevented, diagnosed, and/or ameliorated by Therapeutic protein X or an albumin fusion protein comprising a Therapeutic protein X (or fragment thereof) portion.
  • the "Construct ID” column (column 5 ) provides a link to an exemplary albumin fusion construct disclosed in Table 2 which encodes an albumin fusion protein comprising, or alternatively consisting of the referenced Therapeutic Protein X (or fragment thereof) portion.
  • Therapeutic IO Protein Z inappropriately elevated in selective cation channel. It is mediated by the would be beneficial, as anti-angiogenic or anti-tumor hypertensive diseases and in heart endothelin receptors ETA and ETB, both of agent for treatment of cancer and macular failure. which are G-protein-coupled receptors. degeneration. Antagonists can be identified by their ability to
  • Mechano Growth Factor A muscle growth factor that Myoblast proliferation and differentiation can Wasting disease, cachexia, stroke, MI, CHF, SEQ ID NOs: 783 a (MGF; IGF-IEc; Oenbank appears lost with aging. Can also be assayed in vitro by assays known in the art diseases where neuroprotection or muscle (skeletal 784. Accession No. P05019) act as neuroprotective agent. such as disclosed : in Dluzniewska et al, FASEB or smooth) protection or regeneration would be Unlike mature IGF-I, MGF J. (2005) Sep 6; Goidspink G. J Musculoskelet beneficial. inhibits terminal differentiation Neuronal Interact. (2004) Jun;4(2): 143-7; or whilst increasing myoblast Yang SY and Goldspink G, FEBS Lett. (2002) proliferation. JuI 3, 522(l-3):l56-60.
  • Table 2 provides a non-exhaustive list of polynucleotides of the invention comprising, or alternatively consisting of, nucleic acid molecules encoding an albumin fusion protein.
  • the first column, "Fusion No.” gives a fusion number to each polynucleotide.
  • Column 2, "Construct ID” provides a unique numerical identifier for each polynucleotide of the invention.
  • the Construct IDs may be used to identify polynucleotides which encode albumin fusion proteins comprising, or alternatively consisting of, a Therapeutic protein portion corresponding to a given Therapeutic Protein:X listed in the corresponding row of Table 1 wherein that Construct ID is listed in column 5.
  • the "Construct Name” column (column 3) provides the name of a given albumin fusion construct or polynucleotide.
  • an "expression cassette" comprising, or alternatively consisting of, one or more of (1) a polynucleotide encoding a given albumin fusion protein, (2) a leader sequence, (3) a promoter region, and (4) a transcriptional terminator, may be assembled in a convenient cloning vector and subsequently be moved into an alternative vector, such as, for example, an expression vector including, for example, a yeast expression vector or a mammalian expression vector.
  • an expression cassette comprising, or alternatively consisting of, a nucleic acid molecule encoding an albumin fusion protein is cloned into pSAC3 5 .
  • an expression cassette comprising, or alternatively consisting of, a nucleic acid molecule encoding an albumin fusion protein is cloned into pC4.
  • a polynucleotide comprising or alternatively consisting of a nucleic acid molecule encoding the Therapeutic protein portion of an albumin fusion protein is cloned into pC4:HSA.
  • an expression cassette comprising, or alternatively consisting of, a nucleic acid molecule encoding an albumin fusion protein is cloned into pEE12.
  • Other useful cloning and/or expression vectors will be known to the skilled artisan and are within the scope of the invention.
  • SEQ ID N0:Y provides the full length amino acid sequence of the albumin fusion protein of the invention. Bi most instances, SEQ ID N0:Y shows the unprocessed form of the albumin fusion protein encoded - in other words, SEQ ID NO:Y shows the signal sequence, a HSA portion, and a therapeutic portion all encoded by the particular construct. Specifically contemplated by the present invention are all polynucleotides that encode SEQ ID NO:Y. When these polynucleotides are used to express the encoded protein from a cell, the cell's natural secretion and processing steps produces a protein that lacks the signal sequence listed in columns 4 and/or 11 of Table 2.
  • the seventh column provides the parent nucleic acid sequence from which a polynucleotide encoding a Therapeutic protein portion of a given albumin fusion protein may be derived.
  • the parent nucleic acid sequence from which a polynucleotide encoding a Therapeutic protein portion of an albumin fusion protein may be derived comprises the wild type gene sequence encoding a Therapeutic protein shown in Table 1.
  • the parent nucleic acid sequence from which a polynucleotide encoding a Therapeutic protein portion of an albumin fusion protein may be derived comprises a variant or derivative of a wild type gene sequence encoding a Therapeutic protein shown in Table 1, such as, for example, a synthetic codon optimized variant of a wild type gene sequence encoding a Therapeutic protein.
  • SEQ ID NO:Z provides a predicted translation of the parent nucleic acid sequence (SEQ ID NO:X).
  • This parent sequence can be a full length parent protein used to derive the particular construct, the mature portion of a parent protein, a variant or fragment of a wildtype protein, or an artificial sequence that can be used to create the described construct.
  • One of skill in the art can use this amino acid sequence shown in SEQ ID NO:Z to determine which amino acid residues of an albumin fusion protein encoded by a given construct are provided by the therapeutic protein.
  • SEQ ID NO:Z corresponds to a full length protein, but only a portion of that protein is used to generate the specific CID, it is within the skill of the art to rely on molecular biology techniques, such as PCR, to amplify the specific fragment and clone it into the appropriate vector.
  • Amplification primers provided in columns 9 and 10, "SEQ ID NO:A” and “SEQ ID NO:B” respectively, are exemplary primers used to generate a polynucleotide comprising or alternatively consisting of a nucleic acid molecule encoding the Therapeutic protein portion of a given albumin fusion protein.
  • oligonucleotide primers having the sequences shown in columns 9 and/or 10 (SEQ ID NOS:A and/or B) are used to PCR amplify a polynucleotide encoding the Therapeutic protein portion of an albumin fusion protein using a nucleic acid molecule comprising or alternatively consisting of the nucleotide sequence provided in column 7 (SEQ BD NO:X)of the corresponding row as the template DNA.
  • PCR methods are well-established in the art. Additional useful primer sequences could readily be envisioned and utilized by those of ordinary skill in the art.
  • oligonucleotide primers may be used in overlapping PCR reactions to generate mutations within a template DNA sequence. PCR methods are known in the art.
  • an "expression cassette" comprising, or alternatively consisting of one or more of (1) a polynucleotide encoding a given albumin fusion protein, (2) a leader sequence, (3) a promoter region, and (4) a transcriptional terminator can be moved or "subcloned" from one vector into another.
  • Fragments to be subcloned may be generated by methods well known in the art, such as, for example, PCR amplification (e.g., using oligonucleotide primers having the sequence shown in SEQ ID NO:A or B), and/or restriction enzyme digestion.
  • the albumin fusion proteins of the invention are capable of a therapeutic activity and/or biologic activity corresponding to the therapeutic activity and/or biologic activity of the Therapeutic protein corresponding to the Therapeutic protein portion of the albumin fusion protein listed in the corresponding row of Table 1.
  • the therapeutically active protein portions of the albumin fusion proteins of the invention are fragments or variants of the protein encoded by the sequence shown in SEQ ID NO:X column of Table 2, and are capable of the therapeutic activity and/or biologic activity of the corresponding Therapeutic protein.
  • the present invention is further directed to fragments of the Therapeutic proteins described in Table 1, albumin proteins, and/or albumin fusion proteins of the invention.
  • the present invention is also directed to polynucleotides encoding fragments of the Therapeutic proteins described in Table 1, albumin proteins, and/or albumin fusion proteins of the invention.
  • fragments of a Therapeutic protein corresponding to a Therapeutic protein portion of an albumin fusion protein of the invention include the full length protein as well as polypeptides having one or more residues deleted from the amino terminus of the amino acid sequence of the reference polypeptide (i.e., a Therapeutic protein referred to in Table 1, or a Therapeutic protein portion of an albumin fusion protein encoded by a polynucleotide or albumin fusion construct described in Table 2).
  • N-terminal deletions may be described by the general formula m to q, where q is a whole integer representing the total number of amino acid residues in a reference polypeptide (e.g., a Therapeutic protein referred to in Table 1, or a Therapeutic protein portion of an albumin fusion protein of the invention, or a Therapeutic protein portion of an albumin fusion protein encoded by a polynucleotide or albumin fusion construct described in Table 2), and m is defined as any integer ranging from 2 to q minus 6. Polynucleotides encoding these polypeptides are also encompassed by the invention.
  • a reference polypeptide e.g., a Therapeutic protein referred to in Table 1, or a Therapeutic protein portion of an albumin fusion protein of the invention, or a Therapeutic protein portion of an albumin fusion protein encoded by a polynucleotide or albumin fusion construct described in Table 2
  • m is defined as any integer ranging from 2 to q minus 6.
  • fragments of serum albumin polypeptides corresponding to an albumin protein portion of an albumin fusion protein of the invention include the full length protein as well as polypeptides having one or more residues deleted from the amino terminus of the amino acid sequence of the reference polypeptide (i.e., serum albumin, or a serum albumin portion of an albumin fusion protein encoded by a polynucleotide or albumin fusion construct described in Table 2).
  • N-terminal deletions may be described by the general formula m to 585, where 585 is a whole integer representing the total number of amino acid residues in mature human serum albumin (SEQ ID NO.l), and m is defined as any integer ranging from 2 to 579.
  • N-terminal deletions may be described by the general formula m to 6O9, where ⁇ 5r ⁇ is a whole integer representing the total number of amino acid residues in full length human serum albumin (SEQ ID NO:3), and m is defined as any integer ranging from 2 to 603. Polynucleotides encoding these polypeptides are also encompassed by the invention.
  • fragments of albumin fusion proteins of the invention include the full length albumin fusion protein as well as polypeptides having one or more residues deleted from the amino terminus of the albumin fusion protein (e.g., an albumin fusion protein encoded by a polynucleotide or albumin fusion construct described in Table 2; or an albumin fusion protein having the amino acid sequence disclosed in column 6 of Table 2).
  • N-terminal. deletions may be described by the general formula m to q, where q is a whole integer representing the total number of amino acid residues in the albumin fusion protein, and m is defined as any integer ranging from 2 to q minus 6.
  • Polynucleotides encoding these polypeptides are also encompassed by the invention.
  • a reference polypeptide e.g., a Therapeutic protein; serum albumin protein; or albumin fusion protein of the invention
  • other functional activities e.g., biological activities, ability to multimerize, ability to bind a ligand
  • Therapeutic activities may still be retained.
  • the ability of polypeptides with C-terminal deletions to induce and/or bind to antibodies which recognize the complete or mature forms of the polypeptide generally will be retained when less than the majority of the residues of the complete or mature polypeptide are removed from the C-terminus.
  • Whether a particular polypeptide lacking the N-terminal and/or C-terminal residues of a reference polypeptide retains Therapeutic activity can readily be determined by routine methods described herein and/or otherwise known in the art.
  • the present invention further provides polypeptides having one or more residues deleted from the carboxy terminus of the amino acid sequence of a Therapeutic protein corresponding to a Therapeutic protein portion of an albumin fusion protein of the invention (e.g., a Therapeutic protein referred to in Table 1, or a Therapeutic protein portion of an albumin fusion protein encoded by a polynucleotide or albumin fusion construct described in Table T).
  • a Therapeutic protein corresponding to a Therapeutic protein portion of an albumin fusion protein of the invention e.g., a Therapeutic protein referred to in Table 1, or a Therapeutic protein portion of an albumin fusion protein encoded by a polynucleotide or albumin fusion construct described in Table T.
  • C-terminal deletions may be described by the general formula 1 to n, where n is any whole integer ranging from 6 to q minus 1, and where q is a whole integer representing the total number of amino acid residues in a reference polypeptide (e.g., a Therapeutic protein referred to in Table 1, or a Therapeutic protein portion of an albumin fusionprotein encoded by a polynucleotide-or albumin fusion construct • described in Table 2).
  • a reference polypeptide e.g., a Therapeutic protein referred to in Table 1, or a Therapeutic protein portion of an albumin fusionprotein encoded by a polynucleotide-or albumin fusion construct • described in Table 2.
  • Polynucleotides encoding these polypeptides are also encompassed by the invention.
  • the present invention provides polypeptides having one or more residues deleted from the carboxy terminus of the amino acid sequence of an albumin protein corresponding to an albumin protein portion of an albumin fusion protein of the invention (e.g., serum albumin or an albumin protein portion of an albumin fusion protein encoded by a polynucleotide or albumin fusion construct described in Table 2).
  • C-terminal deletions may be described by the general formula 1 to n, where n is any whole integer ranging from 6 to 584, where 584 is the whole integer representing the total number of amino acid residues in mature human serum albumin (SEQ ID NO:1) minus 1.
  • Polynucleotides encoding these polypeptides are also encompassed by the invention, hi particular, C-terminal deletions may be described by the general formula 1 to n, where n is any whole integer ranging from 6 to 608, where 608 is the whole integer representing the total number of amino acid residues in serum albumin (SEQ ID NO:3) minus 1. Polynucleotides encoding these polypeptides are also encompassed by the invention.
  • the present invention provides polypeptides having one or more residues deleted from the carboxy terminus of an albumin fusion protein of the invention, hi particular, C-terminal deletions may be described by the general formula 1 to n, where n is any whole integer ranging from 6 to q minus 1, and where q is a whole integer representing the total number of amino acid residues in an albumin fusion protein of the invention. Polynucleotides encoding these polypeptides are also encompassed by the invention.
  • any of the above described N- or C-terminal deletions can be combined to produce a N- and C-terminal deleted reference polypeptide.
  • the invention also provides polypeptides having one or more amino acids deleted from both the amino and the carboxyl termini, which may be described generally as having residues m to n of a reference polypeptide (e.g., a Therapeutic protein referred to in Table 1, or a Therapeutic protein portion of an albumin fusion protein of the invention, or a Therapeutic protein portion encoded by a polynucleotide or albumin fusion construct described in Table 2, or serum albumin (e.g., SEQ ID NO:1), or an albumin protein portion of an albumin fusion protein of the invention, or an albumin protein portion encoded by a polynucleotide or albumin fusion construct described in Table 2, or an albumin fusion protein, or an albumin fusion protein encoded by a polynucleotide or albumin fusion construct of the invention) where n and m
  • the present application is also directed to proteins containing polypeptides at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to a reference polypeptide sequence (e.g., a Therapeutic protein referred to in Table 1, or a Therapeutic protein portion of an albumin fusion protein of the invention, or a Therapeutic protein portion encoded by a polynucleotide or albumin fusion construct described in Table 2, or serum albumin (e.g., SEQ ID NO: 1), or an albumin protein portion of an albumin fusion protein of the invention, or an albumin protein portion encoded by a polynucleotide or albumin fusion construct described in Table 2, or an albumin fusion protein, or an albumin fusion protein encoded by a polynucleotide or albumin fusion construct of the invention) set forth herein, or fragments thereof.
  • a reference polypeptide sequence e.g., a Therapeutic protein referred to in Table 1, or a Therapeutic protein portion of an albumin fusion
  • the application is directed to proteins comprising polypeptides at least 80%, 8 5 %, 90%, 95%, 9 6 %, 97%, 98% or 99% identical to reference polypeptides having the amino acid sequence of N- and C-terminal deletions as described above. Polynucleotides encoding these polypeptides are also encompassed by the invention.
  • Preferred polypeptide fragments of the invention are fragments comprising, or alternatively, consisting of, an amino acid sequence that displays a Therapeutic activity and/or functional activity (e.g. biological activity) of the polypeptide sequence of the Therapeutic protein or serum albumin protein of which the amino acid sequence is a fragment.
  • Other preferred polypeptide fragments are biologically active fragments.
  • Biologically active fragments are those exhibiting activity similar, but not necessarily identical, to an activity of the polypeptide of the present invention.
  • the biological activity of the fragments may include an improved desired activity, or a decreased undesirable activity.
  • variant refers to a polynucleotide or nucleic acid differing from a reference nucleic acid or polypeptide, but retaining essential properties thereof. Generally, variants are overall closely similar, and, in many regions, identical to the reference nucleic acid or polypeptide.
  • variant refers to a Therapeutic protein portion of an albumin fusion protein of the invention, albumin portion of an albumin fusion protein of the invention, or albumin fusion protein of the invention differing in sequence from a Therapeutic protein (e.g.
  • variants are overall very similar, and, in many regions, identical to the amino acid sequence of the Therapeutic protein corresponding to a Therapeutic protein portion of an albumin fusion protein, albumin protein corresponding to an albumin protein portion of an albumin fusion protein, and/or albumin fusion protein. Nucleic acids encoding these variants are also encompassed by the invention.
  • the present invention is also directed to proteins which comprise, or alternatively consist of, an amino acid sequence which is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100%, identical to, for example, the amino acid sequence of a Therapeutic protein corresponding to a Therapeutic protein portion of an albumin fusion protein of the invention (e.g., the amino acid sequence of a Therapeutic proteurX disclosed in Table 1; or the amino acid sequence of a Therapeutic protein portion of an albumin fusion protein encoded by a polynucleotide or albumin fusion construct described in Table 1 and 2, or fragments or variants the_reof), albumin.proteins.corresponding to an albumin protein portion of an albumin fusion protein of the invention (e.g., the amino acid sequence of an albumin protein portion of an albumin fusion protein encoded by a polynucleotide or albumin fusion construct described in Table 1 and 2; the amino acid sequence shown in SEQ ID NO: 1; or fragments or variants thereof), and
  • polypeptides encompassed by the invention are polypeptides encoded by polynucleotides which hybridize to the complement of a nucleic acid molecule encoding an albumin fusion protein of the invention under stringent hybridization conditions (e.g., hybridization to filter bound DNA in 6X Sodium chloride/Sodium citrate (SSC) at about 45 degrees Celsius, followed by one or more washes in 0.2X SSC, 0.1% SDS at about 50 - 65 degrees Celsius), under highly stringent conditions (e.g., hybridization to filter bound DNA in 6X sodium chloride/Sodium citrate (SSC) at about 45 degrees Celsius, followed by one or more washes in 0.1X SSC, 0.2% SDS at about 68 degrees Celsius), or under other stringent hybridization conditions which are known to those of skill in the art (see, for example, Ausubel, F.M.
  • stringent hybridization conditions e.g., hybridization to filter bound DNA in 6X Sodium chloride/Sodium citrate (SSC) at about 45 degrees Celsius,
  • a polypeptide having an amino acid sequence at least, for example, 95% "identical" to a query amino acid sequence it is intended that the amino acid sequence of the subject polypeptide is identical to the query sequence except that the subject polypeptide sequence may include up to five amino acid alterations per each 100 amino acids of the query amino acid sequence.
  • the amino acid sequence of the subject polypeptide may include up to five amino acid alterations per each 100 amino acids of the query amino acid sequence.
  • up to 5% of the amino acid residues in the subject sequence may be inserted, deleted, or substituted with another amino acid.
  • These alterations of the reference sequence may occur at the amino- or carboxy-terminal positions of the reference amino acid sequence or anywhere between those terminal positions, interspersed either individually among residues in the reference sequence or in one or more contiguous groups within the reference sequence.
  • any particular polypeptide is at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to, for instance, the amino acid sequence of an albumin fusion protein of the invention or a fragment thereof (such as a Therapeutic protein portion of the albumin fusion protein or an albumin portion of the albumin fusion protein), can be determined conventionally using known computer programs.
  • a preferred method for determining the best overall match between a query sequence (a sequence of the present invention) and a subject sequence also referred to as a global sequence alignment, can be determined using the FASTDB computer program based on the algorithm of Brutlag et al.
  • the percent identity is corrected by calculating the number of residues of the query sequence that are N- and C-terminal of the subject sequence, which are not matched/aligned with a corresponding subject residue, as a percent of the total bases of the query sequence. Whether a residue is matched/aligned is determined by results of the FASTDB sequence alignment.
  • This percentage is then subtracted from the percent identity, calculated by the above FASTDB program using the specified parameters, to arrive at a final percent identity score.
  • This final percent identity score is what is used for the purposes of the present invention. Only residues to the N- and C-termini of the subject sequence, which are not matched/aligned with the query sequence, are considered for the purposes of manually adjusting the percent identity score. That is, only query residue positions outside the farthest N- and C- terminal residues of the subject sequence.
  • a 90 amino acid residue subject sequence is aligned with a 1OQ residue query sequence to determine percent identity.
  • the deletion occurs at the N-terminus of the subject sequence and therefore, the FASTDB alignment does not show a matching/alignment of the first 10 residues at the N-terminus.
  • the 10 unpaired residues represent 10% of the sequence (number of residues at the N- and C- termini not matched/total number of residues in the query sequence) so 10% is subtracted from the percent identity score calculated by the FASTDB program. If the remaining 90 residues were perfectly matched the final percent identity would be 90%.
  • a 90 residue subject sequence is compared with a 100 residue query sequence.
  • deletions are internal deletions so there are no residues at the N- or C-termini of the subject sequence which are not matched/aligned with the query.
  • percent identity calculated by FASTDB is not manually corrected.
  • residue positions outside the N- and C-terminal ends of the subject sequence, as displayed in the FASTDB alignment, which are not matched/aligned with the query sequence are manually corrected for. No other manual corrections are to made for the purposes of the present invention.
  • the variant will usually have at least 75% (preferably at least about 80%, 90%, 95% or 99%) sequence identity with a length of normal HA or Therapeutic protein which is the same length as the variant.
  • Homology or identity at the nucleotide or amino acid sequence level is determined by BLAST (Basic Local Alignment Search Tool) analysis using the algorithm employed by the programs blastp, -blastn, blastx, tblastn and tblastx (Karlin et al, Proc. Natl. Acad. Sci. USA 87: 2264-2268 (1990) and Altschul, J. MoI. Evol. 36: 290-300 (1993), fully incorporated by reference) which are tailored for sequence similarity searching.
  • BLAST Basic Local Alignment Search Tool
  • the approach used by the BLAST program is to first consider similar segments between a query sequence and a database sequence, then to evaluate the statistical significance of all matches that are identified and finally to summarize only those matches which satisfy a preselected threshold of significance.
  • the search parameters for histogram, descriptions, alignments, expect i.e., the statistical significance threshold for reporting matches against database sequences
  • cutoff, matrix and filter are at the default settings.
  • the default scoring matrix used by blastp, blastx, tblastn, and tblastx is the BLOSUM62 matrix (Henikoff et al, Proc.
  • the scoring matrix is set by the ratios of M (i.e., the reward score for a pair of matching residues) to N (i.e., the penalty score for mismatching residues), wherein the default values for M and N are 5 and -4, respectively.
  • M i.e., the reward score for a pair of matching residues
  • N i.e., the penalty score for mismatching residues
  • Nucleotide variants produced by silent substitutions due to the degeneracy of the genetic code are preferred.
  • polypeptide variants in which less than 50, less than 40, less than 30, less than 20, less than 10, or 5-50, 5-25, 5-10, 1-5, or 1-2 amino acids are substituted, deleted, or added in any combination are also preferred.
  • Polynucleotide variants can be produced for a variety of reasons, e.g., to optimize codon expression for a particular host (change codons in the human mRNA to those preferred by a bacterial host, such as, yeast or E. coli).
  • a polynucleotide of the invention which encodes the albumin portion of an albumin fusion protein is optimized for expression in yeasFor mammalian cells.
  • Therapeutic protein portion of an albumin fusion protein is optimized for expression in yeast or mammalian cells.
  • a polynucleotide encoding an albumin fusion protein of the invention is optimized for expression in yeast or mammalian cells.
  • a codon optimized polynucleotide which encodes a Therapeutic protein portion of an albumin fusion protein does not hybridize to the wild type polynucleotide encoding the Therapeutic protein under stringent hybridization conditions as described herein.
  • a codon optimized polynucleotide which encodes an albumin portion of an albumin fusion protein does not hybridize to the wild type polynucleotide encoding the albumin protein under stringent hybridization conditions as described herein.
  • a codon optimized polynucleotide which encodes an albumin fusion protein does not hybridize to the wild type polynucleotide encoding the Therapeutic protein portion or the albumin protein portion under stringent hybridization conditions as described herein.
  • a polynucleotide which encodes a Therapeutic protein portion of an albumin fusion protein does not comprise, or alternatively consist of, the naturally occurring sequence of that Therapeutic protein.
  • a polynucleotide which encodes an albumin protein portion of an albumin fusion protein does not comprise, or alternatively consist of, the naturally occurring sequence of albumin protein.
  • a polynucleotide which encodes an albumin fusion protein does not comprise, or alternatively consist of, the naturally occurring sequence of a Therapeutic protein portion or the albumin protein portion.
  • Naturally occurring variants are called "allelic variants," and refer to one of several alternate forms of a gene occupying a given locus on a chromosome of an organism. (Genes ⁇ , Lewin, B., ed., John Wiley & Sons, New York (1985)). These allelic variants can vary at either the polynucleotide and/or polypeptide level and are included in the present invention. Alternatively, non-naturally occurring variants may be produced by mutagenesis techniques or by direct synthesis.
  • variants may be generated to improve or alter the characteristics of the polypeptides of the present invention.
  • one or more amino acids can be deleted from the N-terminus or C- terminus of the polypeptide of the present invention without substantial loss of biological function.
  • Ron et al. J. Biol. Chem. 2 ⁇ 8: 2984-2988 (1993)
  • variant KGF proteins having heparin binding activity even after deleting 3, 8, or 27 amino-terminal amino acid residues.
  • Interferon gamma exhibited up to ten times higher activity after deleting 8-10 amino acid residues from the carboxy terminus of this protein.
  • the invention further includes polypeptide variants which have a functional activity (e.g., biological activity and/or therapeutic activity).
  • the invention provides variants of albumin fusion proteins that have a functional activity (e.g., biological activity and/or therapeutic activity) that corresponds to one or more biological and/or therapeutic activities of the Therapeutic protein corresponding to the Therapeutic protein portion of the albumin fusion protein.
  • the invention provides variants of albumin fusion proteins that have a functional activity (e.g., biological activity and/or therapeutic activity) that corresponds to one or more biological and/or therapeutic activities of the Therapeutic protein corresponding to the Therapeutic protein portion of the albumin fusion protein.
  • Such variants include deletions, insertions, inversions, repeats, and substitutions selected according to general rules known in the art so as have little effect on activity. Polynucleotides encoding such variants are also encompassed by the invention.
  • the variants of the invention have conservative substitutions.
  • conservative substitutions is intended swaps within groups such as replacement of the aliphatic or hydrophobic amino acids Ala, VaI, Leu and JJe; replacement of the hydroxyl residues Ser and Thr; replacement of the acidic residues Asp and GIu; replacement of the amide residues Asn and GIn, replacement of the basic residues Lys, Arg, and His; replacement of the aromatic residues Phe, Tyr, and Trp, and replacement of the small-sized amino acids Ala, Ser, Thr, Met, and GIy.
  • the first strategy exploits the tolerance of amino acid substitutions by natural selection during the process of evolution. By comparing amino acid sequences in different species, conserved amino acids can be identified. These conserved amino acids are likely important for protein function. In contrast, the amino acid positions where substitutions have been tolerated by natural selection indicates that these positions are not critical for protein function. Thus, positions tolerating amino acid substitution could be modified while still maintaining biological activity of the protein.
  • the second strategy uses genetic engineering to introduce amino acid changes at specific positions of a cloned gene to identify regions critical for protein function. For example, site directed mutagenesis or alanine-scanning mutagenesis (introduction of single alanine mutations at every residue in the molecule) can be used. See Cunningham and Wells, Science 244:1O81-1Q8 5 (1989). The resulting mutant molecules can then be tested for biological activity.
  • tolerated conservative amino acid substitutions involve replacement of the aliphatic or hydrophobic amino acids Ala, VaI, Leu and Ue; replacement of the hydroxyl residues Ser and Thr; replacement of the acidic residues Asp and GIu; replacement of the amide residues Asn and GIn, replacement of the basic residues Lys, Arg, and His; replacement of the aromatic residues Phe, Tyr, and Trp, and replacement of the small-sized amino acids Ala, Ser, Thr, Met, and GIy.
  • variants of the present invention include (i) polypeptides containing substitutions of one or more of the non-conserved amino acid residues, where the substituted amino acid residues may or may not be one encoded by the genetic code, or (ii) polypeptides containing substitutions of one or more of the amino acid residues having a substiruent group, or (iii) polypeptides which have been fused with or chemically conjugated to another compound, such as a compound to increase the stability and/or solubility of the polypeptide (for example, polyethylene glycol), (iv) polypeptide containing additional amino acids, such as, for example, an IgG Fc fusion region peptide.
  • polypeptides are deemed to be within the scope of those skilled in the art from the teachings herein.
  • polypeptide variants containing amino acid substitutions of charged amino acids with other charged or neutral amino acids may produce proteins with improved characteristics, such as less aggregation. Aggregation of pharmaceutical formulations both reduces activity and increases clearance due to the aggregate's immunogenic activity. See Pinckard et al., Clin. Exp. Immunol. 2:331-340 (1967); Robbins et al., Diabetes 36: 838-845 (1987); Cleland et al., Crit. Rev. Therapeutic Drug Carrier Systems 10:307-377 (1993).
  • the polypeptides of the invention comprise, or alternatively, consist of, fragments or variants of the amino acid sequence of an albumin fusion protein, the amino acid sequence of a Therapeutic protein and/or human serum albumin, wherein the fragments or variants have 1-5, 5-10, 5-25, 5-50, 10-50 or 50-150, amino acid residue additions, substitutions, and/or deletions when compared to the reference amino acid sequence.
  • _the amino acid substitutions- are conservative.
  • -Nucleic acids encoding these polypeptides are also encompassed by the invention.
  • the polypeptide of the present invention can be composed of amino acids joined to each other by peptide bonds or modified peptide bonds, i.e., peptide isosteres, and may contain amino acids other than the 20 gene-encoded amino acids.
  • the polypeptides may be modified by either natural processes, such as post-translational processing, or by chemical modification techniques which are well known in the art. Such modifications are well described in basic texts and in more detailed monographs, as well as in a voluminous research literature. Modifications can occur anywhere in a polypeptide, including the peptide backbone, the amino acid side-chains and the amino or carboxyl termini.
  • polypeptides may be branched, for example, as a result of ubiquitination, and they may be cyclic, with or without branching. Cyclic, branched, and branched cyclic polypeptides may result from posttranslation natural processes or may be made by synthetic methods.
  • Modifications include acetylation, acylation, ADP-ribosylation, amidation, covalent attachment of flavin, covalent attachment of a heme moiety, covalent attachment of a nucleotide or nucleotide derivative, covalent attachment of a lipid or lipid derivative, covalent attachment of phosphotidylinositol, cross-linking, cyclization, disulfide bond formation, demethylation, formation of covalent cross-links, formation of cysteine, formation of pyroglutamate, formylation, gamma-carboxylation, glycosylation, GPI anchor formation, hydroxylation, iodination, methylation, myristylation, oxidation, pegylation, proteolytic processing, phosphorylation, prenylation, racemization, selenoylation, sulfation, transfer-RNA mediated addition of amino acids to proteins such as arginylation, and ubiquitination.
  • a polypeptide having functional activity refers to a polypeptide capable of displaying one or more known functional activities associated with the full-length, pro-protein, and/or mature form of a Therapeutic protein.
  • Such functional activities include, but are not limited to, biological activity, antigenicity [ability to bind (or compete with a polypeptide for binding) to an anti-polypeptide antibody], immunogenicity (ability to generate antibody which binds to a specific polypeptide of the invention), ability to form multimers with polypeptides of the invention, and ability to bind to a receptor or ligand for a polypeptide.
  • a polypeptide having biological activity refers to a polypeptide exhibiting activity similar to, but not necessarily identical to, an activity of a Therapeutic protein of the present invention, including mature forms, as measured in a particular biological assay, with or without dose dependency. In the case where dose dependency does exist, it need not be identical to that of the polypeptide, but rather substantially similar to the dose-dependence in a given activity as compared to the polypeptide of the present invention (i.e., the candidate polypeptide will exhibit greater activity or not more than about 2 5 -fold less and, preferably, not more than about tenfold less activity, and most preferably, not more than about three-fold less activity relative to the polypeptide of the present invention).
  • an albumin fusion protein of the invention has at least one biological and/or therapeutic activity associated with the Therapeutic protein portion (or fragment or variant thereof) when it is not fused to albumin.
  • the albumin fusion protein of the invention has an increased plasma stability compared to the Therapeutic protein portion (or fragment or variant thereof) in an unfused state.
  • Plasma stability of the albumin fusion protein of the invention or of the unfused Therapeutic protein portion (or fragment or variant thereof) can be assayed using or routinely modifying assays known in the art.
  • the albumin fusion proteins of the invention can be assayed for functional activity (e.g., biological activity) using or routinely modifying assays known in the art, as well as assays described herein.
  • one of skill in the art may routinely assay fragments of a Therapeutic protein corresponding to a Therapeutic protein portion of an albumin fusion protein, for activity using assays referenced in its corresponding row of Table 1 (e.g., in column 3 of Table 1). Further, one of skill in the art may routinely assay fragments of an albumin protein corresponding to an albumin protein portion of an albumin fusion protein, for activity using assays known in the art and/or as described in the Examples section below.
  • various immunoassays known in the art can be used, including but not limited to, competitive and non-competitive assay systems using techniques such as radioimmunoassays, ELISA (enzyme linked immunosorbent assay), "sandwich” immunoassays, immunoradiometric assays, gel diffusion precipitation reactions, immunodiffusion assays, in situ immunoassays (using colloidal gold, enzyme or radioisotope labels, for example), western blots, precipitation reactions, agglutination assays (e.g., gel agglutination assays, hemagglutination assays), complement fixation assays, immunofluorescence assays, protein A assays, and Immunoelectrophoresis assays,
  • antibody binding is detected by detecting a label on the primary antibody.
  • the primary antibody is detected by detecting binding of a secondary antibody or reagent to the primary antibody.
  • the secondary antibody is labeled. Many means are known in the art for detecting binding in an immunoassay and are within the scope of the present invention.
  • a binding partner.(e.g.,.a receptor or -a ligand) -of a- Therapeutic protein is identified
  • an albumin fusion protein which comprises that Therapeutic protein as the Therapeutic protein portion of the fusion
  • an albumin fusion protein which comprises that Therapeutic protein as the Therapeutic protein portion of the fusion
  • the ability of physiological correlates of an albumin fusion protein to bind to a substrate(s) of the Therapeutic polypeptide corresponding to the Therapeutic protein portion of the fusion can be routinely assayed using techniques known in the art.
  • association with other components of the multimer can be assayed, e.g., by means well-known in the art, such as, for example, reducing and non-reducing gel chromatography, protein affinity chromatography, and affinity blotting. See generally, Phizicky et al., supra.
  • an albumin fusion protein comprising all or a portion of an antibody that binds a Therapeutic protein, has at least one biological and/or therapeutic activity (e.g., to specifically bind a polypeptide or epitope) associated with the antibody that binds a Therapeutic protein (or fragment or variant thereof) when it is not fused to albumin.
  • the biological activity and/or therapeutic activity of an albumin fusion protein comprising all or a portion of an antibody that binds a Therapeutic protein is the inhibition (i.e., antagonism) or activation (i.e., agonism) of one or more of the biological activities and/or therapeutic activities associated with the polypeptide that is specifically bound by antibody that binds a Therapeutic protein.
  • Albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein may be characterized in a variety of ways.
  • albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein may be assayed for the ability to specifically bind to the same antigens specifically bound by the antibody that binds a Therapeutic protein corresponding to the Therapeutic protein portion of the albumin fusion protein using techniques described herein or routinely modifying techniques known in the art.
  • albumin fusion proteins e.g., comprising at least a fragment or variant of an antibody that binds a Therapeutic protein
  • Assays for the ability of the albumin fusion proteins to (specifically) bind a specific protein or epitope may be performed in solution (e.g., Houghten, Bio/Techniques 13:412-421(1992)), on beads (e.g., Lam, Nature 354:82-84 (1991)), on chips (e.g., Fodor, Nature 364:555-556 (1993)), on bacteria (e.g., U.S. v. disturb ⁇ ' ⁇ ue '. " .Ii IU* ii...ii .•' «».,.
  • Albumin fusion proteins comprising at least a fragment or variant of a Therapeutic antibody may also be assayed for their specificity and affinity for a specific protein or epitope using or routinely modifying techniques described herein or otherwise known in the art.
  • the albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein may be assayed for cross-reactivity with other antigens (e.g., molecules that have sequence/structure conservation with the molecule(s) specifically bound by the antibody that binds a Therapeutic protein (or fragment or variant thereof) corresponding to the Therapeutic protein portion of the albumin fusion protein of the invention) by any method known in the art.
  • other antigens e.g., molecules that have sequence/structure conservation with the molecule(s) specifically bound by the antibody that binds a Therapeutic protein (or fragment or variant thereof) corresponding to the Therapeutic protein portion of the albumin fusion protein of the invention
  • Immunoassays which can be used to analyze (immunospecific) binding and cross-reactivity include, but are not limited to, competitive and non-competitive assay systems using techniques such as western blots, radioimmunoassays, ELISA (enzyme linked immunosorbent assay), "sandwich” immunoassays, immunoprecipitation assays, precipitin reactions, gel diffusion precipitin reactions, immunodiffusion assays, agglutination assays, complement-fixation assays, immunoradiometric assays, fluorescent immunoassays, and protein A immunoassays, to name but a few.
  • competitive and non-competitive assay systems using techniques such as western blots, radioimmunoassays, ELISA (enzyme linked immunosorbent assay), "sandwich” immunoassays, immunoprecipitation assays, precipitin reactions, gel diffusion precipitin reactions, immunodiffusion assays, aggluti
  • Immunoprecipitation protocols generally comprise lysing a population of cells in a lysis buffer such as RIPA buffer (1% NP-40 or Triton X-100, 1% sodium deoxycholate, 0.1% SDS, 0.15 M NaCl, 0.01 M sodium phosphate at pH 7.2, 1% Trasylol) supplemented with protein phosphatase and/or protease inhibitors (e.g., EDTA, PMSF, aprotinin, sodium vanadate), adding the albumin fusion protein of the invention (e.g., comprising at least a fragment or variant of an antibody that binds a Therapeutic protein) to the cell lysate, incubating for a period of time (e.g., 1 to 4 hours) at 40 degrees C, adding sepharose beads coupled to an anti-albumin antibody, for example, to the cell lysate, incubating for about an hour or more at 40 degrees C, washing the beads in lysis buffer and resuspending the
  • albumin fusion protein to immunoprecipitate a particular antigen can be assessed by, e.g., western blot analysis.
  • One of skill in the art would be knowledgeable as to the parameters that can be modified to increase the binding of the albumin fusion protein to an antigen and decrease the background (e.g., pre- clearing the cell lysate with sepharose beads).
  • immunoprecipitation protocols see, e.g., Ausubel et al, eds, 1994, Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York at 10.16.1.
  • Western blot analysis generally comprises preparing protein samples, electrophoresis of the protein samples in a polyacrylamide gel (e.g., 8%- 20% SDS-PAGE depending on the molecular weight of the antigen), transferring the protein sample from the polyacrylamide gel to a membrane such as nitrocellulose, PVDF or nylon,-blocking the membrane in blocking solution (e.g:, PBS -with 3% BSA or non-fat milk), washing "the membrane in washing buffer (e.g., PBS-Tween 20), applying the albumin fusion protein of the invention (diluted in blocking buffer) to the membrane, washing the membrane in washing buffer, applying a secondary antibody (which recognizes the albumin fusion protein, e.g., an anti- human serum albumin antibody) conjugated to an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase) or radioactive molecule (e.g., 32 P or 125 I) diluted in blocking buffer, washing the membrane in wash
  • ELISAs comprise preparing antigen, coating the well of a 96-well microtiter plate with the antigen, washing away antigen that did not bind the wells, adding the albumin fusion protein (e.g., comprising at least a fragment or variant of an antibody that binds a Therapeutic protein) of the invention conjugated to a detectable compound such as an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase) to the wells and incubating for a period of time, washing away unbound or non-specifically bound albumin fusion proteins, and detecting the presence of the albumin fusion proteins specifically bound to the antigen coating the well.
  • an enzymatic substrate e.g., horseradish peroxidase or alkaline phosphatase
  • the albumin fusion protein does not have to be conjugated to a detectable compound; instead, a second antibody (which recognizes albumin fusion protein) conjugated to a detectable compound may be added to the well.
  • the albumin fusion protein may be coated to the well, hi this case, the detectable molecule could be the antigen conjugated to a detectable compound such as an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase).
  • an enzymatic substrate e.g., horseradish peroxidase or alkaline phosphatase.
  • the binding affinity of an albumin fusion protein to a protein, antigen, or epitope and the off-rate of an albumin fusion protein- protein/antigen/epitope interaction can be determined by competitive binding assays.
  • a competitive binding assay is a radioimmunoassay comprising the incubation of labeled antigen (e.g., 3 H or 125 I) with the albumin fusion protein of the invention in the presence of increasing amounts of unlabeled antigen, and the detection of the antibody bound to the labeled antigen.
  • labeled antigen e.g., 3 H or 125 I
  • the affinity of the albumin fusion protein br a specific protein, antigen, or epitope and the binding off-rates can be determined from the data by Scatchard plot analysis.
  • Competition with a second protein that binds the same protein, antigen or epitope as the albumin fusion protein can also be determined using radioimmunoassays.
  • the protein, antigen or epitope is incubated with an albumin fusion protein conjugated to a labeled compound (e.g., 3 H or 125 I) in the presence of increasing amounts of an unlabeled second protein that binds the same protein, antigen, or epitope as the albumin fusion protein of the invention.
  • a labeled compound e.g., 3 H or 125 I
  • BIAcore kinetic analysis is used to determine the binding on and off rates of albumin fusion proteins of the invention to a protein, antigen or epitope.
  • BIAcore kinetic analysis comprises analyzing the binding and dissociation of albumin fusion proteins, or specific polypeptides, antigens or epitopes from chips with immobilized specific polypeptides, antigens or epitopes or albumin fusion proteins, respectively, on their surface.
  • Antibodies that bind a Therapeutic protein corresponding to the Therapeutic protein portion of an albumin fusion protein may also be described or specified in terms of their binding affinity for a given protein or antigen, preferably the antigen which they specifically bind.
  • Preferred binding affinities include those with a dissociation constant or Kd less than 5 X 10 '2 M, I 0 "2 M, S X I 0 "3 M, 10 '3 M, 5 X IO 4 M, 10 "4 M.
  • More preferred binding affinities include those with a dissociation constant or Kd less than 5 X I 0 "5 M, 10 "5 M, 5 X 10 " ⁇ M, 10 "6 M, 5 X 10 "7 M, 10 7 M, 5 X lO "8 M or 1O 'S M.
  • binding affinities include those with a dissociation constant or Kd less than 5 X 10 "9 M, 10 "9 M, 5 X 10 '10 M, 10 " '° M, 5 X Kr 11 M, 10 " " M, 5 X I 0 ' 12 M, 10'12 M, 5 X I 0 "13 M, 10 "13 M, 5 X 10 "14 M, 10 'M M, 5 X 10 "15 M, or 10 "15 M.
  • albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein, has an affinity for a given protein or epitope similar to that of the corresponding antibody (not fused to albumin) that binds a Therapeutic protein, taking into account the valency of the albumin fusion protein (comprising at least a fragment or variant of an antibody that binds a Therapeutic protein) and the valency of the corresponding antibody.
  • albumin fusion proteins and fragments, variants and derivatives thereof may routinely be applied to measure the ability of albumin fusion proteins and fragments, variants and derivatives thereof to elicit biological activity and/or Therapeutic activity (either in vitro or in vivo) related to either the Therapeutic protein portion and/or albumin portion of the albumin fusion protein.
  • Therapeutic activity either in vitro or in vivo
  • Other methods will be known to the skilled artisan and are within the scope of the invention.
  • an albumin fusion protein of the invention comprises at least a fragment or variant of a Therapeutic protein and at least a fragment or variant of human serum albumin, which are associated with one another, preferably by genetic fusion.
  • An additional embodiment comprises at least a fragment or variant of a Therapeutic protein and at least a fragment or variant of human serum albumin, which are linked to one another by chemical conjugation.
  • HSA human serum albumin
  • HA human albumin
  • albumin and HA are broader, and encompass human serum albumin (and fragments and- variants- thereof) as well as albumin from other species " (and fragments and variants thereof).
  • albumin refers collectively to albumin protein or amino acid sequence, or an albumin fragment or variant, having one or more functional activities (e.g., biological activities) of albumin.
  • albumin refers to human albumin or fragments thereof (see for example, EP 201 239, EP 322 094 WO 97/2444 5 , WO95/23857) especially the mature form of human albumin as shown in Figure 1 and SEQ ID NO: 1, or albumin from other vertebrates or fragments thereof, or analogs or variants of these molecules or fragments thereof.
  • the human serum albumin protein used in the albumin fusion proteins of the invention contains one or both of the following sets of point mutations with reference to SEQ ID NO: 1: Leu-407 to Ala, Leu-408 to VaI, Val-409 to Ala, and Arg-410 to Ala; or Arg- 410 to A, Lys-413 to GIn, and Lys-414 to GIn (see, e.g., International Publication No. WO95/23857, hereby incorporated in its entirety by reference herein).
  • albumin fusion proteins of the invention that contain one or both of above-described sets of point mutations have improved stability/resistance to yeast Ya ⁇ 3p proteolytic cleavage, allowing increased production of recombinant albumin fusion proteins expressed in yeast host cells.
  • a portion of albumin sufficient to prolong the therapeutic activity or plasma stability or shelf-life of the Therapeutic protein refers to a portion of albumin sufficient in length or structure to stabilize or prolong the therapeutic activity or plasma stability of the protein so that the shelf life or plasma stability of the Therapeutic protein portion of the albumin fusion protein is prolonged or extended compared to the shelf-life or plasma stability in the non-fusion state.
  • the albumin portion of the albumin fusion proteins may comprise the full length of the HA sequence as described above, or may include one or more fragments thereof that are capable of stabilizing or prolonging the therapeutic activity.
  • Such fragments may be of 10 or more amino acids in length or may include about 15, 20, 25, 30, 50, or more contiguous amino acids from the HA sequence or may include part or all of specific domains of HA. For instance, one or more fragments of HA spanning the first two immunoglobulin- like domains may be used. In a preferred embodiment, the HA fragment is the mature form of HA.
  • the albumin portion of the albumin fusion proteins of the invention may be a variant of normal HA.
  • the Therapeutic protein portion of the albumin fusion proteins of the invention may also be variants of the Therapeutic proteins as described herein.
  • variants includes insertions, deletions and substitutions, either conservative or non conservative, where such changes do not substantially alter one or more of the oncotic, usefiil ligand-binding and non-immunogenic properties of albumin, or the active site, or active domain which confers the therapeutic activities of the Therapeutic proteins.
  • the albumin fusion proteins of the invention may include naturally occurring polymorphic variants of human albumin and fragments of human albumin, for example those fragments disclosed in EP 322 094 (namely HA (Pn), where n is 369 to 419).
  • the albumin may be derived from any vertebrate, especially any mammal, for example human, cow, sheep, or pig.
  • Non-mammalian albumins include, but are not limited to, hen and salmon.
  • the albumin portion of the albumin fusion protein may be from a different animal than the Therapeutic protein portion.
  • an HA fragment or variant will be at least 100 amino acids long, preferably at least 150 amino acids long.
  • the HA variant may consist of or alternatively comprise at least one whole domain of HA, for example domains 1 (amino acids 1-194 of SEQ ID NO: 1), domain 2 (amino acids 195-387 of SEQ ID N ⁇ :l), domain 3 (amino acids 388-585 of SEQ ID NO:1), domains 1 and 2 (1-387 of SEQ ID NO:1), domains 2 and 3 (195-585 of SEQ ID N ⁇ :l) or domains 1 and 3 (amino acids 1-194 of SEQ ID NO:1 and amino acids 388-585 of SEQ ID N ⁇ :l).
  • domains 1 amino acids 1-194 of SEQ ID NO: 1
  • domain 2 amino acids 195-387 of SEQ ID N ⁇ :l
  • domain 3 amino acids 388-585 of SEQ ID NO:1
  • domains 1 and 2 1-387 of SEQ ID NO:1
  • domains 2 and 3 195-585 of SEQ ID N ⁇ :l
  • domains 1 and 3 amino acids 1-194 of SEQ
  • Each domain is itself made up of two homologous subdomains namely 1-105, 120-194, 195-291, 316-387, 388-491 and 512-585, with flexible inter-subdomain linker regions comprising residues LyslO6 to Glull9, Glu292 to Val315 and Glu492 to Ala511.
  • the albumin portion of an albumin fusion protein of the invention comprises at least one subdomain or domain of HA or conservative modifications thereof. If the fusion is based on subdomains, some or all of the adjacent linker is preferably used to link to the Therapeutic protein moiety.
  • Antibodies that Specifically bind Therapeutic proteins are also Therapeutic proteins
  • the present invention also encompasses albumin fusion proteins that comprise at least a fragment or variant of an antibody that specifically binds a Therapeutic protein disclosed in Table 1. It is specifically contemplated that the term "Therapeutic protein” encompasses antibodies that bind a Therapeutic protein (e.g., as Described in column I of Table 1) and fragments and variants thereof. Thus an albumin fusion protein of the invention may contain at least a fragment or variant of a Therapeutic protein, and/or at least a fragment or variant of an antibody that binds a Therapeutic protein.
  • the basic antibody structural unit is known to comprise a tetramer.
  • Each tetramer is composed of two identical pairs of polypeptide chains, each pair having one "light” (about 25 kDa) and one "heavy" chain (about 50-70 kDa).
  • the amino-terminal portion of each chain includes a variable region of about 100 to 110 or more amino acids primarily responsible for antigen recognition.
  • the carboxy-terminal portion of each chain defines a constant region primarily responsible for effector function.
  • Human light chains are classified as kappa and lambda light chains.
  • Heavy chains are classified as mu, delta, gamma, alpha, or epsilon, and define the antibody's isorype as IgM, IgD, IgG, IgA, and IgE, respectively. See generally, Fundamental Immunology Chapters 3-5 (Paul, W., ed., 4th ed. Raven Press, N. Y. (1998)) (incorporated by reference in its entirety for all purposes).
  • the variable regions of each light/heavy chain pair fo ⁇ ji.the antibody binding site. - - - ⁇ -
  • an intact IgG antibody has two binding sites. Except in bifunctional or bispecific antibodies, the two binding sites are the same.
  • the chains all exhibit the same general structure of relatively conserved framework regions (FR) joined by three hypervariable regions, also called complementarity determining regions or CDRs.
  • the CDR regions in general, are the portions of the antibody which make contact with the antigen and determine its specificity.
  • the CDRs from the heavy and the light chains of each pair are aligned by the framework regions, enabling binding to a specific epitope. From N-terminal to C-terminal, both light and heavy chains variable regions comprise the domains FRl, CDRl, FR2, CDR2, FR3, CDR3 and FR4.
  • variable regions are connected to the heavy or light chain constant region.
  • the assignment of amino acids to each domain is in accordance with the definitions of Kabat Sequences of Proteins of Immunological Interest (National Institutes of Health, Bethesda, Md. (1987 and 1991)), or Chothia & Lesk J MoI. Biol. 196:901-917 (1987); Chothia et al. Nature 342:878-883 (1989).
  • antibody refers to immunoglobulin molecules and immunologically active portions of immunoglobulin molecules, i.e., molecules that contain an antigen binding site that specifically binds an antigen (e.g., a molecule containing one or more CDR regions of an antibody).
  • Antibodies that may correspond to a Therapeutic protein portion of an albumin fusion protein include, but are not limited to, monoclonal, multispecific, human, humanized or chimeric antibodies, single chain antibodies (e.g., single chain Fvs), Fab fragments, F(ab') fragments, fragments produced by a Fab expression library, anti-idiotypic (anti-Id) antibodies (including, e.g., anti-Id antibodies specific to antibodies of the invention), and epitope-binding fragments of any of the above (e.g., VH domains, VL domains, or one or more CDR regions).
  • single chain antibodies e.g., single chain Fvs
  • Fab fragments fragments
  • F(ab') fragments fragments produced by a Fab expression library
  • anti-idiotypic antibodies including, e.g., anti-Id antibodies specific to antibodies of the invention
  • epitope-binding fragments of any of the above e.g., VH domains, V
  • the present invention encompasses albumin fusion proteins that comprise at least a fragment or variant of an antibody that binds a Therapeutic Protein (e.g., as disclosed in Table 1) or fragment or variant thereof.
  • Antibodies that bind a Therapeutic protein may be from any animal origin, including birds and mammals.
  • the antibodies are human, murine ⁇ e.g., mouse and rat), donkey, sheep, rabbit, goat, guinea pig, camel, horse, or chicken antibodies.
  • the antibodies are human antibodies.
  • "human" antibodies include antibodies having the amino acid sequence of a human immunoglobulin and include antibodies isolated from human immunoglobulin libraries and xenomice or other organisms that have been genetically engineered to produce human antibodies.
  • the antibody molecules that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention can be of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., IgGl, IgG2, IgG3, IgG4, IgAl and IgA2) or subclass of immunoglobulin molecule.
  • the antibody molecules that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein are IgGl, In other preferred embodiments, the immunoglobulin molecules that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein are IgG2. In other preferred embodiments, the immunoglobulin molecules that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein are IgG4.
  • the antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein are human antigen-binding antibody fragments of the present invention and include, but are not limited to, Fab, Fab' and F(ab')2, Fd, single-chain Fvs (scFv), single-chain antibodies, disulfide-linked Fvs (sdFv) and fragments comprising either a VL or VH domain.
  • Antigen-binding antibody fragments, including single-chain antibodies may comprise the variable region(s) alone or in combination with the entirety or a portion of the following: hinge region, CHl, CH2, and CH3 domains.
  • the antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein may be monospecific, bispecif ⁇ c, trispecif ⁇ c or of greater multispecif ⁇ city.
  • Multispecific antibodies may be specific for different epitopes of a Therapeutic protein or may be specific for both a Therapeutic protein as well as for a heterologous epitope, such as a heterologous polypeptide or solid support material. See, e.g., PCT publications WO 93/17715; WO 92/08802; WO 91/00360; WO 92/05793; Tutt, et al., J. Immunol. 147:60-69 (1991); U.S. Patent Nos.
  • Antibodies that bind a Therapeutic protein may be bispecific or bifunctional which means that the antibody is an artificial hybrid antibody having two different heavy/light chain pairs and two different binding sites.
  • Bispecific antibodies can be produced by a variety of methods including fusion of hybridomas or linking of Fab' fragments. See, e.g., Songsivilai & Lachmann Clin. Exp. Immunol. 79: 315-321 (1990), Kostelny et al. J Immunol.
  • bispecific antibodies may be formed as "diabodies” (Holliger et al. '"Diabodies 1 : small bivalent and bispecific antibody fragments” PNAS USA 90:6444-6448 (1993)) or "Janusins” (Traunecker et al. "Bispecific single chain molecules (Janusins) target cytotoxic lymphocytes on HTV infected cells” EMBO J 10:3655-3659 (1991) and Traunecker et al. "Janusin: new molecular design for bispecific reagents" bit J Cancer Si ⁇ pll ':51-52 (1992)).
  • the present invention also provides albumin fusion proteins that comprise, fragments or variants (including derivatives) of an antibody described herein or known elsewhere in the art. Standard techniques known to those of skill in the art can be used to introduce mutations in the nucleotide sequence encoding a molecule of the invention, including, for example, site-directed mutagenesis and PCR-mediated mutagenesis which result in amino acid substitutions.
  • the variants encode less than 50 amino acid substitutions, less than 40 amino acid substitutions, less than 30 amino _acid_substitutions, less than 25 amino acid substitutions,- less than 20 amino acid substitutions, less than " 15 amino acid substitutions, less than 10 amino acid substitutions, less than 5 amino acid substitutions, less than 4 amino acid substitutions, less than 3 amino acid substitutions, or less than 2 amino acid substitutions relative to the reference VH domain, VHCDRl, VHCDR2, VHCDR3, VL domain, VLCDRl, VLCDR2, or VLCDR3.
  • the variants encode substitutions of VHCDR3.
  • the variants have conservative amino acid substitutions at one or more predicted non-essential amino acid residues.
  • Antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein may be described or specified in terms of the epitope(s) or portion(s) of a Therapeutic protein which they recognize or specifically bind.
  • Antibodies which specifically bind a Therapeutic protein or a specific epitope of a Therapeutic protein may also be excluded. Therefore, the present invention encompasses antibodies that specifically bind Therapeutic proteins, and allows for the exclusion of the same, hi preferred embodiments, albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein, binds the same epitopes as the unfused fragment or variant of that antibody itself.
  • Antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein may also be described or specified in terms of their cross-reactivity. Antibodies that do not bind any other analog, ortholog, or homolog of a Therapeutic protein are included. Antibodies that bind polypeptides with at least 95%, at least 90%, at least 85%, at least 80%, at least 75%, at least 70%, at least 65%, at least 60%, at least 55%, and at least 50% sequence identity (as calculated using methods known in the art and described herein) to a Therapeutic protein are also included in the present invention.
  • antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein cross-react with murine, rat and/or rabbit homologs of human proteins and the corresponding epitopes thereof.
  • Antibodies that do not bind polypeptides with less than 95%, less than 90%, less than 85%, less than 80%, less than 75%, less than 70%, less than 65%, less than 60%, less than 55%, and less than 50% sequence identity (as calculated using methods known in the art and described herein) to a Therapeutic protein are also included in the present invention.
  • albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein, has similar or substantially identical cross reactivity characteristics compared to the fragment or variant or that particular antibody itself.
  • antibodies which bind polypeptides encoded by polynucleotides which hybridize to a polynucleotide encoding a Therapeutic protein under stringent hybridization conditions are also included in the present invention.
  • Antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention may also be described or specified in terms of their binding affinity to a polypeptide of the invention.
  • Preferred binding affinities include those with a dissociation constant or Kd less than 5 X 10 '2 M, 1C 2 M, 5 X 10 "3 M 1 10 "3 M, 5 X 10 "4 M, 10 "4 M.
  • More preferred binding affinities include those with a dissociation constant or Kd less than 5 X 10 "5 M, 10 '5 M 1 5 X 1O -6 M, 10 '6 M, 5 X I 0 "7 M, I 0 7 M, 5 X 10 "8 M or 10 "8 M.
  • binding affinities include those with a dissociation constant or Kd less than 5 X 10 "9 M, 10 '9 M, 5 X W 10 M, lO "10 M, 5 X 1O-" M, 10 ' " M, 5 X 10 "12 M, I0"12 M, 5 X 1O -13 M, 10 '13 M, 5 X 10 "14 M, 10 " ' 4 M, 5 X 10 '15 M, or 10 " ' 5 M.
  • albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein, has an affinity for a given protein or epitope similar to that of the corresponding antibody (not fused to albumin) that binds a Therapeutic protein, taking into account the valency of the albumin fusion protein (comprising at least a fragment or variant of an antibody that binds a Therapeutic protein) and the valency of the corresponding antibody.
  • the invention also provides antibodies that competitively inhibit binding of an antibody to an epitope of a Therapeutic protein as determined by any method known in the art for determining competitive binding, for example, the immunoassays described herein.
  • the antibody competitively inhibits binding to the epitope by at least 95%, at least 90%, at least 85%, at least 80%, at least 75%, at least 70%, at least 60%, or at least 50%.
  • albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein, competitively inhibits binding of a second antibody to an epitope of a Therapeutic protein.
  • albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein, competitively inhibits binding of a second antibody to an epitope of a Therapeutic protein by at least 9 5 %, at least 90%, at least 85 %, at least 80%, at least 75%, at least 70%, at least 60%, or at least 50%.
  • Antibodies may bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention may act as agonists or antagonists of the Therapeutic protein.
  • the present invention includes antibodies which disrupt the receptor/ligand interactions with the polypeptides of the invention either partially or fully.
  • the invention features both receptor-specific antibodies and ligand-specific antibodies.
  • the invention also features receptor-specific antibodies which do not prevent ligand binding but prevent receptor activation. Receptor activation (i.e., signaling) may be determined by techniques described herein or otherwise known in the art.
  • receptor activation can be determined by detecting the phosphorylation (e.g., tyrosine or serine/threonine) of the receptor or its substrate by immunoprecipitation followed by western blot analysis (for example, as described supra).
  • phosphorylation e.g., tyrosine or serine/threonine
  • antibodies are provided that inhibit ligand activity or receptor activity by at least 95%, at least 90%, at least 85%, at least 80%, at least 75%, at least 70%, at least 60%, or at least 50% of the activity in absence of the antibody.
  • albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein, has similar or substantially similar characteristics with regard to preventing ligand binding and/or preventing receptor activation compared to an un-fused fragment or variant of the antibody that binds the Therapeutic protein.
  • the invention also features receptor-specific antibodies which both prevent ligand binding and receptor activation as well as antibodies that recognize the receptor-ligand complex, and, preferably, do not specifically recognize the unbound receptor or the unbound ligand.
  • antibodies which bind the ligand and prevent binding of the ligand to the receptor are included in the invention.
  • antibodies which activate the receptor may act as receptor agonists, i.e., potentiate or activate either all or a subset of the biological activities of the ligand-mediated receptor activation, for example, by inducing dimerization of the receptor.
  • the antibodies may be specified as agonists, antagonists or inverse agonists for biological activities comprising the specific biological activities of the Therapeutic proteins (e.g. as disclosed in Table 1).
  • the above antibody agonists can be made using methods known in the art.
  • albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein, have similar or substantially identical agonist or antagonist properties as an un-fused fragment or variant of the antibody that binds the Therapeutic protein.
  • Antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention may be used, for example, to purify, detect, and target Therapeutic proteins, including both in m vitro and in vivo diagnostic and therapeutic methods.
  • the antibodies have utility in immunoassays for qualitatively and quantitatively measuring levels of the Therapeutic protein in biological samples.
  • albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein, may be used, for example, to purify, detect, and target Therapeutic proteins, including both in vitro and in vivo diagnostic and therapeutic methods.
  • Antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein include derivatives that are modified, i.e., by the covalent attachment of any type of molecule to the antibody.
  • the antibody derivatives include antibodies that have been modified, e.g., by glycosylation, acetylation, pegylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to a cellular Iigand or other protein, etc.
  • any of numerous chemical modifications may be carried out by known techniques, including, but not limited to specific chemical cleavage, acetylation, formylation, metabolic synthesis of tunicamycin, etc. Additionally, the derivative may contain one or more non-classical amino acids. Albumin fusion proteins of the invention may also be modified as described above.
  • the antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention may be generated by any suitable method known in the art.
  • Polyclonal antibodies to an antigen-of-interest can be produced by various procedures well known in the art.
  • a Therapeutic protein may be administered to various host animals including, but not limited to, rabbits, mice, rats, etc. to induce the production of sera containing polyclonal antibodies specific for the antigen.
  • adjuvants may be used to increase the immunological response, depending on the host species, and include but are not limited to, Freund's (complete and incomplete), mineral gels such as aluminum hydroxide, surface active substances such as lysol ⁇ cithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanins, dinitrophenol, and potentially useful human adjuvants such as BCG (bacille Calmette-Guerin) and corynebacterium parvum. Such adjuvants are also well known in the art.
  • Monoclonal antibodies can be prepared using a wide variety of techniques known in the art including the use of hybridoma, recombinant, and phage display technologies, or a combination thereof.
  • monoclonal antibodies can be produced using hybridoma techniques including those known in the art and taught, for example, in Harlow et al., Antibodies: A Laboratory Manual, (Cold Spring Harbor Laboratory Press, 2nd ed. 1988); Hammerling, et al., in: Monoclonal Antibodies and T-CeIl Hybridomas 563-681 (Elsevier, N.Y., 1981) (said references incorporated by reference in their entireties).
  • the term “monoclonal antibody” as used herein is not limited to antibodies produced through hybridoma technology.
  • the term “monoclonal antibody” refers to an antibody that is derived from a single clone, including any eukaryotic, prokaryotic, or phage clone, and not the method by which it is produced.
  • mice can be immunized with a Therapeutic protein or fragment or variant thereof, an albumin fusion protein, or a cell expressing such a Therapeutic protein or fragment or variant thereof or albumin fusion protein.
  • an immune response e.g., antibodies specific for the antigen are detected in the mouse s_erurn
  • the .mouse spleen is harvested and splenocytes isolated.
  • the -splenocytes are then fused by well known techniques to any suitable myeloma cells, for example cells from cell line SP20 available from the ATCC.
  • Hybridomas are selected and cloned by limited dilution. The hybridoma clones are then assayed by methods known in the art for cells that secrete antibodies capable of binding a polypeptide of the invention. Ascites fluid, which generally contains high levels of antibodies, can be generated by immunizing mice with positive hybridoma clones.
  • the present invention provides methods of generating monoclonal antibodies as well as antibodies produced by the method comprising culturing a hybridoma cell secreting an antibody wherein, preferably, the hybridoma is generated by fusing splenocytes isolated from a mouse immunized with an antigen of the invention with myeloma cells and then screening the hybridomas resulting from the fusion for hybridoma clones that secrete an antibody able to bind a polypeptide of the invention.
  • EBV Epstein Barr Virus
  • Protocols for generating EBV-transformed B cell lines are commonly known in the art, such as, for example, the protocol outlined in Chapter 7.22 of Current Protocols in Immunology, Coligan et al., Eds., 1994, John Wiley & Sons, NY, which is hereby incorporated in its entirety by reference.
  • the source of B cells for transformation is commonly human peripheral blood, but B cells for transformation may also be derived from other sources including, but not limited to, lymph nodes, tonsil, spleen, tumor tissue, and infected tissues.
  • Tissues are generally made into single cell suspensions prior to EBV transformation. Additionally, steps may be taken to either physically remove or inactivate T cells (e.g., by treatment with cyclosporin A) in B cell-containing samples, because T cells from individuals seropositive for anti-EBV antibodies can suppress B cell immortalization by EBV.
  • EBV lines are generally polyclonal. However, over prolonged periods of cell cultures, EBV lines may become monoclonal or polyclonal as a result of the selective outgrowth of particular B cell clones.
  • polyclonal EBV transformed lines may be subcloned (e.g., by limiting dilution culture) or fused with a suitable fusion partner and plated at limiting dilution to obtain monoclonal B cell lines.
  • the present invention also provides a method of generating polyclonal or monoclonal human antibodies against polypeptides of the invention or fragments thereof, comprising EBV-transformation of human B cells.
  • Antibody fragments which recognize specific epitopes may be generated by known techniques.
  • Fab and F(ab')2 fragments of the invention may be produced by proteolytic cleavage of immunoglobulin molecules, using enzymes such as papain (to produce Fab fragments) or pepsin (to produce F(ab')2 fragments).
  • F(ab')2 fragments contain the variable region, the light chain constant region and the CHl domain of the heavy chain.
  • antibodies that bind to a Therapeutic protein can also be generated using various phage display methods known in the art.
  • phage display methods functional antibody domains are displayed on the surface of phage particles which carry the polynucleotide sequences encoding them.
  • phage can be utilized to display antigen binding domains expressed from a repertoire or combinatorial antibody library (e.g., human or murine).
  • Phage expressing an antigen binding domain that binds the antigen of interest can be selected or identified with antigen, e.g., using labeled antigen or antigen bound or captured to a solid surface or bead.
  • Phage used in these methods are typically filamentous phage including fd and M13 binding domains expressed from phage with Fab, Fv or disulfide stabilized Fv antibody domains recombinantly fused to either the phage gene HI or gene VIII protein.
  • phage display methods that can be used to make antibodies that bind to a Therapeutic protein include those disclosed in Brinkman et al., J. Immunol. Methods 182:41-50 (1995); Ames et al., J. Immunol. Methods 184:177-186 (1995); Kettleborough et al., Eur. J. Immunol.
  • the antibody coding regions from the phage can be isolated and used to generate whole antibodies, including human antibodies, or any other desired antigen binding fragment, and expressed in any desired host, including mammalian cells, insect cells, plant cells, yeast, and bacteria, e.g., as described in detail below.
  • a chimeric antibody is a molecule in which different portions of the antibody are derived from different animal species, such as antibodies having a variable region derived from a murine monoclonal antibody and a human immunoglobulin constant region.
  • Methods for producing chimeric antibodies are known in the art. See e.g., Morrison, Science 229:1202 (1985); Oi et al., BioTechniques 4:214 (1986); Gillies et al., (1989) J. Immunol. Methods 125:191-202; U.S. Patent Nos. 5,807,715; 4,816,567; and 4,816397, which are incorporated herein by reference in their entirety.
  • Humanized antibodies are antibody molecules from non-human species antibody that binds the desired antigen having one or more complementarity determining regions (CDRs) from the non-human species and a framework regions from a human immunoglobulin molecule.
  • CDRs complementarity determining regions
  • framework residues in the human framework regions will be substituted with the corresponding residue from the CDR donor antibody to alter, preferably improve, antigen binding.
  • These framework substitutions are identified by methods well known in the art, e.g., by modeling of the interactions of the CDR and framework residues to identify framework residues important for antigen binding and sequence comparison to identify unusual framework residues at particular positions. (See, e.g., Queen et al., U.S. Patent No.
  • Antibodies can be humanized using a variety of techniques known in the art including, for example, CDR-grafting (EP 239,400; PCT publication WO 91/09967; U.S. Patent Nos. 5,225,539; 5,530,101; and 5,585,089), veneering or resurfacing (EP 592,106; EP 519,596; Padlan, Molecular Immunology 28(4/5):489-498 (1991); Studnicka et al., Protein Engineering 7(6):805-814 (1994); Roguska. et al., PNAS 91:969-973 (1994)), and chain shuffling (U.S. Patent No. 5,565,332).
  • Human antibodies are particularly desirable for therapeutic treatment of human patients.
  • Human antibodies can be made by a variety of methods known in the art including phage display methods described above using antibody libraries derived from human immunoglobulin sequences. See also, U.S. Patent Nos. 4,444,887 and 4,716,111; and PCT publications WO 98/46645, WO 98/50433, WO 98/24893, WO 98/16654, WO 96/34096, WO 96/33735, and WO 91/10741; each of which is incorporated herein by reference in its entirety.
  • Human antibodies can also be produced using transgenic mice which are incapable of expressing functional endogenous immunoglobulins, but which can express human immunoglobulin genes.
  • the human heavy and light chain immunoglobulin gene """ complexes' may b" ⁇ T ⁇ trodu'ced randomly or " by homologous recombination into mouse embryonic stem cells.
  • the human variable region, constant region, and diversity region may be introduced into mouse embryonic stem cells in addition to the human heavy and light chain genes.
  • the mouse heavy and light chain immunoglobulin genes may be rendered non-functional separately or simultaneously with the introduction of human immunoglobulin loci by homologous recombination.
  • homozygous deletion of the JH region prevents endogenous antibody production.
  • the modified embryonic stem cells are expanded and microinjected into blastocysts to produce chimeric mice.
  • the chimeric mice are then bred to produce homozygous offspring which express human antibodies.
  • the transgenic mice are immunized in the normal fashion with a selected antigen, e.g., all or a portion of a polypeptide of the invention.
  • Monoclonal antibodies directed against the antigen can be obtained from the immunized, transgenic mice using conventional hybridoma technology.
  • the human immunoglobulin transgenes harbored by the transgenic mice rearrange during B cell differentiation, and subsequently undergo class switching and somatic mutation.
  • Completely human antibodies which recognize a selected epitope can be generated using a technique referred to as "guided selection.”
  • a selected non-human monoclonal antibody e.g., a mouse antibody, is used to guide the selection of a completely human antibody recognizing the same epitope. (Jespers et al., Bio/technology 12:899-903 (1988)).
  • the invention further provides polynucleotides comprising a nucleotide sequence encoding an antibody and fragments thereof.
  • the invention also encompasses polynucleotides that hybridize under stringent or alternatively, under lower stringency hybridization conditions, e.g., as defined supra, to polynucleotides that encode an antibody, preferably, that specifically binds to a Therapeutic protein, and more preferably, an antibody that binds to a polypeptide having the amino acid sequence of a "Therapeutic protein:X" as disclosed in the "SEQ ID NO:Z" column of Table 2.
  • the polynucleotides may be obtained, and the nucleotide sequence of the polynucleotides determined, by any method known in the art.
  • a polynucleotide encoding the antibody may be assembled from chemically synthesized oligonucleotides (e.g., as described in Kutmeier et al., BioTechniques 17:242 (1994)), which, briefly, involves the synthesis of overlapping oligonucleotides containing portions of the sequence encoding the antibody, annealing and ligating of those oligonucleotides ⁇ and then " ⁇ amplification of the ligated oligonucleotides by PCR.
  • a polynucleotide encoding an antibody may be generated from nucleic acid from a suitable source. If a clone containing a nucleic acid encoding a particular antibody is not available, but the sequence of the antibody molecule is known, a nucleic acid encoding the immunoglobulin may be chemically synthesized or obtained from a suitable source (e.g., an antibody cDNA library, or a cDNA library generated from, or nucleic acid, preferably poly A+ RNA, isolated from, any tissue or cells expressing the antibody, such as hybridoma cells selected to express an antibody) by PCR amplification using synthetic primers hybridizable to the 3' and 5 1 ends of the sequence or by cloning using an oligonucleotide probe specific for the particular gene sequence to identify, e.g., a cDNA clone from a cDNA library that encodes the antibody. Amplified nucleic acids generated by PCR may then
  • nucleotide sequence and corresponding amino acid sequence of the antibody may be manipulated using methods well known in the art for the manipulation of nucleotide sequences, e.g., recombinant DNA techniques, site directed mutagenesis, PCR, etc.
  • the amino acid sequence of the heavy and/or light chain variable domains may be inspected to identify the sequences of the complementarity determining regions (CDRs) by methods that are well know in the art, e.g., by comparison to known amino acid sequences of other heavy and light chain variable regions to determine the regions of sequence hypervariability.
  • CDRs complementarity determining regions
  • one or more of the CDRs may be inserted within framework regions, e.g., into human framework regions to humanize a non-human antibody, as described supra.
  • the framework regions may be naturally occurring or consensus framework regions, and preferably human framework regions (see, e.g., Chothia et al., J. MoI. Biol.
  • the polynucleotide generated by the combination of the framework regions and CDRs encodes an antibody that specifically binds a polypeptide of the " invention.
  • one or more amino acid substitutions may be made within the framework regions, and, preferably, the amino acid substitutions improve binding of the antibody to its antigen. Additionally, such methods may be used to make amino acid substitutions or deletions of one or more variable region cysteine residues participating in an intrachain disulfide bond to generate antibody molecules lacking one or more intrachain disulfide bonds.
  • a chimeric antibody is a molecule in which different portions are derived from different animal species, such as those having a variable region derived from a murine mAb and a human immunoglobulin constant region, e.g., humanized antibodies.
  • an antibody, or fragment, derivative or analog thereof e.g., a heavy or light chain of an antibody or a single chain antibody
  • an expression vector containing a polynucleotide that encodes the antibody Once a polynucleotide encoding an antibody molecule or a heavy or light chain of an antibody, or portion thereof (preferably containing the heavy or light chain variable domain), of the invention has been obtained, the vector for the production of the antibody molecule may be produced by recombinant DNA technology using techniques well known in the art. Thus, methods for preparing a protein by expressing a polynucleotide containing an antibody encoding nucleotide sequence are described herein.
  • the invention provides replicable vectors comprising a nucleotide sequence encoding an antibody molecule of the invention, or a heavy or light chain thereof, or a heavy or light chain variable domain, operably linked to a promoter.
  • Such vectors may include the nucleotide sequence encoding the constant region of the antibody molecule (see, e.g., PCT Publication WO 86/05807; PCT Publication WO 89/01036; and U.S. Patent No. 5,122,464) and the variable domain of the antibody may be cloned into such a vector for expression of the entire heavy or light chain.
  • the expression vector is transferred to a host cell by conventional techniques and the transfected cells are then cultured by conventional techniques to produce an antibody.
  • the invention includes host cells containing a polynucleotide encoding an antibody of the invention, or a - heavy or light chain thereof, or a single chain antibody, operably linked to a heterologous promoter.
  • vectors encoding both the heavy and light chains may be co-expressed in the host cell for expression of the entire immunoglobulin molecule, as detailed below.
  • host-expression vector systems may be utilized to express the antibody molecules of the invention.
  • Such host-expression systems represent vehicles by which the coding sequences of interest may be produced and subsequently purified, but also represent cells which may, when transformed or transfected with the appropriate nucleotide coding sequences, express an antibody molecule of the invention in situ.
  • These include but are not limited to microorganisms such as bacteria (e.g., E. coli, B.
  • subtilis transformed with recombinant bacteriophage DNA, plasmid DNA or cosmid DNA expression vectors containing antibody coding sequences; yeast (e.g., Saccharomyces, Pichia) transformed with recombinant yeast expression vectors containing antibody coding sequences; insect cell systems infected with recombinant virus expression vectors (e.g., baculovirus) containing antibody coding sequences; plant cell systems infected with recombinant virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or transformed with recombinant plasmid expression vectors (e.g., Ti plasmid) containing antibody coding sequences; or mammalian cell systems (e.g., COS, CHO, BHK, 293, 3T3 cells) harboring recombinant expression constructs containing promoters derived from the genome of mammalian cells (e.g., metallothionein promoter) or from mamm
  • bacterial cells such as Escherichia coli, and more preferably, eukaryotic cells, especially for the expression of whole recombinant antibody molecule, are used for the expression of a recombinant antibody molecule.
  • mammalian cells such as Chinese hamster ovary cells (CHO), in conjunction with a vector such as the major intermediate early gene promoter element from human cytomegalovirus is an effective expression system for antibodies (Foecking et al., Gene 45:101 (1986); Cockett et al., Bio/Technology 8:2 (1990)).
  • a number of expression vectors may be advantageously selected depending upon the use intended for the antibody molecule being expressed.
  • vectors which direct the expression of high levels of fusion protein products that are readily purified may be desirable.
  • Such vectors include, but are not limited, to the E. coli expression vector pUR278 (Ruther et al., EMBO J. 2:1791 (1983)), in which the antibody coding sequence may be ligated individually into the vector in frame with the lac Z coding region so that a fusion protein is produced; pIN vectors (Inouye & Inouye, Nucleic Acids Res.
  • pGEX vectors may also be used to express foreign polypeptides as fusion proteins with glutathione S-transferase (GST).
  • GST glutathione S-transferase
  • fusion proteins are soluble and can easily be purified from lysed cells by adsorption and binding to matrix glutathione-agarose beads followed by elution in the presence of free glutathione.
  • the pGEX vectors are designed to include thrombin or factor Xa protease cleavage sites so that the cloned target gene product can be released from the GST moiety.
  • Autographa californica nuclear polyhedrosis virus (AcNPV) is used as a vector to express foreign genes.
  • the virus grows in Spodoplera ⁇ - ⁇ giperda cells.
  • the antibody coding sequence may be cloned individually into non-essential regions (for example the polyhedrin gene) of the virus and placed under control of an AcNPV promoter (for example the polyhedrin promoter).
  • a number of viral-based expression systems may be utilized.
  • the antibody coding sequence of interest may be ligated to an adenovirus transcription/translation control complex, e.g., the late promoter and tripartite leader sequence.
  • This chimeric gene may then be inserted in the adenovirus genome by in vitro or in vivo recombination. Insertion in a non- essential region of the viral genome (e.g., region El or E3) will result in a recombinant virus that is viable and capable of expressing the antibody molecule in infected hosts, (e.g., see Logan & Shenk, Proc. Natl.
  • Specific initiation signals may also be required for efficient translation of inserted antibody coding sequences. These signals include the ATG initiation codon and adjacent sequences. Furthermore, the initiation codon must be in phase with the reading frame of the desired coding sequence to ensure translation of the entire insert. These exogenous translational control signals and initiation codons can be of a variety of origins, both natural and synthetic. The efficiency of expression may be enhanced by the inclusion of appropriate transcription enhancer elements, transcription terminators, etc. (see Bittner et al., Methods in Enzymol. 153:51-544 (1987)).
  • a host cell strain may be chosen which modulates the expression of the inserted sequences, or modifies and processes the gene product in the specific fashion desired. Such modifications (e.g., glycosylation) and processing (e.g., cleavage) of protein products may be important for the function of the protein.
  • Different host cells have characteristic and specific mechanisms for the post-translational processing and modification of proteins and gene products. Appropriate cell lines or host systems can be chosen to ensure the correct modification and processing of the foreign protein expressed.
  • eukaryotic host cells which possess the cellular machinery for proper processing of the primary transcript, glycosylation, and phosphorylation of the gene product may be used.
  • Such mammalian host cells include but are not limited to CHO, VERY, BHK, HeIa, COS, MDCK, 293, 3T3, WI38, and in particular, breast cancer cell lines such as, for example, BT483, Hs578T, HTB2, BT20 and T47D, and normal mammary gland cell line such as, for example, CRL7030 and Hs578Bst.
  • cell lines which stably express the antibody molecule may be engineered.
  • host cells can be - transformed with DNA controlled by appropriate expression control elements (e.g., promoter, enhancer, sequences, transcription terminators, polyadenylation sites, etc.), and a selectable marker.
  • appropriate expression control elements e.g., promoter, enhancer, sequences, transcription terminators, polyadenylation sites, etc.
  • engineered cells may be allowed to grow for 1-2 days in an enriched media, and then are switched to a selective media.
  • the selectable marker in the recombinant plasmid confers resistance to the selection and allows cells to stably integrate the plasmid into their chromosomes and grow to form foci which in turn can be cloned and expanded into cell lines.
  • This method may advantageously be used to engineer cell lines which express the antibody molecule.
  • Such engineered cell lines may be particularly useful in screening and evaluation of compounds that interact directly or indirectly with the antibody molecule.
  • a number of selection systems may be used, including but not limited to the herpes simplex virus thymidine kinase (Wigler et al., Cell 11:223 (1977)), hypoxanthine-guanine phosphoribosyltransferase (Szybalska & Szybalski, Proc. Natl. Acad. Sci. USA 48:202 (1992)), and adenine phosphoribosyltransferase (Lowy et al., Cell 22:817 (1980)) genes can be employed in tk-, hgprt- or aprt- cells, respectively.
  • antimetabolite resistance can be used as the basis of selection for the following genes: dhfr, which confers resistance to methotrexate (Wigler et al., Natl. Acad. Sci. USA 77:357 (1980); O'Hare et al., Proc. Natl. Acad. Sci. USA 78:1527 (1981)); gpt, which confers resistance to mycophenolic acid (Mulligan & Berg, Proc. Natl. Acad. Sci.
  • the expression levels of an antibody molecule can be increased by vector amplification (for a review, see Bebbington and Hentschel, The use of vectors based on gene amplification for the expression of cloned genes in mammalian cells in DNA cloning, Vol.3. (Academic Press, New York, 1987)).
  • vector amplification for a review, see Bebbington and Hentschel, The use of vectors based on gene amplification for the expression of cloned genes in mammalian cells in DNA cloning, Vol.3. (Academic Press, New York, 1987)).
  • a marker in the vector system expressing antibody is amplifiable, increase in the level of inhibitor present in culture of ' host' cel ⁇ "wnT'i ⁇ crease the number "of copies of the marker gene. Since the amplified region is associated with the antibody gene, production of the antibody will also increase (Crouse et al., MoI. Cell. Biol. 3:2 5 7 (1983)
  • Vectors which use glutamine synthase (GS) or DHFR as the selectable markers can be amplified in the presence of the drugs methionine sulphoximine or methotrexate, respectively.
  • An advantage of glutamine synthase based vectors are the availability of cell lines (e.g., the murine myeloma cell line, NSO) which are glutamine synthase negative.
  • Glutamine synthase expression systems can also function in glutamine synthase expressing cells (e.g. Chinese Hamster Ovary (CHO) cells) by providing additional inhibitor to prevent the functioning of the endogenous gene.
  • a glutamine synthase expression system and components thereof are detailed in PCT publications: WO87/04462; WO86/05807; WO89/01036; WO89/10404; and WO91/066 5 7 which are incorporated in their entireties by reference herein. Additionally, glutamine synthase expression vectors that may be used according to the present invention are commercially available from suppliers, including, for example Lonza Biologies, Inc. (Portsmouth, NH). Expression and production of monoclonal antibodies using a GS expression system in murine myeloma cells is described in Bebbington et al., Bio/iechnology 10:169(1992) and in Biblia and Robinson Biotechnol. Prog. 11:1 (1995) which are incorporated in their entireties by reference herein.
  • the host cell may be co-transfected with two expression vectors of the invention, the first vector encoding a heavy chain derived polypeptide and the second vector encoding a light chain derived polypeptide.
  • the two vectors may contain identical selectable markers which enable equal expression of heavy and light chain polypeptides.
  • a single vector may be used which encodes, and is capable of expressing, both heavy and light chain polypeptides. In such situations, the light chain should be placed before the heavy chain to avoid an excess of toxic free heavy chain (Proudfoot, Nature 322:52 (1986); Kohler, Proc. Natl. Acad. Sci. USA 77:2197 (1980)).
  • the coding sequences for the heavy and light chains may comprise cDNA or genomic DNA.
  • an antibody molecule of the invention may be purified by any method known in the art for purification of an immunoglobulin molecule, for example, by chromatography (e.g., ion exchange, affinity, particularly by affinity for the specific antigen after Protein A, and sizing column chromatography), centrifugation, differential solubility, or by any other standard technique for the purification of proteins.
  • chromatography e.g., ion exchange, affinity, particularly by affinity for the specific antigen after Protein A, and sizing column chromatography
  • centrifugation e.g., ion exchange, affinity, particularly by affinity for the specific antigen after Protein A, and sizing column chromatography
  • differential solubility e.g., differential solubility
  • the antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention or fragments thereof can be fused to heterologous polypeptide sequences described herein or otherwise known in the art, to facilitate purification.
  • Antibodies that bind a Therapeutic protein or fragments or variants can be fused to marker sequences, such as a peptide to facilitate purification.
  • the marker amino acid sequence is a hexa-histidine peptide, such as the tag provided in a pQE vector (QIAGEN, Inc., 9259 Eton Avenue, Chatsworth, CA, 91311), among others, many of which are commercially available.
  • a hexa-histidine provides for convenient purification of the fusion protein.
  • peptide tags useful for purification include, but are not limited to, the hemagglutinin tag (also called the "HA tag”), which corresponds to an epitope derived from the influenza hemagglutinin protein (Wilson et al., Cell 37:767 (1984)) and the "flag" tag.
  • HA tag also called the "HA tag”
  • the present invention further encompasses antibodies or fragments thereof conjugated to a diagnostic or therapeutic agent.
  • the antibodies can be used diagnostically to, for example, monitor the development or progression of a tumor as part of a clinical testing procedure to, e.g., determine the efficacy of a given treatment regimen. Detection can be facilitated by coupling the antibody to a detectable substance. Examples of detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials, bioluminescent materials, radioactive materials, positron emitting metals using various positron emission tomographies, and nonradioactive paramagnetic metal ions.
  • the detectable substance may be coupled or conjugated either directly to the antibody (or fragment thereof) or indirectly, through an intermediate (such as, for example, a linker known in the art) using techniques known in the art. See, for example, U.S. Patent No. 4,741,900 for metal ions which can be conjugated to antibodies for use as diagnostics according to the present invention.
  • suitable enzymes include horseradish peroxidase, alkaline phosphatase, beta-galactosidase, or acetylcholinesterase;
  • suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin;
  • suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin;
  • an example of a luminescent material includes luminol;
  • examples of bioluminescent materials include luciferase, luciferin, and aequorin;
  • suitable radioactive material include 1251, 1311, 11 ISi or 99Tc. Other examples of detectable substances have been described elsewhere herein.
  • an antibody of the invention may be conjugated to a therapeutic moiety such as a cytotoxin, e.g., a cytostatic or cytocidal agent, a therapeutic agent or a radioactive metal ion, e.g., alpha-emitters such as, for example, 213Bi.
  • a cytotoxin or cytotoxic agent includes any agent that is detrimental to cells.
  • Examples include paclitaxol, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicin, doxorubicin, daunorubicin, dihydroxy anthracin dione, mitoxantrone, mithramycin, actinomycin D, 1-dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, and puromycin and analogs or homologs thereof.
  • Therapeutic agents include, but are not limited to, antimetabolites (e.g., methotrexate, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-fluorouracil decarbazine), alkylating agents (e.g., mechlorethamine, thioepa chlorambucil, melphalan, carmustine (BSNU) and lomustine (CCNU), cyclothosphamide, tfusulfa ⁇ ' ⁇ romomannitol ' .
  • antimetabolites e.g., methotrexate, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-fluorouracil decarbazine
  • alkylating agents e.g., mechlorethamine, thioepa chlorambucil, melphalan, carmustine (BSNU) and lomustine (CCNU)
  • BSNU thioepa chlorambucil
  • streptozotocm " mitomycin C, and cis- diohlorodiamine platinum (H) (DDP) cisplatin), anthracyolines (e.g., daunorubicin (formerly daunomycin) and doxorubicin), antibiotics (e.g., dactinomycin (formerly actinomycin), bleomycin, mithramycin, and anthramycin (AMC)), and anti-mitotic agents (e.g., vincristine and vinblastine).
  • DDP diohlorodiamine platinum
  • anthracyolines e.g., daunorubicin (formerly daunomycin) and doxorubicin
  • antibiotics e.g., dactinomycin (formerly actinomycin), bleomycin, mithramycin, and anthramycin (AMC)
  • anti-mitotic agents e.g., vincristine and vinblastine.
  • the conjugates of the invention can be used for modifying a given biological response, the therapeutic agent or drug moiety is not to be construed as limited to classical chemical therapeutic agents.
  • the drug moiety may be a protein or polypeptide possessing a desired biological activity.
  • Such proteins may include, for example, a toxin such as abrin, ricin A, pseudomonas exotoxin, or diphtheria toxin; a protein such as tumor necrosis factor, alpha-interferon, ⁇ -interferon, nerve growth factor, platelet derived growth factor, tissue plasminogen activator, an apoptotic agent, e.g., TNF-alpha, TNF-beta, AIM I (See, International Publication No. WO 97/33899), AM ⁇ (See, International Publication No. WO 97/34911), Fas Ligand (Takahashi et al, Int.
  • a toxin such as abrin, ricin A, pseudomonas exotoxin, or diphtheria toxin
  • a protein such as tumor necrosis factor, alpha-interferon, ⁇ -interferon, nerve growth factor, platelet derived growth factor, tissue plasminogen activator, an a
  • VEGI See, International Publication No. WO 99/23105
  • a thrombotic agent or an anti- angiogenic agent e.g., angiostatin or endostatin
  • biological response modifiers such as, for example, lymphokines, interleukin-1 ("DL-I”), interleukin-2 (“IL-2”), interleukin-6 (“IL-6”), granulocyte macrophage colony stimulating factor (“GM-CSF”), granulocyte colony stimulating factor (“G-CSF”), or other growth factors.
  • DL-I lymphokines
  • IL-2 interleukin-2
  • IL-6 interleukin-6
  • GM-CSF granulocyte macrophage colony stimulating factor
  • G-CSF granulocyte colony stimulating factor
  • Antibodies may also be attached to solid supports, which are particularly useful for immunoassays or purification of the target antigen.
  • solid supports include, but are not limited to, glass, cellulose, polyacrylamide, nylon, polystyrene, polyvinyl chloride or polypropylene.
  • Techniques for conjugating such therapeutic moiety to antibodies are well known. See, for example, Arnon et al., "Monoclonal Antibodies For Immunotargeting Of Drugs In Cancer Therapy", in Monoclonal Antibodies And Cancer Therapy, Reisfeld et al. (eds.), pp. 243-56 (Alan R. Liss, Inc.
  • an antibody can be conjugated to a second antibody to form an antibody heteroconjugate as described by Segal in U.S. Patent No. 4,676,980, which is incorporated herein by reference in its entirety.
  • An antibody, with or without a therapeutic moiety conjugated to it, administered alone or in combination with cytotoxic factor(s) and/or cytokine(s) can be used as a therapeutic.
  • Antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention include, but are not limited to, antibodies that bind A Therapeutic protein disclosed-in the-Therapeutic Protein X" column of Table 1, or a fragment or variant thereof.
  • the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of, the VH domain.
  • the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of, one, two or three VH CDRs.
  • the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of, the VH CDRl.
  • the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of, the VH CDR2.
  • the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of, the VH CDR3.
  • the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of, the VL domain.
  • the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of, one, two or three VL CDRs.
  • the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of, the VL CDRl.
  • the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of, the VL CDR2.
  • the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists o ⁇ the VL CDR3.
  • the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of, one, two, three, four, five, or six VH and/or VL CDRs.
  • the "fragment or variant of an antibody that immunospecifically binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of, an scFv comprising the VH domain of the Therapeutic antibody, linked to the VL domain of the therapeutic antibody by a peptide linker such as (Gly 4 Ser) 3 (SEQ ID NO:4).
  • the antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein (or fragment or variant thereof) may be utilized for immunophenotyping of cell lines and biological samples.
  • Therapeutic proteins of the present invention may be useful as cell-specific markers, or more specifically as cellular markers that are differentially expressed at various stages of differentiation and/or maturation of particular cell types.
  • Monoclonal antibodies (or albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein) directed against a specific epitope, or combination of epitopes, will allow for the screening of cellular populations expressing the marker.
  • Various techniques can be utilized using monoclonal antibodies (or albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein) to screen for cellular populations expressing the marker(s), and include magnetic separation using antibody-coated magnetic beads, "panning" with antibody attached to a solid matrix (i.e., plate), and flow cytometry (See, e.g., U.S. Patent 5,985,660; and Morrison et al., Cell, 96:131-19 (1999)).
  • These techniques allow for the screening of particular populations of cells, such as might be found with hematological malignancies (i.e.
  • MRD minimal residual disease
  • GVHD Graft-versus-Host Disease
  • the antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein (or fragment or variant thereof) may be characterized in a variety of ways.
  • Albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein may be assayed for the ability to specifically bind to the same antigens specifically bound by the antibody that binds a Therapeutic protein corresponding to the antibody that binds a Therapeutic protein portion of the albumin fusion protein using techniques described herein or routinely modifying techniques known in the art.
  • Assays for the ability of the antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein (or fragment or variant thereof) to (specifically) bind a specific protein or epitope may be performed in solution (e.g., Houghten, Bio/Techniques 13:412-421(1992)), on beads ⁇ e.g., Lam, Nature 354:82-84 (1991)), on chips (e.g., Fodor, Nature 364:555-556 (1993)), on bacteria (e.g., U.S. Patent No. 5,223,409), on spores (e.g., Patent Nos.
  • the antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein (or fragment or variant thereof) may also be assayed for their specificity and affinity for a specific protein or epitope using or routinely modifying techniques described herein or otherwise known in the art.
  • the albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein may be assayed for cross-reactivity with other antigens (e.g., molecules that have sequence/structure conservation with the molecule(s) specifically bound by the antibody that binds a Therapeutic protein (or fragment or variant thereof) corresponding to the Therapeutic protein portion of the albumin fusion protein of the invention) by any method known in the art.
  • antigens e.g., molecules that have sequence/structure conservation with the molecule(s) specifically bound by the antibody that binds a Therapeutic protein (or fragment or variant thereof) corresponding to the Therapeutic protein portion of the albumin fusion protein of the invention
  • Immunoassays which can be used to analyze (immunospecific) binding and cross-reactivity include, but are not limited to, competitive and non-competitive assay systems using techniques such as western blots, radioimmunoassays, ELISA (enzyme linked immunosorbent assay), "sandwich” immunoassays, immunoprecipitation assays, precipitin reactions, gel diffusion precipitin reactions, immunodiffusion assays, agglutination assays, complement-fixation assays, immunoradiometric assays, fluorescent immunoassays, and protein A immunoassays, to name but a few.
  • competitive and non-competitive assay systems using techniques such as western blots, radioimmunoassays, ELISA (enzyme linked immunosorbent assay), "sandwich” immunoassays, immunoprecipitation assays, precipitin reactions, gel diffusion precipitin reactions, immunodiffusion assays, aggluti
  • Immunoprecipitation protocols generally comprise lysing a population of cells in a lysis buffer such as RIPA buffer (1% NP-40 or Triton X-100, 1% sodium deoxycholate, 0.1% SDS, 0.15 M NaCl, 0.01 M sodium phosphate at pH 7.2, 1% Trasylol) supplemented with protein phosphatase and/or protease inhibitors (e.g., EDTA, PMSF, aprotinin, sodium vanadate), adding an antibody of the invention or albumin fusion protein of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein (or fragment or variant thereof) to the cell Iysate, incubating for a period of time (e.g., 1 to 4 hours) at 40 degrees C, adding protein A and/or protein G sepharose beads (or beads coated with an appropriate anti-idiotypic antibody or anti-albumin antibody in the case when an albumin fusion protein comprising at least a fragment
  • the ability of the antibody or albumin fusion protein of the invention to immunoprecipitate a particular antigen can be assessed by, e.g., western blot analysis.
  • One of skill in the art would be knowledgeable as to the parameters that can be modified to increase the binding of the antibody or albumin fusion protein to an antigen and decrease the background (e.g., pre-clearing the cell lysate with sepharose beads).
  • immunoprecipitation protocols see, e.g., Ausubel et al, eds, 1994, Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York at 10.16.1.
  • Western blot analysis generally comprises preparing protein samples, electrophoresis of the protein samples in a polyacrylamide gel (e.g., 8%- 20% SDS-PAGE depending on the molecular weight of the antigen), transferring the protein sample from the polyacrylamide gel to a membrane such as nitrocellulose, PVDF or nylon, blocking the membrane in blocking solution (e.g., PBS with 3% BSA or non-fat milk), washing the membrane in washing buffer (e.g., PBS-T ween 20), applying the antibody or albumin fusion protein of the invention (diluted in blocking buffer) to the membrane, washing the membrane in washing buffer, applying a secondary antibody (which recognizes the albumin fusion protein, e.g., an anti-human serum albumin antibody) conjugated to an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase) or radioactive molecule (e.g., 32 P or 125 I) diluted in blocking buffer, washing the membrane in wash buffer, and
  • ELISAs comprise preparing antigen, coating the well of a 96-well microtiter plate with the antigen, washing away antigen that did not bind the wells, adding the antibody or albumin fusion protein (comprising at least a fragment or variant of an antibody that binds a Therapeutic protein) of the invention conjugated to a detectable compound such as an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase) to the wells and incubating for a period of time, washing away unbound or non-specif ⁇ cally bound albumin fusion proteins, and detecting the presence of the antibody or albumin fusion proteins specifically bound to the antigen coating the well.
  • an enzymatic substrate e.g., horseradish peroxidase or alkaline phosphatase
  • the antibody or albumin fusion protein does not have to be conjugated to a detectable compound; instead, a second antibody (which recognizes the antibody or albumin fusion protein, respectively) conjugated to a detectable compound may be added to the well. Further, instead of coating the well with the antigen, antibody or the albumin fusion protein may be coated to the well.
  • the detectable molecule could be the antigen conjugated to a detectable compound such as an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase).
  • an enzymatic substrate e.g., horseradish peroxidase or alkaline phosphatase.
  • the binding affinity of an albumin fusion protein to a protein, antigen, or epitope and the off-rate of an antibody- or albumin fusion protein-protein/antigen/epitope interaction can be determined by competitive binding -assays.
  • a competitive binding assay is a " radioimmunoassay comprising the incubation of labeled antigen (e.g., 3 H or 125 I) with the antibody or albumin fusion protein of the invention in the presence of increasing amounts of unlabeled antigen, and the detection of the antibody bound to the labeled antigen.
  • labeled antigen e.g., 3 H or 125 I
  • the affinity of the antibody or albumin fusion protein of the invention for a specific protein, antigen, or epitope and the binding off-rates can be determined from the data by Scatehard plot analysis. Competition with a second protein that binds the same protein, antigen or epitope as the antibody or albumin fusion protein, can also be determined using radioimmunoassays.
  • the protein, antigen or epitope is incubated with an antibody or albumin fusion protein of the invention conjugated to a labeled compound (e.g., 3 H or 115 I) in the presence of increasing amounts of an unlabeled second protein that binds the same protein, antigen, or epitope as the albumin fusion protein of the invention.
  • a labeled compound e.g., 3 H or 115 I
  • BIAcore kinetic analysis is used to determine the binding on and off rates of antibody or albumin fusion proteins of the invention to a protein, antigen or epitope.
  • BIAcore kinetic analysis comprises analyzing the binding and dissociation of antibodies, albumin fusion proteins, or specific polypeptides, antigens or epitopes from chips with immobilized specific polypeptides, antigens or epitopes, antibodies or albumin fusion proteins, respectively, on their surface.
  • the present invention is further directed to antibody-based therapies which involve administering antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein to an animal, preferably a mammal, and most preferably a human, patient for treating one or more of the disclosed diseases, disorders, or conditions.
  • Therapeutic compounds of the invention include, but are not limited to, antibodies of the invention (including fragments, analogs and derivatives thereof as described herein), nucleic acids encoding antibodies of the invention (including fragments, analogs and derivatives thereof and anti-idiotypic antibodies as described herein), albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein, and nucleic acids encoding such albumin fusion proteins.
  • the antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein can be used to treat, inhibit or prevent diseases, disorders or conditions associated with aberrant expression and/or activity of a Therapeutic protein, including, but not limited to, any one or more of the diseases, disorders, or conditions described herein.
  • the treatment and/or prevention of diseases, disorders, or conditions associated with aberrant expression and/or activity of a Therapeutic protein includes, but is not limited to, alleviating symptoms associated with those diseases, disorders or conditions, antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein may be provided in pharmaceutically acceptable compositions as known in the art or as described herein.
  • the present invention is directed to antibody-based therapies which involve administering antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein to an animal, preferably a mammal, and most preferably a human, patient for treating one or more diseases, disorders, or conditions, including but not limited to: neural disorders, immune system disorders, muscular disorders, reproductive disorders, gastrointestinal disorders, pulmonary disorders, cardiovascular disorders, renal disorders, proliferative disorders, and/or cancerous diseases and conditions., and/or as described elsewhere herein.
  • Therapeutic compounds of the invention include, but are not limited to, antibodies of the invention (e.g., antibodies directed to the full length protein expressed on the cell surface of a mammalian cell; antibodies directed to an epitope of a Therapeutic protein and nucleic acids encoding antibodies of the invention (including fragments, analogs and derivatives thereof and anti-idiotypic antibodies as described herein).
  • the antibodies of the invention can be used to treat, inhibit or prevent diseases, disorders or conditions associated with aberrant expression and/or activity of a Therapeutic protein, including, but not limited to, any one or more of the diseases, disorders, or conditions described herein.
  • the treatment and/or prevention of diseases, disorders, or conditions associated with aberrant expression and/or activity of a Therapeutic protein includes, but is not limited to, alleviating symptoms associated with those diseases, disorders or conditions.
  • Antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein may be provided in pharmaceutically acceptable compositions as known in the art or as described herein.
  • a summary of the ways in which the antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein may be used therapeutically includes binding Therapeutic proteins locally or systemically in the body or by direct cytotoxicity of the antibody, e.g. as mediated by complement (CDC) or by effector cells (ADCC). Some of these approaches are described in more detail below.
  • CDC complement
  • ADCC effector cells
  • the antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein may be advantageously utilized in combination with other monoclonal or chimeric antibodies, or with lymphokines or hematopoietic growth factors (such as, e.g., IL-2, IL-3 and EL-7), for example, which serve to increase the number or activity of effector cells which interact with the antibodies.
  • lymphokines or hematopoietic growth factors such as, e.g., IL-2, IL-3 and EL-7
  • the antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein may be administered alone or in combination with other-types of -treatments (e.g., radiation therapy, chemotherapy, ⁇ hormonal therapy, immunotherapy and anti-tumor agents). Generally, administration of products of a species origin or species reactivity (in the case of antibodies) that is the same species as that of the patient is preferred. Thus, in a preferred embodiment, human antibodies, fragments derivatives, analogs, or nucleic acids, are administered to a human patient for therapy or prophylaxis.
  • other-types of -treatments e.g., radiation therapy, chemotherapy, ⁇ hormonal therapy, immunotherapy and anti-tumor agents.
  • administration of products of a species origin or species reactivity in the case of antibodies
  • human antibodies, fragments derivatives, analogs, or nucleic acids are administered to a human patient for therapy or prophylaxis.
  • binding affinities include dissociation constants or Kd's less than 5 X 10 ⁇ 2 M, 10 "1 M, 5 X 10 '3 M, 10 '3 M, 5 X 10 "4 M, 10 "4 M.
  • More preferred binding affinities include those with a dissociation constant or Kd less than 5 X 10 "5 M, 10 "5 M, 5 X 10 "6 M, IC 6 M, 5 X 10 "7 M, I 0 7 M, 5 X 10 "8 M or 10 "8 M.
  • binding affinities include those with a dissociation constant or Kd less than 5 X 10 "9 M, 1(T 9 M, 5 X 1(T 10 M, 10 "10 M, 5 X 10 "n M, 1(T 11 M, 5 X 10 " 12 M, 10 "12 M, 5 X lO "13 M, lO "13 M, 5 X 10 "14 M, lO "14 M, 5 X 10 "15 M, or 10 "15 M.
  • nucleic acids comprising sequences encoding antibodies that bind therapeutic proteins or albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein are administered to treat, inhibit or prevent a disease or disorder associated with aberrant expression and/or activity of a Therapeutic protein, by way of gene therapy.
  • Gene therapy refers to therapy performed by the administration to a subject of an expressed or expressible nucleic acid.
  • the nucleic acids produce their encoded protein that mediates a therapeutic effect.
  • the compounds or pharmaceutical compositions of the invention are preferably tested in vitro, and then in vivo for the desired therapeutic or prophylactic activity, prior to use in humans.
  • in vitro assays to demonstrate the therapeutic or prophylactic utility of a compound or pharmaceutical composition include, the effect of a compound on a cell line or a patient tissue sample.
  • the effect of the compound or composition on the cell line and/or tissue sample can be determined utilizing techniques known to those of skill in the art including, but not limited to, rosette formation assays and cell lysis assays.
  • in vitro assays which can be used to determine whether administration of a specific compound is indicated, include in vitro cell culture assays in which a patient tissue sample is grown in culture, and exposed to or otherwise administered a compound, and the effect of such compound upon the tissue sample is observed.
  • the invention provides methods of treatment, inhibition and prophylaxis by administration to a subject of an effective amount of a compound or pharmaceutical composition of the invention.
  • the compound is substantially purified (e.g., substantially free from substances that limit its effect or produce undesired side-effects).
  • the subject is preferably an animal, including but not limited to animals such as cows, pigs, horses, chickens, cats, dogs, etc., and is preferably a mammal, and most preferably human.
  • Formulations and methods of administration that can be employed when the compound comprises a nucleic acid or an immunoglobulin are described above; additional appropriate formulations and routes of administration can be selected from among those described herein below.
  • Various delivery systems are known and can be used to administer a compound of the invention, e.g., encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of expressing the compound, receptor-mediated endocytosis (see, e.g., Wu and Wu, J. Biol. Chem. 262:4429-4432 (1987)), construction of a nucleic acid as part of a retroviral or other vector, etc.
  • Methods of introduction include but are not limited to intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, and oral routes.
  • the compounds or compositions may be administered by any convenient route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents. Administration can be systemic or local.
  • intraventricular injection may be facilitated by an intraventricular catheter, for example, attached to a reservoir, such as an Ommaya reservoir.
  • Pulmonary administration can also be employed, e.g., by use of an inhaler or nebulizer, and formulation with an aerosolizing agent.
  • a protein, including an antibody, of the invention care must be taken to use materials to which the protein does not absorb.
  • the compound or composition can be delivered in a vesicle, in particular a liposome (see Langer, Science 249:1527-1533 (1990); Treat et al., in Liposomes in the Therapy of Infectious Disease and Cancer, Lopez-Berestein and Fidler (eds.),- Liss, New York, pp. 353- 365 (1989); Lopez-Berestein, ibid., pp. 317-327; see generally ibid.)
  • the compound or composition can be delivered in a controlled release system.
  • a pump may be used (see Langer, supra; Sefton, CRC Crit. Ref. Biomed. Eng. 14:201 (1987); Buchwald et al., Surgery 88:507 (1980); Saudek et al., N. Engl. J. Med. 321:574 (1989)).
  • polymeric materials can be used (see Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Pres., Boca Raton, Florida (1974); Controlled Drug Bioavailability, Drug Product Design and Performance, Smolen and Ball (eds.), Wiley, New York (1984); Ranger and Peppas, J., Macromol. Sci. Rev. Macromol. Chem. 23:61 (1983); see also Levy et al., Science 228:190 (1985); During et al., Ann. Neurol. 25:351 (1989); Howard et al., J.Neurosurg. 71:105 (1989)).
  • a controlled release system can be placed in proximity of the therapeutic target, e.g., the brain, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol. 2, pp. 115-138 (1984)).
  • Other controlled release systems are discussed in the review by Langer (Science 249: 1527-1533 (1990)).
  • the nucleic acid can be administered in vivo to promote expression of its encoded protein, by constructing it as part of an appropriate nucleic acid expression vector and administering it so that it becomes intracellular, e.g., by use of a retroviral vector (see U.S. Patent No.
  • a nucleic acid can be introduced intracellularly and incorporated within host cell DNA for expression, by homologous recombination.
  • compositions comprise a therapeutically effective amount of a compound, and a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans.
  • carrier refers to a diluent, adjuvant, excipient, or vehicle with which the therapeutic is administered.
  • Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like.
  • Water is a preferred carrier when the pharmaceutical composition is administered intravenously.
  • Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions.
  • Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like.
  • the composition if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents.
  • compositions can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations and the like.
  • the composition can be formulated as a suppository, with traditional binders and carriers such as triglycerides.
  • Oral formulation can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc. Examples of suitable pharmaceutical carriers are described in "Remington's Pharmaceutical Sciences" by E. W. Martin.
  • Such compositions will contain a therapeutically effective amount of the compound, preferably in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the patient.
  • the formulation should suit the mode of administration.
  • the composition is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous administration to human beings.
  • compositions for intravenous administration are solutions in sterile isotonic aqueous buffer.
  • the composition may also include a solubilizing agent and a local anesthetic such as lignocaine to ease pain at the site of the injection.
  • the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent.
  • composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline.
  • an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
  • the compounds of the invention can be formulated as neutral or salt forms.
  • Pharmaceutically acceptable salts include those formed with anions such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, etc., and those formed with cations such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2-ethylamino ethanol, histidine, procaine, etc.
  • the amount of the compound of the invention which will be effective in the treatment, inhibition and prevention of a disease or disorder associated with aberrant expression and/or activity of a Therapeutic protein can be determined by standard clinical techniques, hi addition, in vitro assays may optionally be employed to help identify optimal dosage ranges.
  • the precise dose to be employed in the formulation will also depend on the route of administration, and the seriousness of the disease or disorder, and should be decided according to the judgment of the practitioner and each patient's circumstances. Effective doses may be extrapolated from dose-response curves derived from in vitro or animal model test systems.
  • the dosage administered to a patient is typically 0.1 mg/kg to 100 mg/kg of the patient's body weight.
  • the dosage administered to a patient is between 0.1 mg/kg and 20 mg/kg of the . patient's Jjody. weight, more preferably 1 mg/kg to-10 mg/kg of the- patient's body weight.
  • human antibodies have a longer half-life within the human body than antibodies from other species due to the immune response to the foreign polypeptides. Thus, lower dosages of human antibodies and less frequent administration is often possible.
  • the dosage and frequency of administration of antibodies of the invention may be reduced by enhancing uptake and tissue penetration (e.g., into the brain) of the antibodies by modifications such as, for example, lipidation.
  • Labeled antibodies and derivatives and analogs thereof that bind a Therapeutic protein (or fragment or variant thereof) can be used for diagnostic purposes to detect, diagnose, or monitor diseases, disorders, and/or conditions associated with the aberrant expression and/or activity of Therapeutic protein.
  • the invention provides for the detection of aberrant expression of a Therapeutic protein, comprising (a) assaying the expression of the Therapeutic protein in cells or body fluid of an individual using one or more antibodies specific to the polypeptide interest and (b) comparing the level of gene expression with a standard gene expression level, whereby an increase or decrease in the assayed Therapeutic protein expression level compared to the standard expression level is indicative of aberrant expression.
  • the invention provides a diagnostic assay for diagnosing a disorder, comprising (a) assaying the expression of the Therapeutic protein in cells or body fluid of an individual using one or more antibodies specific to the Therapeutic protein or albumin fusion proteins comprising at least a fragment of variant of an antibody specific to a Therapeutic protein, and (b) comparing the level of gene expression with a standard gene expression level, whereby an increase or decrease in the assayed Therapeutic protein gene expression level compared to the standard expression level is indicative of a particular disorder.
  • the presence of a relatively high amount of transcript in biopsied tissue from an individual may indicate a predisposition for the development of the disease, or may provide a means for detecting the disease prior to the appearance of actual clinical symptoms. A more definitive diagnosis of this type may allow health professionals to employ preventative measures or aggressive treatment earlier thereby preventing the development or further progression of the cancer.
  • Antibodies of the invention or albumin fusion proteins comprising at least a fragment of variant of an antibody specific to a Therapeutic protein can be used to assay protein levels in a biological sample using classical immunohistological methods known to those of skill in the art (e.g., see Jalkanen et al., J. Cell. Biol. 101:976-985 (1985); Jalkanen et al., J. Cell . Biol. 105:3087-3096 (1987)).
  • Other antibody-based methods useful for detecting protein gene expression include immunoassays, such as the enzyme linked immunosorbent assay (ELISA) and the radioimmunoassay (RIA).
  • Suitable antibody assay labels include enzyme labels, such as, glucose oxidase; radioisotopes, such as iodine (1251, 1211), carbon (14C), sulfur (35S), tritium (3H), indium (112In), and technetium (99Tc); luminescent labels, such as luminol; and fluorescent labels, such as fluorescein and rhodamine, and biotin.
  • enzyme labels such as, glucose oxidase
  • radioisotopes such as iodine (1251, 1211), carbon (14C), sulfur (35S), tritium (3H), indium (112In), and technetium (99Tc)
  • luminescent labels such as luminol
  • fluorescent labels such as fluorescein and rhodamine, and biotin.
  • diagnosis comprises: a) administering (for example, parenterally, subcutaneously, or intraperitoneally) to a subject an effective amount of a labeled molecule which specifically binds to the polypeptide of interest; b) waiting for a time interval following the administering for permitting the labeled molecule to preferentially concentrate at sites in the subject where the Therapeutic protein is expressed (and for unbound labeled molecule to be cleared to background level); c) determining background level; and d) detecting the labeled molecule in the subject, such mat detection of labeled molecule above the background level indicates that the subject has a particular disease or disorder associated with aberrant expression of the therapeutic protein.
  • Background level can be determined by various methods including, comparing the amount of labeled molecule detected to a standard value previously determined for a particular system.
  • the size of the subject and the imaging system used will determine the quantity of imaging moiety needed to produce diagnostic images.
  • the quantity of radioactivity injected will normally range from about 5 to 20 millicuries of 99mTc.
  • the labeled antibody, antibody fragment, or albumin fusion protein comprising at least a fragment or variant of an antibody that binds a Therapeutic protein will then preferentially accumulate at the location of cells which contain the specific Therapeutic protein. In vivo tumor imaging is described in S.W.
  • the time interval following the administration for permitting the labeled molecule to preferentially concentrate at sites in the subject and for unbound labeled molecule to be cleared to background level is 6 to 48 hours or 6 to 24 hours or 6 to 12 hours. In another embodiment the time interval following administration is 5 to 20 days or 5 to 10 days.
  • monitoring of the disease or disorder is carried out by repeating the method for diagnosing the disease or disease, for example, one month after initial diagnosis, six months after initial diagnosis, one year after initial diagnosis, etc.
  • Presence of the labeled molecule can be detected in the patient using methods known in the art for in vivo scanning. These methods depend upon the type of label used. Skilled artisans will be _able to determine-the appropriate method for detecting a particular label; Methods and devices that may be used in the diagnostic methods of the invention include, but are not limited to, computed tomography (CT), whole body scan such as position emission tomography (PET), magnetic resonance imaging (MRI), and sonography.
  • CT computed tomography
  • PET position emission tomography
  • MRI magnetic resonance imaging
  • sonography sonography
  • the molecule is labeled with a radioisotope and is detected in the patient using a radiation responsive surgical instrument (Thurston et al., U.S. Patent No. 5,441,050).
  • the molecule is labeled with a fluorescent compound and is detected in the patient using a fluorescence responsive scanning instrument.
  • the molecule is labeled with a positron emitting metal and is detected in the patent using positron emission-tomography.
  • the molecule is labeled with a paramagnetic label and is detected in a patient using magnetic resonance imaging (MRI).
  • MRI magnetic resonance imaging
  • kits that can be used in the above methods.
  • a kit comprises an antibody, preferably a purified antibody, in one or more containers.
  • the kits of the present invention contain a substantially isolated polypeptide comprising an epitope which is specifically immunoreactive with an antibody included in the kit.
  • the kits of the present invention further comprise a control antibody which does not react with the polypeptide of interest.
  • kits of the present invention contain a means for detecting the binding of an antibody to a polypeptide of interest (e.g., the antibody may be conjugated to a detectable substrate such as a fluorescent compound, an enzymatic substrate, a radioactive compound or a luminescent compound, or a second antibody which recognizes the first antibody may be conjugated to a detectable substrate).
  • a detectable substrate such as a fluorescent compound, an enzymatic substrate, a radioactive compound or a luminescent compound, or a second antibody which recognizes the first antibody may be conjugated to a detectable substrate.
  • the kit is a diagnostic kit for use in screening serum containing antibodies specific against proliferative and/or cancerous polynucleotides and polypeptides.
  • a kit may include a control antibody that does not react with the polypeptide of interest.
  • a kit may include a substantially isolated polypeptide antigen comprising an epitope which is specifically immunoreactive with at least one anti-polypeptide antigen antibody.
  • a kit includes means for detecting the binding of said antibody to the antigen (e.g., the antibody may be conjugated to a fluorescent compound such as fluorescein or rhodamine which can be detected by flow cytometry).
  • the kit may include a recombinantly produced or chemically synthesized polypeptide antigen.
  • the polypeptide antigen of the kit may also be attached to a solid support.
  • the detecting means of the above-described kit includes a solid support to which said polypeptide antigen is attached.
  • a kit may also include a non-attached reporter-labeled anti-human antibody.
  • binding of the antibody to the polypeptide antigen can be detected by binding of the said reporter-labeled antibody.
  • the invention includes a diagnostic kit for use in screening serum containing antigens of the polypeptide of the invention.
  • the diagnostic kit includes a substantially isolated antibody specifically immunoreaetive with polypeptide or polynucleotide antigens, and means for detecting the binding of the polynucleotide or polypeptide antigen to the antibody.
  • the antibody is attached to a solid support.
  • the antibody may be a monoclonal antibody.
  • the detecting means of the kit may include a second, labeled monoclonal antibody. Alternatively, or in addition, the detecting means may include a labeled, competing antigen.
  • test serum is reacted with a solid phase reagent having a surface-bound antigen obtained by the methods of the present invention.
  • the reagent After binding with specific antigen antibody to the reagent and removing unbound serum components by washing, the reagent is reacted with reporter-labeled anti-human antibody to bind reporter to the reagent in proportion to the amount of bound anti-antigen antibody on the solid support.
  • the reagent is again washed to remove unbound labeled antibody, and the amount of reporter associated with the reagent is determined.
  • the reporter is an enzyme which is detected by incubating the solid phase in the presence of a suitable fluorometric, luminescent or colorimetric substrate (Sigma, St. Louis, MO).
  • the solid surface reagent in the above assay is prepared by known techniques for attaching protein material to solid support material, such as polymeric beads, dip sticks, 96-well plate or filter material. These attachment methods generally include non-specific adsorption of the protein to the support or covalent attachment of the protein, typically through a free amine group, to a chemically reactive group on the solid support, such as an activated carboxyl, hydroxyl, or aldehyde group. Alternatively, streptavidin coated plates can be used in conjunction with biotinylated antigen(s).
  • the invention provides an assay system or kit for carrying out this diagnostic method.
  • the kit generally includes a support with surface-bound recombinant antigens, and a reporter-labeled anti-human antibody for detecting surface-bound anti-antigen antibody.
  • albumin fusion protein refers to a protein formed by the fusion of at least one molecule of albumin (or a fragment or variant thereof) to at least one molecule of a Therapeutic protein (or fragment or variant thereof).
  • An albumin fusion protein of the invention comprises at least a fragment or variant of a Therapeutic protein and at least a fragment or variant of human serum albumin, which are associated with one another, preferably by genetic fusion (i.e., the albumin fusion .
  • the Therapeutic protein is generatedby translation of a nucleic acid in which a polynucleotide encoding all or a portion of a Therapeutic protein is joined in-frame with a polynucleotide encoding all or a portion of albumin) or to one another.
  • the Therapeutic protein and albumin protein, once part of the albumin fusion protein, may each be referred to as a "portion", "region” or "moiety" of the albumin fusion protein.
  • the invention provides an albumin fusion protein encoded by a polynucleotide or albumin fusion construct described in Table 1 or Table 2. Polynucleotides encoding these albumin fusion proteins are also encompassed by the invention.
  • Preferred albumin fusion proteins of the invention include, but are not limited to, albumin fusion proteins encoded by a nucleic acid molecule comprising, or alternatively consisting of, a polynucleotide encoding at least one molecule of albumin (or a fragment or variant thereof) joined in frame to at least one polynucleotide encoding at least one molecule of a Therapeutic protein (or fragment or variant thereof); a nucleic acid molecule comprising, or alternatively consisting of, a polynucleotide encoding at least one molecule of albumin (or a fragment or variant thereof) joined in frame to at least one polynucleotide encoding at least one molecule of a Therapeutic protein (or fragment or variant thereof) generated as described in Table 1, Table 2 or in the Examples; or a nucleic acid molecule comprising, or alternatively consisting of, a polynucleotide encoding at least one molecule of albumin (or a fragment or variant thereof) joined
  • the invention provides an albumin fusion protein comprising, or alternatively consisting of, a Therapeutic protein (e.g., as described in Table 1) and a serum albumin protein.
  • the invention provides an albumin fusion protein comprising, or alternatively consisting of, a biologically active and/or therapeutically active fragment of a Therapeutic protein and a serum albumin protein.
  • the invention provides an albumin fusion protein comprising, or alternatively consisting of, a biologically active and/or therapeutically active variant of a Therapeutic protein and a serum albumin protein.
  • the serum albumin protein component of the albumin fusion protein is the mature portion of serum albumin.
  • BoSmr ⁇ tsrt ⁇ 'e ' invention provides an albumin fusion protein comprising, or alternatively consisting of, a Therapeutic protein, and a biologically active and/or therapeutically active fragment of serum albumin.
  • the invention provides an albumin fusion protein comprising, or alternatively consisting of, a Therapeutic protein and a biologically active and/or therapeutically active variant of serum albumin.
  • the Therapeutic protein portion of the albumin fusion protein is the mature portion of the Therapeutic protein.
  • the invention provides an albumin fusion protein comprising, or alternatively consisting of, a biologically active and/or therapeutically active fragment or variant of a Therapeutic protein and a biologically active and/or therapeutically active fragment or variant of serum albumin.
  • the invention provides an albumin fusion protein comprising, or alternatively consisting of, the mature portion of a Therapeutic protein and the mature portion of serum albumin.
  • the albumin fusion protein comprises HA as the N-terminal portion, and a Therapeutic protein as the C-terminal portion.
  • a Therapeutic protein as the C-terminal portion may also be used.
  • the albumin fusion protein has a Therapeutic protein fused to both the N-terminus and the C-terminus of albumin.
  • the Therapeutic proteins fused at the N- and C- termini are the same Therapeutic proteins.
  • the Therapeutic proteins fused at the N- and C- termini are different Therapeutic proteins.
  • the Therapeutic proteins fused at the N- and C- termini are different Therapeutic proteins which may be used to treat or prevent the same or a related disease, disorder, or condition (e.g. as listed in the "Preferred Indication Y" column of Table 1).
  • the Therapeutic proteins fused at the N- and C- termini are different Therapeutic proteins which may be used to treat, ameliorate, or prevent diseases or disorders (e.g. as listed in the "Preferred Indication Y" column of Table 1) which are known in the art to commonly occur in patients simultaneously, concurrently, or consecutively, or which commonly occur in patients in association with one another.
  • diseases or disorders e.g. as listed in the "Preferred Indication Y" column of Table 1
  • Albumin fusion proteins of the invention encompass proteins containing one, two, three, four, or more molecules of a given Therapeutic protein X or variant thereof fused to the N- or C- terminus of an albumin fusion protein of the invention, and/or to the N- and/or C- terminus of albumin or variant thereof.
  • Molecules of a given Therapeutic protein X or variants thereof may be in any number of orientations, including, but not limited to, a 'head to head' orientation (e.g., wherein the N-terminus of one molecule of a Therapeutic protein X is fused to the N-terminus of another molecule of the Therapeutic protein X), or a 'head to tail' orientation (e.g., wherein the C-terminus of one molecule of a Therapeutic protein X is fused to the N-terminus of another molecule of Therapeutic protein X).
  • a 'head to head' orientation e.g., wherein the N-terminus of one molecule of a Therapeutic protein X is fused to the N-terminus of another molecule of the Therapeutic protein X
  • a 'head to tail' orientation e.g., wherein the C-terminus of one molecule of a Therapeutic protein X is fused to the N-terminus of another molecule of Therapeutic protein X.
  • one, two, three, or more tandemly oriented Therapeutic protein X polypeptides are fused to the N- or C- terminus of an albumin fusion protein of the invention, and/or to the N- and/or C- terminus of albumin or variant thereof.
  • Albumin fusion proteins of the invention further encompass proteins containing one, two, three, four, or more -molecules of a given " Therapeutic protein X or variant thereof fused to the N- or C- terminus of an albumin fusion protein of the invention, and/or to the N- and/or C- terminus of albumin or variant thereof, wherein the molecules are joined through peptide linkers.
  • Examples include those peptide linkers described in U.S. Pat. No. 5,073,627 (hereby incorporated by reference).
  • Albumin fusion proteins comprising multiple Therapeutic protein X polypeptides separated by peptide linkers may be produced using conventional recombinant DNA technology. Linkers are particularly important when fusing a small peptide to the large HSA molecule.
  • the peptide itself can be a linker by fusing tandem copies of the peptide or other known linkers can be used. Constructs that incorporate linkers are described in Table 2 or are apparent when examining SEQ ID NO: Y.
  • albumin fusion proteins of the invention may also be produced by fusing a Therapeutic protein X or variants thereof to the N- terminal and/or C-terminal of albumin or variants thereof in such a way as to allow the formation of intramolecular and/or intermolecular multimeric forms.
  • albumin fusion proteins may be in monomeric or multimeric forms (i.e., dimers, trimers, tetramers and. higher multimers).
  • the Therapeutic protein portion of an albumin fusion protein may be in monomeric form or multimeric form (i.e., dimers, trimers, tetramers and higher multimers).
  • the Therapeutic protein portion of an albumin fusion protein is in multimeric form (i.e., dimers, trimers, tetramers and higher multimers), and the albumin protein portion is in monomeric form.
  • albumin fusion proteins of the invention may also be produced by inserting the Therapeutic protein or peptide of interest (e.g., a Therapeutic protein X as disclosed in Table 1, or an antibody that binds a Therapeutic protein or a fragment or variant thereof) into an internal region of HA. For instance, within the protein sequence of the HA molecule a number of loops or turns exist between the end and beginning of ⁇ -helices, which are stabilized by disulphide bonds.
  • Therapeutic protein or peptide of interest e.g., a Therapeutic protein X as disclosed in Table 1, or an antibody that binds a Therapeutic protein or a fragment or variant thereof
  • the loops as determined from the crystal structure of HA (PDB identifiers 1AO6, 1BJ5, IBKE, IBMO, 1E7E to 1E7I and IUOR) for the most part extend away from the body of the molecule. These loops are useful for the insertion, or internal fusion, of therapeutically active peptides, particularly those requiring a secondary structure to be functional, or Therapeutic proteins, to essentially generate an albumin molecule with specific biological activity.
  • peptides or polypeptides are inserted into the VaI 5 4-Asn61, GIn 170-AIa 176, and/or Lys 5 ⁇ 0-Thr 5 66 loops of mature human albumin (SEQ ID NO:1).
  • Peptides to be inserted may be derived from either phage display or synthetic peptide libraries screened for specific biological activity or from the active portions of a molecule with the desired function. Additionally, random peptide libraries may be generated within particular loops or by insertions of randomized peptides into particular loops of the HA molecule and in which all possible combinations of amino acids are represented.
  • Such library(s) could be generated on HA or domain fragments of HA by one of the following methods: randomized mutation of amino acids within one or more peptide loops of HA or HA domain fragments. Either one, more or all the residues within a loop could be mutated in this manner; replacement of, or insertion into one or more loops of HA or HA domain fragments (i.e., internal fusion) of a randomized peptide(s) of length X n (where X is an amino acid and n is the number of residues;
  • N-, C- or N- and C- terminal peptide/protein fusions in addition to (a) and/or (b).
  • the HA or HA domain fragment may also be made multifunctional by grafting the peptides derived from different screens of different loops against different targets into the same HA or HA domain fragment.
  • peptides inserted into a loop of human serum albumin are peptide fragments or peptide variants of the Therapeutic proteins disclosed in Table 1. More particularly, the invention encompasses albumin fusion proteins which comprise peptide fragments or peptide variants at least 7 at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 20, at least 25, at least 30, at least 35, or at least 40 amino acids in length inserted into a loop of human serum albumin.
  • the invention also encompasses albumin fusion proteins which comprise peptide fragments or peptide variants at least 7 at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 20, at least 25, at least 30, at least 35, or at least 40 amino acids fused to the N-terminus of human serum albumin.
  • the invention also encompasses albumin fusion proteins which comprise peptide fragments or peptide variants at least 7 at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 20, at least 25, at least 30, at least 35, or at least 40 amino acids fused to the C- terminus of human serum albumin.
  • short peptides described in Table 1 and 2 e.g., Therapeutic Y
  • Therapeutic Y can be inserted into the albumin loops.
  • the albumin fusion proteins of the invention may have one HA-derived region and one Therapeutic protein-derived region. Multiple regions of each protein, however, may be used to make an albumin fusion protein of the invention. Similarly, more than one Therapeutic protein may be used to make an albumin fusion protein of the invention. For instance, a Therapeutic protein may be fused to both the N- and C- terminal ends of the HA. In such a configuration, the Therapeutic protein portions may be Jhe same or different Therapeutic protein molecules.
  • the - structure of bifunctional albumin fusion proteins may be represented as: X-HA-Y or Y-HA-X.
  • an anti-BLySTM scFv-HA-IFN ⁇ -2b fusion may be prepared to modulate the immune response to IFN ⁇ -2b by anti-BLySTM scFv.
  • An alternative is making a bi (or even multi) functional dose of HA-fusions e.g. HA-IFN ⁇ -2b fusion mixed with HA-anti-BLySTM scFv fusion or other HA-fusions in various ratio's depending on function, half-life etc.
  • Bi- or multi-functional albumin fusion proteins may also be prepared to target the Therapeutic protein portion of a fusion to a target organ or cell type via protein or peptide at the opposite terminus of HA.
  • the peptides could be obtained by screening libraries constructed as fusions to the N-, C- or N- and C- termini of HA, or domain fragment of HA, of typically 6, 8, 12, 20 or 25 or X n (where X is an amino acid (aa) and n equals the number of residues) randomized amino acids, and in which all possible combinations of amino acids were represented.
  • X is an amino acid (aa) and n equals the number of residues) randomized amino acids, and in which all possible combinations of amino acids were represented.
  • a particular advantage of this approach is that the peptides may be selected in situ on the HA molecule and the properties of the peptide would therefore be as selected for rather than, potentially, modified as might be the case for a peptide derived by any other method then being attached to HA.
  • the albumin fusion proteins of the invention may include a linker peptide between the fused portions to provide greater physical separation between the moieties and thus maximize the accessibility of the Therapeutic protein portion, for instance, for binding to its cognate receptor.
  • the linker peptide may consist of amino acids such that it is flexible or more rigid.
  • the linker sequence may be cleavable by a protease or chemically to yield the growth hormone related moiety.
  • the protease is one which is produced naturally by the host, for example the S. cerevisiae protease kex2 or equivalent proteases.
  • the albumin fusion proteins of the invention may have the following formula R1-L-R2; R2-L-R1; or Rl- L-R2-L-R1, wherein Rl is at least one Therapeutic protein, peptide or polypeptide sequence, and not necessarily the same Therapeutic protein, L is a linker and R2 is a serum albumin sequence.
  • albumin fusion proteins of the invention comprising a Therapeutic protein have a higher plasma stability compared to the plasma stability of the same Therapeutic protein when not fused to albumin.
  • Plasma stability typically refers to the time period T*iei®lIdl ⁇ k. ⁇ >!rd ⁇ n, " Jk administered in vivo and carried into the bloodstream and when the therapeutic protein is degraded and cleared from the bloodstream, into an organ, such as the kidney or liver, that ultimately clears the Therapeutic protein from the body.
  • Plasma stability is calculated in terms of the half-life of the Therapeutic protein in the bloodstream.
  • the half-life of the Therapeutic protein in the bloodstream can be readily determined by common assays known in the art.
  • Albumin fusion proteins of the invention comprising a Therapeutic protein have extended shelf life compared to the shelf life the same Therapeutic protein when not fused to albumin. Shelf-life typically refers to the time period over which the therapeutic activity of a Therapeutic protein in solution or in some other storage formulation, is stable without undue loss of therapeutic activity. Many of the Therapeutic proteins are highly labile in their unfused state. As described below, the typical shelf-life of these Therapeutic proteins is markedly prolonged upon incorporation into the albumin fusion protein of the invention.
  • Albumin fusion proteins of the invention with "prolonged” or “extended” shelf-life exhibit greater therapeutic activity relative to a standard that has been subjected to the same storage and handling conditions.
  • the standard may be the unfused full-length Therapeutic protein.
  • the Therapeutic protein portion of the albumin fusion protein is an analog, a variant, or is otherwise altered or does not include the complete sequence for that protein, the prolongation of therapeutic activity may alternatively be compared to the unfused equivalent of that analog, variant, altered peptide or incomplete sequence.
  • an albumin fusion protein of the invention may retain greater than about 10Q% of the therapeutic activity, or greater than about 10 5 %, 110%, 120%, 130%, 1 5 O% or 200% of the therapeutic activity of a standard when subjected to the same storage and handling conditions as the standard when compared at a given time point.
  • Shelf-life may also be assessed in terms of therapeutic activity remaining after storage, normalized to therapeutic activity when storage began.
  • Albumin fusion proteins of the invention with prolonged or extended shelf-life as exhibited by prolonged or extended therapeutic activity may retain greater than about 50% of the therapeutic activity, about ⁇ %, 70%, 80%, or 90% or more of the therapeutic activity of the equivalent unfused Therapeutic protein when subjected to the same conditions.
  • the albumin fusion proteins of the invention may be produced as recombinant molecules by secretion from yeast, a microorganism such as a bacterium, or a human or animal cell line.
  • the polypeptide is secreted from the host cells.
  • a particular embodiment of the invention comprises a DNA construct encoding a signal sequence effective for directing secretion in yeast, particularly a yeast-derived signal sequence (especially one which is homologous to the yeast host), and the fused molecule of the first aspect of the invention, there being no yeast-derived pro sequence between the signal and the mature polypeptide.
  • the Saccharomyces cerevisiae invertase signal is a preferred example of a yeast-derived signal sequence.
  • Conjugates of the kind prepared by Poznansky et ah, (FEBS Lett. 239:18 (1988)), in which separately-prepared polypeptides are joined by chemical cross-linking, are not contemplated. _ _ . . .
  • the present invention also includes a cell, preferably a yeast cell transformed to express an albumin fusion protein of the invention.
  • a cell preferably a yeast cell transformed to express an albumin fusion protein of the invention.
  • the present invention also contemplates a culture of those cells, preferably a monoclonal (clonally homogeneous) culture, or a culture derived from a monoclonal culture, in a nutrient medium. If the polypeptide is secreted, the medium will contain the polypeptide, with the cells, or without the cells if they have been filtered or centrifuged away.
  • Many expression systems are known and may be used, including bacteria (for example E.
  • yeasts for example Saccharomyces cerevisiae, Kluyveromyces lactis and Pichia pastoris
  • filamentous fungi for example Aspergillus
  • plant cells animal cells and insect cells.
  • D88 [leu2-3, Ieu2-122, canl, pral, ubc4] is a derivative of parent strain AH22his* (also known as DBl; see, e.g., Sleep et a Biotechnology 8:42-46 (1990)).
  • the strain contains a Ieu2 mutation which allows for auxotropic selection of 2 micron-based plasmids that contain the LEU2 gene.
  • D88 also exhibits a derepression of PRBl in glucose excess.
  • the PRBl promoter is normally controlled by two checkpoints that monitor glucose levels and growth stage.
  • the promoter is activated in wild type yeast upon glucose depletion and entry into stationary phase.
  • Strain D88 exhibits the repression by glucose but maintains the induction upon entry into stationary phase.
  • the PRAl gene encodes a yeast vacuolar protease, YscA endoprotease A, that is localized in the ER.
  • the UBC4 gene is in the ubiquitination pathway and is involved in targeting short lived and abnormal proteins for ubiquitin dependant degradation. Isolation of this ubc4 mutation was found to increase the copy number of an expression plasmid in the cell and cause an increased level of expression of a desired protein expressed from the plasmid (see, e.g., International Publication No. WO99/00504, hereby incorporated in its entirety by reference herein).
  • DXYl a derivative of D88, has the following genotype: [leu2-3, Ieu2-122, canl, pral, vbc4, ura3::yap3 ⁇ .
  • this strain also has a knockout of the YAP3 protease. This protease causes cleavage of mostly di-basic residues (RR, RK, KR, KK) but can also promote cleavage at single basic residues in proteins. Isolation of this yap3 mutation resulted in higher levels of full length HSA production (see, e.g., U.S. Patent No. 5,965,386 and Kerry-Williams et al., Yeast 14:161-169 (1998), hereby incorporated in their entireties by reference herein).
  • BXPlO has the following genotype: leu2-3, Ieu2-122, canl, pral, ubc4, ura3, yap3::URA3, Iys2, hspl50::LYS2, pmtl::URA3.
  • this strain also has a knockout of the PMTl gene and the HSP150 gene.
  • the PMTl gene is a member of the evolutionarily conserved family of dolichyl-phosphate-D-mannose protein O-mannosyltransferases (Pmts). The transmembrane topology of
  • Pmtlp suggests that it is an integral membrane protein of the endoplasmic reticulum with a role in O-Iinked glycosylation.
  • This mutation serves to reduce/eliminate O-linked glycosylation of HSA fusions (see, e.g., International Publication No. WO00/44772, hereby incorporated in its entirety by reference herein).
  • Studies revealed that the Hspl50 protein is inefficiently separated from rHA by ion exchange chromatography.
  • the mutation in the HSPl 5 O gene removes a potential contaminant that has proven difficult to remove by standard purification techniques. See, e.g., U.S. Patent No.
  • the desired protein is produced in conventional ways, for example from a coding sequence inserted in the host chromosome or on a free plasmid.
  • the yeasts are transformed with a coding sequence for the desired protein in any of the usual ways, for example electroporation. Methods for transformation of yeast by electroporation are disclosed in Becker & Guarente (1990) Methods Enzymol. 194, 182.
  • Successfully transformed cells i.e., cells that contain a DNA construct of the present invention, can be identified by well known techniques. For example, cells resulting from the introduction of an expression construct can be grown to produce the desired polypeptide. Cells can be harvested and lysed and their DNA content examined for the presence of the DNA using a method such as that described by Southern (1975)
  • the presence of the protein in the supernatant can be detected using antibodies.
  • Useful yeast plasmid vectors include pRS403-406 and pRS413-416 and are generally available from Stratagene Cloning Systems, La
  • Plasmids pRS403, pRS404, pRS405 and pRS406 are Yeast Integrating plasmids (Yips) and incorporate the yeast selectable markers HIS3, 7RPl, LEU2 and URA3.
  • Plasmids pRS413-416 are Yeast Centromere plasmids (Ycps).
  • Preferred vectors for making albumin fusion proteins for expression in yeast include pPPC000 5 , pScCHSA, pScNHSA, and pC4:HSA which are described in detail in Example 1.
  • Figure 2 shows a map of the pPPC0005 plasmid that can be used as the base vector into which polynucleotides encoding Therapeutic proteins may be cloned to form HA-fusions. It contains a PRBl S. cerevisiae promoter (PRBIp), a Fusion leader sequence (FL), DNA encoding HA (rHA) and an ADHl S. cerevisiae terminator sequence.
  • PRBIp PRBl S. cerevisiae promoter
  • FL Fusion leader sequence
  • rHA DNA encoding HA
  • ADHl S. cerevisiae terminator sequence ADHl S. cerevisiae terminator sequence.
  • sequence of the fusion leader sequence consists of the first 19 amino acids of the signal peptide of human serum albumin (SEQ ID NO-.3) and the last five amino acids of the mating factor alpha 1 promoter (SLDKR, see EP-A-387319 which is hereby incorporated by reference in its entirety).
  • Another vector useful for expressing an albumin fusion protein in yeast the pSAC35 vector which is described in
  • a yeast promoter that can be used to express the albumin fusion protein is the MET25 promoter. _See, for example, Dominik-Mumburg,
  • the Met25 promoter is 383 bases long (bases -382 to -1) and the genes expressed by this promoter are also known as Metl5, Metl7, and YLR303W.
  • a preferred embodiment uses the sequence below, where, at the 5' end of the sequence below, the Not 1 site used in the cloning is underlined and at the 3' end, the ATG start codon is underlined:
  • Additional promoters that can be used to express the albumin fusion protein in yeast include the following: a) the cbhl promoter:
  • CACAGAAACCCAATAGTCAACCGCGGACTGGCATC (SEQ ID NO: 11 5 ) d) a cysD promoter from Aspergillus nidulans having the sequence: AGATCTGGTTCCTGAGTACATCTACCGATGCGCCTCGA GTTCAGGGTCCTATTTGGACCGCTAGAAATAGACTCTGCTCGATTTGTTTCCATTATTCACGCAATTACGATAGTATTTGGC
  • a variety of methods have been developed to operably link DNA to vectors via complementary cohesive termini. For instance, complementary homopolymer tracts can be added to the DNA segment to be inserted to the vector DNA. The vector and DNA segment are then joined by hydrogen bonding between the complementary homopolymeric tails to form recombinant DNA molecules.
  • Synthetic linkers containing one or more restriction sites provide an alternative method of joining the DNA segment to vectors.
  • the DNA segment generated by endonuclease restriction digestion, is treated with bacteriophage T4 DNA polymerase or E. coli DNA polymerase I, enzymes that remove protruding, gamma-single-stranded termini with their 3' 5 '-exonucIeolytio activities, and fill in recessed 3'-ends with their polymerizing activities.
  • a desirable way to modify the DNA in accordance with the invention is to use the polymerase chain reaction as disclosed by Saiki et al. (1988) Science 239, 487-491.
  • the DNA to be enzymatically amplified is flanked by two specific oligonucleotide primers which themselves become incorporated into the amplified DNA
  • the specific primers may contain restriction endonuclease recognition sites which can be used for cloning into expression vectors using methods known in the art.
  • Exemplary genera of yeast contemplated to be useful in the practice of the present invention as hosts for expressing the albumin fusion proteins are Pichia (Hansenula), Saccharomyces, Kluyveromyces, Candida, Torulopsis, Torulaspora, Schtosaccharomyces, Citeromyces, Pachysolen, D ⁇ baromyces, Metschitnikowia, Rhodosporidium, Leucosporidium, Botryoascus, Sporidiobolus, Endomycopsis, and the like.
  • Preferred genera are those selected from the group consisting of Saccharomyces, Schtosaccharomyces, Kluyveromyces, Pichia and Torulaspora. Examples of Saccharomyces spp. are S. cerevisiae, S. italiciis and S. rouxii.
  • Examples of Kluyveromyces spp. are K. fragilis, K. lactis and K. marxianus.
  • a suitable Torulaspora species is T. delbrueckii.
  • Examples of Pichia (Hansenula) spp. are P. angusta (formerly H. polymorphs), P. anomala (formerly H. anomala) and P. pastoris.
  • Methods for the transformation of S. cerevisiae are taught generally in EP 251 744, EP 258 067 and WO 90/01063, all of which are incorporated herein by reference.
  • Preferred exemplary species of Saccharomyces include S. cerevisiae, S. italicus, S.
  • Preferred exemplary species of Kluyveromyces include K. fragilis and K. lactis.
  • Preferred exemplary species of Hansenula include H. polymorpha (now Pichia angusta), H. anomala (now Pichia anomala), and Pichia capsulata. Additional preferred exemplary species of Pichia include P. pastoris.
  • Preferred exemplary species of Aspergillus include A. niger and A. nidulans.
  • Preferred exemplary species of Yarrowia include Y. lipolytica. Many preferred yeast species are available from the ATCC.
  • yeast species are available from the ATCC and are useful in the expression of albumin fusion proteins: Saccharomyces cerevisiae Hansen, teleomorph strain BY4743 yap3 mutant (ATCC Accession No. 4022731); Saccharomyces cerevisiae Hansen, teleomorph strain BY4743 hspl50 mutant (ATCC Accession No. 4021266); Saccharomyces cerevisiae Hansen, teleomorph strain BY4743 pmtl mutant (ATCC Accession No. 4023792); Saccharomyces cerevisiae Hansen, teleomorph (ATCC Accession No ⁇ .
  • Suitable promoters for S. cerevisiae include those associated with the PGKI gene, GALl or GALlO genes, CYCL PHO5, TRPI, ADHL ADH2, the genes for glyceraldehyde-3-phosphate dehydrogenase, hexokinase, pyruvate decarboxylase, phosphofructokinase, triose phosphate isomerase, phosphoghicose isomerase, glucokinase, alpha-mating factor pheromone, [a mating factor pheromone], the PRBI promoter, the GUT2 promoter, the GPDI promoter, and hybrid promoters involving hybrids of parts of 5' regulatory regions with parts of 5' regulatory regions of other promoters or with upstream activation sites (e.g. the promoter of EP-A-2 5 8 067).
  • Convenient regulatable promoters for use in Schizosaccharomyces pombe are the thiamine-repressible promoter from the nmt gene as described by Maundrell (1990,) J. Biol. Chem. 265, 10857-10864 and the glucose repressible jbpl gene promoter as described by Hoffman & Winston (1990) Genetics 124, 807-816.
  • Pichia expression kits are commercially available from Livitrogen BV, Leek, Netherlands, and Invitrogen Corp., San Diego, California.
  • Suitable promoters include AOXI and A0X2.
  • Gleeson et al. (1986) J. Gen. Microbiol. 132, 3459-3465 include information on Hansenula vectors and transformation, suitable promoters being MOXl and FMDl; whilst EP 361 991, Fleer et al. (1991) and other- publications from Rhone-Poulenc Rorer teach how to express foreign proteins in Kluyveromyces spp., a suitable promoter being PGKI.
  • the transcription termination signal is preferably the 3' flanking sequence of a eukaryotic gene which contains proper signals for transcription termination and polyadenylation.
  • Suitable 3' flanking sequences may, for example, be those of the gene naturally linked to the expression control sequence used, i.e. may correspond to the promoter. Alternatively, they may be different in which case the termination signal of the S. cerevisiae ADHI gene is preferred.
  • ⁇ " LM " 1[033OjUI IMkttfred ' alBli'iM fli ⁇ WprStein may be initially expressed with a secretion leader sequence, which may be any leader effective in the yeast chosen.
  • Leaders useful in yeast include any of the following: a) the MPIF-I signal sequence (e.g., amino acids 1-21 of GenBank Accession number AAB51134) MKVSVAALSCLMLVTALGSQA (SEQ ID N ⁇ :6) b) the stanniocalcin signal sequence (MLQNSAVLLLLVISASA, SEQ ID NO:7) c) the pre-pro region of the HSA signal sequence (e.g., MKWVTFISLLFLFS S AYSRGVFRR, SEQ ID NO:8) d) the pre region of the HSA signal sequence (e.g., MKWVTFISLLFLFSSAYS, SEQ ID NO:9) or variants thereof, such as, for example, MKWVSFISLLFLFSSAYS, (SEQ ID NO: 10) e) the invertase signal sequence (e.g., MLLQAFLFLLAGFAAKISA, SEQ ID NO: 11) f) the yeast mating factor alpha signal sequence (e.g.,
  • K. lactis killer toxin leader sequence h) a hybrid signal sequence (e.g., MKWVSFISLLFLFSSAYSRSLEKR, SEQ ID N ⁇ :13)
  • a hybrid signal sequence e.g., MKWVSFISLLFLFSSAYSRSLEKR, SEQ ID N ⁇ :13
  • HSA/MF ⁇ -1 hybrid signal sequence also known as HSA/kex2
  • MKWVSFISLLFLFSSAYSRSLDKR also known as HSA/kex2
  • lactis killer/ MF ⁇ -1 fusion leader sequence e.g., MNIFYIFLFLLSFVQGSLDKR, SEQ ID NO: 15
  • the Immunoglobulin Ig signal sequence e.g., MGWSCIILFLV ATATGVHS, SEQ ID N ⁇ :16
  • Fibulin B precursor signal sequence e.g., MERAAPSRRVPLPLLLLGGLALLAAGVDA, SEQ ID NO: 17
  • the clusterin precursor signal sequence e.g., MMKTLLLFVGLLLTWESGQVLG, SEQ ID N ⁇ :18
  • the insulin-like growth factor-binding protein 4 signal sequence e.g., MLPLCLV AALLLAAGPGPSLG, SEQ ID NO: 19
  • variants of the pre-pro-region of the HSA signal sequence such as, for example,
  • Modified HSA (S 14) leader also known as modified HSA #65
  • MKWVTFISLLFLFSGVSG SEQ ID NO:27
  • Modified HSA (G14) leader - MKWVTFISLLFLFGGVSG (SEQ ID NO:28), or MKWVTFISLLFLFGGVLGDLHKS (SEQ ID NO:29) p) a consensus signal sequence (MPTWAWWLFLVLLLALWAPARG, SEQ ID N ⁇ :30) q) acid phosphatase (PH05) leader (e.g., MFKSWYSILAASLANA SEQ ID N ⁇ :31) r) the pre-sequence of MFoz-1 s) the pre-sequence of 0 glucanase (BGL2) t) killer toxin leader u) the presequence of killer toxin v) k. lactis killer toxin prepro (29 amino acids; 16 amino acids of pre and 13 amino acids of pro)
  • MNIFYIFLFLLSFVQGLEHTHRRGSLDKR (SEQ ID NO:32) w) 5 1 . diastaticus glucoarnylase Il secretion leader sequence x) S. carlsbergensis ⁇ -galactosidase (MELl) secretion leader sequence y) Candida glitcoamylase leader sequence z)
  • the hybrid leaders disclosed in EP-A-387 319 (herin incorporated by reference) aa) the gp67 signal sequence (in conjunction with baculoviral expression systems) (e.g., amino acids 1-19 of GenBank Accession Number
  • AAA72759 or bb) the natural leader of the therapeutic protein X; cc) S. cerevisiae invertase (SUC2) leader, as disclosed in JP 62-096086 (granted as 911036516, herein incorporate by reference); or dd) Inulinase- MKLAYSLLLPLAGVSASVINYKR (SEQ ID NO:33). ee) A modified TA57 propeptide leader variant # 1 -
  • the present invention also relates to vectors containing a polynucleotide encoding an albumin fusion protein of the present invention, host cells, and the production of albumin fusion proteins by synthetic and recombinant techniques.
  • the vector may be, for example, a phage, plasmid, viral, or retroviral vector.
  • Retroviral vectors may be replication competent or replication defective. In the latter case, viral propagation generally will occur only in complementing host cells.
  • the polynucleotides encoding albumin fusion proteins of the invention may be joined to a vector containing a selectable marker for propagation in a host.
  • a plasmid vector is introduced in a precipitate, such as a calcium phosphate precipitate, or in a complex with a charged lipid. If the vector is a virus, it may be packaged in vitro using an appropriate packaging cell line and then transduced into host cells.
  • the polynucleotide insert should be operatively linked to an appropriate promoter, such as the phage lambda PL promoter, the E.
  • the expression constructs will further contain sites for transcription initiation, termination, and, in the transcribed region, a ribosome binding site for translation.
  • the coding portion of the transcripts expressed by the constructs will preferably include a translation initiating codon at the beginning and a termination codon (UAA, UGA or UAG) appropriately positioned at the end of the polypeptide to be translated.
  • the expression vectors will preferably include at least one selectable marker.
  • markers include dihydrofolate reductase, G418, glutamine synthase, or neomycin resistance for eukaryotic cell culture, and tetracycline, kanamycin or ampicillin resistance genes for culturing in E. coli and other bacteria.
  • Representative examples of appropriate hosts include, but are not limited to, bacterial cells, such as E. coli, Streptomyces and Salmonella typhimurium cells; fungal cells, such as yeast cells (e.g., Saccharomyces cerevisiae or Pichia pastoris (ATCC Accession No.
  • insect cells such as Drosophila S2 and Spodoptera Sf9 cells
  • animal cells such as CHO, COS, NSO, 293, and Bowes melanoma cells
  • plant cells Appropriate culture mediums and conditions for the above-described host cells are known in the art.
  • vectors preferred for use in bacteria include pQE70, pQE60 and pQE-9, available from QIAGEN, Inc.; pBluescript vectors, Phagescript vectors, pNH8A, pNH16a, pNH18A, pNH46A, available from Stratagene Cloning Systems, Inc.; and ptrc99a, pKK223-3, pKK233-3, pDR540, pRIT 5 available from Pharmacia Biotech, Inc.
  • preferred eukaryotic vectors are pWLNEO, pSV2CAT, pOG44, pXTl and pSG available from Stratagene; and pSVK3, pBPV, pMSG and pSVL available from Pharmacia.
  • Preferred expression vectors for use in.yeast systems ⁇ _ include, but are not limited to pYES2, pYDl, pTEFl/Zeo, pYES2/GS, pPICZ, pGAPZ, pGAPZalph, pPIC9, pPIC3.
  • polynucleotides encoding an albumin fusion protein of the invention may be fused to signal sequences which will direct the localization of a protein of the invention to particular compartments of a prokaryotic or eukaryotic cell and/or direct the secretion of a protein of the invention from a prokaryotic or eukaryotic cell.
  • signal sequences which will direct the localization of a protein of the invention to particular compartments of a prokaryotic or eukaryotic cell and/or direct the secretion of a protein of the invention from a prokaryotic or eukaryotic cell.
  • E. coli one may wish to direct the expression of the protein to the periplasmic space.
  • Examples of signal sequences or proteins (or fragments thereof) to which the albumin fusion proteins of the invention may be fused in order to direct the expression of the polypeptide to the periplasmic space of bacteria include, but are not limited to, the pelB signal sequence, the maltose binding protein (MBP) signal sequence, MBP, the ompA signal sequence, the signal sequence of the periplasmic E. coli heat- labile enterotoxin B-subunit, and the signal sequence of alkaline phosphatase.
  • MBP maltose binding protein
  • ompA the signal sequence of the periplasmic E. coli heat- labile enterotoxin B-subunit
  • alkaline phosphatase Several vectors are commercially available for the construction of fusion proteins which will direct the localization of a protein, such as the pMAL series of vectors (particularly the pMAL-p series) available from New England Biolabs.
  • polynucleotides albumin fusion proteins of the invention may be fused to the pelB pectate lyase signal sequence to increase the efficiency of expression and purification of such polypeptides in Gram-negative bacteria. See, U.S. Patent Nos. 5 ,576,195 and 5,846,818, the contents of which are herein incorporated by reference in their entireties.
  • Examples of signal peptides that may be fused to an albumin fusion protein of the invention in order to direct its secretion in mammalian cells include, but are not limited to: a) the MPIF-I signal sequence (e.g., amino acids 1-21 of GenBank Accession number AAB51134) MKVSVAALSCLMLVTALGSQA (SEQ IDNO:6) b) the stanniocalcin signal sequence (MLQNSAVLLLLVISASA, SEQ ID N ⁇ :7) c) the pre-pro region of the HSA signal sequence (e.g., MKWVTFISLLFLFSSAYSRGVFRR, SEQ ID N ⁇ :8) d) the pre region of the HSA signal sequence (e.g., MKWVTFISLLFLFSSAYS, SEQ ID N ⁇ :9) or variants thereof, such as, for example, MKWVSFISLLFLFSSAYS, (SEQ ID N ⁇ :10) e) the invertase signal sequence (e.g., MLLQAFL
  • SEQ ID NO: 12 g) K. lactis killer toxin leader sequence h) a hybrid signal sequence (e.g., MKWVSFISLLFLFSSAYSRSLEKR, SEQ ID NO: 13) i) an HSA/MF ⁇ -1 hybrid signal sequence (also known as HSA/kex2) (e.g., MKWVSFISLLFLFSSAYSRSLDKR, SEQ ID N ⁇ :14) j) a JST.
  • a hybrid signal sequence e.g., MKWVSFISLLFLFSSAYSRSLEKR, SEQ ID NO: 13
  • HSA/MF ⁇ -1 hybrid signal sequence also known as HSA/kex2
  • MKWVSFISLLFLFLFSSAYSRSLDKR SEQ ID N ⁇ :14
  • lactis killer/ MFa- 1 fusion leader sequence e.g., MNIFYIFLFLLSFVQGSLDKR, SEQ ID NO: 15
  • the Immunoglobulin Ig signal sequence e.g., MGWSCIILFLVATATGVHS, SEQ ID NO: 16
  • the Fibulin B precursor signal sequence e.g., MERAAPSRRVPLPLLLLGGLALLAAGVDA, SEQ ID NO: 17
  • the clusterin precursor signal sequence e.g., MMKTLLLFVGLLLTWESGQVLG, SEQ ID N0:18
  • the insulin-like growth factor-binding protein 4 signal sequence e.g., MLPLCLVAALLLAAGPGPSLG, SEQ ID NO: 19
  • variants of the pre-pro-region of the HSA signal sequence such as, for example,
  • Modified HSA (S 14) leader also known as modified HSA #65
  • MKWVTFISLLFLFSGVSG SEQ ID NO:27
  • Modified HSA (G14) leader - MKWVTFISLLFLFGGVSG (SEQ ID NO:28), or MKWVTFISLLFLFGGVLGDLHKS (SEQ ID NO:29) p) a consensus signal sequence (MPTWAWWLFLVLLLALWAPARG, SEQ ID N ⁇ :30) q) acid phosphatase (PH05) leader (e.g., MFKSWYSILAASLANA SEQ ID N ⁇ :31) r) the pre-sequence ofMFoz-1 s) the pre-sequence of 0 glucanase (BGL2) t) killer toxin leader u) the presequence of killer toxin _ __ _ _ _ . - - - v) k. lactis killer toxin prepro (29 amino acids; 16 amino acids of pre and 13 amino acids of pro)
  • MNIFYIFLFLLSFVQGLEHTHRRGSLDKR SEQ IDNO:32
  • the hybrid leaders disclosed in EP-A-387319 (herin incorporated by reference) aa) the gp67 signal sequence (in conjunction with baculoviral expression systems) (e.g., amino acids 1-19 of GenBank Accession Number
  • AAA72759 or bb) the natural leader of the therapeutic protein X; cc) S. cerevisiae invertase (SUC2) leader, as disclosed in JP 62-096086 (granted as 911036516, herein incorporate by reference); or dd) Inulinase- MKLAYSLLLPLAGVSASVINYKR (SEQ ID NO:33). ee) A modified TA57 propeptide leader variant # 1 -
  • the modified HSA/kex2 signal sequence (SEQ ID NO: 112) is fused to the amino terminus of an albumin fusion protein, including fusion proteins comprising albumin and a therapeutic protein as described herein, as well as albumin fusion proteins disclosed in WO93/15199; WO97/24445; WO03/60071; WO03/59934; and PCT/US04/01369, each of which are incorporated herein by reference in their entireties.
  • the modified HSA/kex2 signal sequence is based on the HSA/kex2 signal sequence (SEQ ID NO: 14) disclosed, e.g., in Sleep et ' !L» llal 1 .
  • modified HSA/kex2 leader sequence disclosed herein contains a non-conservative amino acid substitution (Arg to GIy) at residue 19 of the parent signal peptide.
  • the modified HSA/kex2 signal peptide has been found to produce unexpectedly better expression yield and/or better cleavage efficiency of albumin fusion proteins when expressed in yeast than the unmodified HSA/kex2 signal sequence. Variants of the modified HSA/kex2 signal peptide are also encompassed by the invention.
  • GIy residue at position 19 of SEQ ID NO: 112 may be substituted with a Pro residue.
  • Other conservative substitution variants of the modified HSA/kex2 signal sequence are also contemplated.
  • Nucleic acids encoding the modified HSA/kex2 signal sequence of SEQ ID NO: 112, as well as conservative substitution variants thereof, are also encompassed by the invention.
  • Vectors which use glutamine synthase (GS) or DHFR as the selectable markers can be amplified in the presence of the drugs methionine sulphoximine or methotrexate, respectively.
  • An advantage of glutamine synthase based vectors are the availability of cell lines (e.g., the murine myeloma cell line, NSO) which are glutamine synthase negative.
  • Glutamine synthase expression systems can also function in glutamine synthase expressing cells (e.g., Chinese Hamster Ovary (CHO) cells) by providing additional inhibitor to prevent the functioning of the endogenous gene.
  • glutamine synthase expression system and components thereof are detailed in PCT publications: WO87/04462; WO86/05807; WO89/01036; WO89/10404; and WO91/06657, which are hereby incorporated in their entireties by reference herein. Additionally, glutamine synthase expression vectors can be obtained from Lonza Biologies, Inc. (Portsmouth, NH). Expression and production of monoclonal antibodies using a GS expression system in murine myeloma cells is described in Bebbington et al, Bio/technology 10:169(1992) and in Biblia and Robinson Biotechnol. Prog. 11:1 (1995) which are herein incorporated by reference.
  • the present invention also relates to host cells containing the above-described vector constructs described herein, and additionally encompasses host cells containing nucleotide sequences of the invention that are operably associated with one or more heterologous control regions (e.g., promoter and/or enhancer) using techniques known of in the art.
  • the host cell can be a higher eukaryotic cell, such as a mammalian cell (e.g., a human derived cell), or a lower eukaryotic cell, such as a yeast cell, or the host cell can be a prokaryotic cell, such as a bacterial cell.
  • a host strain may be chosen which modulates the expression of the inserted gene sequences, or modifies and processes the gene product in the specific fashion desired.
  • Expression from certain promoters can be elevated in the presence of certain inducers; thus expression of the genetically engineered polypeptide may be controlled.
  • different host cells have characteristics and specific mechanisms for the translational and post- translational processing and modification (e.g., phosphorylation, cleavage) of proteins. Appropriate cell lines can be chosen to ensure the desired modifications and processing of the foreign protein expressed.
  • nucleic acids and nucleic acid constructs of the invention into the host cell can be effected by calcium phosphate transfection, DEAE-dextran mediated transfection, cationic lipid-mediated transfection, electroporation, transduction, infection, or other methods. Such methods are described in many standard laboratory manuals, such as Davis et al ⁇ Basic Methods . In Molecular Biology _(19_86). . It js.. _ specifically contemplated that the polypeptides of the present invention may in fact be expressed by a host cell lacking a recombinant vector.
  • the invention also encompasses primary, secondary, and immortalized host cells of vertebrate origin, particularly mammalian origin, that have been engineered to delete or replace endogenous genetic material (e.g., the coding sequence corresponding to a Therapeutic protein may be replaced with an albumin fusion protein corresponding to the Therapeutic protein), and/or to include genetic material (e.g., heterologous polynucleotide sequences such as for example, an albumin fusion protein of the invention corresponding to the Therapeutic protein may be included).
  • the genetic material operably associated with the endogenous polynucleotide may activate, alter, and/or amplify endogenous polynucleotides.
  • heterologous polynucleotides e.g., polynucleotides encoding an albumin protein, or a fragment or variant thereof
  • heterologous control regions e.g., promoter and/or enhancer
  • endogenous polynucleotide sequences encoding a Therapeutic protein via homologous recombination (see, e.g., US Patent Number 5,641,670, issued June 24, 1997; International Publication Number WO 96/29411; International Publication Number WO 94/12650; Koller et al., Proc. Natl. Acad. Sd. USA ⁇ 6:8932-8935 (1989); and Zijlstra et al., Nature 342:435-438 (1989), the disclosures of each of which are incorporated by reference in their entireties).
  • Albumin fusion proteins of the invention can be recovered and purified from recombinant cell cultures by well-known methods including ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography, hydrophobic charge interaction chromatography and lectin chromatography. Most preferably, high performance liquid chromatography (“HPLC”) is employed for purification.
  • HPLC high performance liquid chromatography
  • the albumin fusion proteins of the invention are purified using Anion Exchange Chromatography including, but not limited to, chromatography on Q-sepharose, DEAE sepharose, poros HQ, poros DEAE, Toyopearl Q, Toyopearl QAE, Toyopearl DEAE, Resource/Source Q and DEAE, Fractogel Q and DEAE columns.
  • Anion Exchange Chromatography including, but not limited to, chromatography on Q-sepharose, DEAE sepharose, poros HQ, poros DEAE, Toyopearl Q, Toyopearl QAE, Toyopearl DEAE, Resource/Source Q and DEAE, Fractogel Q and DEAE columns.
  • the albumin fusion proteins of the invention are purified using Cation Exchange Chromatography including, but not limited to, SP-sepharose, CM sepharose, poros HS, poros CM, Toyopearl SP, Toyopearl CM, Resource/Source S and CM, Fractogel S and CM ioW'niMnitfeir'Silkival& ' a'Bd-amillara'bles.
  • Cation Exchange Chromatography including, but not limited to, SP-sepharose, CM sepharose, poros HS, poros CM, Toyopearl SP, Toyopearl CM, Resource/Source S and CM, Fractogel S and CM ioW'niMnitfeir'Silkival& ' a'Bd-amillara'bles.
  • the albumin fusion proteins of the invention are purified using Hydrophobic Interaction Chromatography including, but not limited to, Phenyl, Butyl, Methyl, Octyl, Hexyl-sepharose, poros Phenyl, Butyl, Methyl, Octyl, Hexyl, Toyopearl Phenyl, Butyl, Methyl, Octyl, Hexyl Resource/Source Phenyl, Butyl, Methyl, Octyl, Hexyl, Fractogel Phenyl, Butyl, Methyl, Octyl, Hexyl columns and their equivalents and comparables.
  • Hydrophobic Interaction Chromatography including, but not limited to, Phenyl, Butyl, Methyl, Octyl, Hexyl-sepharose, poros Phenyl, Butyl, Methyl, Octyl, Hexyl, Toyopearl Phenyl, Butyl
  • albumin fusion proteins of the invention are purified using Size Exclusion Chromatography including, but not limited to, sepharose SlOO, S200, S300, superdex resin columns and their equivalents and comparables.
  • albumin fusion proteins of the invention are purified using Affinity Chromatography including, but not limited to, Mimetic Dye affinity, peptide affinity and antibody affinity columns that are selective for either the HSA or the "fusion target" molecules.
  • albumin fusion proteins of the invention are purified using one or more Chromatography methods listed above.
  • albumin fusion proteins of the invention are purified using one or more of the following Chromatography columns, Q sepharose FF column, SP Sepharose FF column, Q Sepharose High Performance Column, Blue Sepharose FF column , Blue Column, Phenyl Sepharose FF column, DEAE Sepharose FF, or Methyl Column.
  • albumin fusion proteins of the invention may be purified using the process described in PCT International Publication WO 00/44772 which is herein incorporated by reference in its entirety.
  • One of skill in the art could easily modify the process described therein for use in the purification of albumin fusion proteins of the invention.
  • Albumin fusion proteins of the present invention may be recovered from: products of chemical synthetic procedures; and products produced by recombinant techniques from a prokaryotic or eukaryotic host, including, for example, bacterial, yeast, higher plant, insect, and mammalian cells. Depending upon the host employed in a recombinant production procedure, the polypeptides of the present invention may be glycosylated or may be non-glycosylated. In addition, albumin fusion proteins of the invention may also include an initial modified methionine residue, in some cases as a result of host-mediated processes.
  • N-terminal methionine encoded by the translation initiation codon generally is removed with high efficiency from any protein after translation in all eukaryotic cells. While the N-terminal methionine on most proteins also is efficiently removed in most prokaryotes, for some proteins, this prokaryotic removal process is inefficient, depending on the nature of the amino acid to which the N-terminal methionine is covalently linked.
  • the yeast Pichia pasioris is used to express albumin fusion proteins of the invention in a eukaryotic system.
  • Pichia pastoris is a methylotrophic yeast which can metabolize methanol as its sole carbon source.
  • a main step in the methanol metabolization pathway is the oxidation of methanol to formaldehyde using O 2 . This reaction is catalyzed by the enzyme alcohol oxidase.
  • Pichia pastoris In order to metabolize methanol as its sole carbon source, Pichia pastoris must generate high levels of alcohol oxidase due, in part, to the relatively low affinity of alcohol oxidase for O2.
  • alcohol oxidase produced from the AOXl gene comprises up to approximately 30% of the total soluble protein in Pichia pastoris. See Ellis, S.B., et al, MoI. Cell. Biol 5:1111-21 (1985); Koutz, PJ, et a!., Yeast 5:167-77 (1989); Tschopp, J.F., et al., Niicl. Acids Res. 15:3859-76 (1987).
  • a heterologous coding sequence such as, for example, a polynucleotide of the present invention, under the transcriptional regulation of all or part of the AOXl regulatory sequence is expressed at exceptionally high levels in Pichia yeast grown in the presence of methanol.
  • the plasmid vector pPIC9K is used to express DNA encoding an albumin fusion protein of the invention, as set forth herein, in a Pichea yeast system essentially as described in '"Pichia Protocols: Methods in Molecular Biology," D.R. Higgins and J. Cregg, eds. The Humana Press, Totowa, NJ, 1998.
  • This expression vector allows expression and secretion of a polypeptide of the invention by virtue of the strong AOXl promoter linked to the Pichia pastoris alkaline phosphatase (PHO) secretory signal peptide (i.e., leader) located upstream of a multiple cloning site.
  • PHO alkaline phosphatase
  • yeast vectors could be used in place of pPIC9K, such as, pYES2, pYDl, pTEFl/Zeo, pYES2/GS, pPICZ, pGAPZ, pGAPZalpha, pPIC9, pPIC3.5, pH!L-D2, pHIL-Sl, pPIC3.5K, and PAO815, as one skilled in the art would readily appreciate, as long as the proposed expression construct provides appropriately located signals for transcription, translation, secretion (if desired), and the like, including an in-frame AUG as required.
  • high-level expression of a heterologous coding sequence such as, for example, a polynucleotide encoding an albumin fusion protein of the present invention, may be achieved by cloning the heterologous polynucleotide of the invention into an expression vector such as, for example, pGAPZ or pGAPZalpha, and growing the yeast culture in the absence of methanol.
  • albumin fusion proteins of the invention can be chemically synthesized using techniques known in the art (e.g., see Creighton, 1983, Proteins: Structures and Molecular Principles, W.H. Freeman & Co., N. Y., and Hunkapiller et al., Nature, 310:105-111 (1984)).
  • a polypeptide corresponding to a fragment of a polypeptide can be synthesized by use of a peptide synthesizer.
  • nonclassical amino acids or chemical amino acid analogs can be introduced as a substitution or addition into the polypeptide sequence.
  • Non-classical amino acids include, but are not limited to, to the D-isomers of the common amino acids, 2,4-diaminobutyric acid, a-amino isobutyric abidj Am ⁇ nBbm ⁇ ffi'a ⁇ ' idjIv ⁇ bujl ⁇ 'Mnfe ⁇ yric acid, g-Abu, e-Ahx, 6 -amino hexanoic acid, Aib, 2-amino isobutyric acid, 3-amino propionic acid, ornithine, norleucine, norvaline, hydroxyproline, sarcosine, citrulline, homocitrulline, cysteic acid, t-butylglycine, t-butylalanine, phenylglyci ⁇ e, cyclohexylalanine, b-alanine, fluoro-amino acids, designer amino acids such as b-methyl amino acids, Ca
  • the invention encompasses albumin fusion proteins of the present invention which are differentially modified during or after translation, e.g., by glycosylation, acetylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to an antibody molecule or other cellular ligand, etc. Any of numerous chemical modifications may be carried out by known techniques, including but not limited, to specific chemical cleavage by cyanogen bromide, trypsin, chymotrypsin, papain, V8 protease, NaBHU; acetylation, formylation, oxidation, reduction; metabolic synthesis in the presence of tunicamycin; etc.
  • Additional post-translational modifications encompassed by the invention include, for example, e.g., N-linked or O-linked carbohydrate chains, processing of N-terminal or C-terminal ends), attachment of chemical moieties to the amino acid backbone, chemical modifications of N-linked or O-linked carbohydrate chains, and addition or deletion of an N-terminal methionine residue as a result of procaryotic host cell expression.
  • the albumin fusion proteins may also be modified with a detectable label, such as an enzymatic, fluorescent, isotopic or affinity label to allow for detection and isolation of the protein.
  • suitable enzymes include horseradish peroxidase, alkaline phosphatase, beta-galactosidase, or acetylcholinesterase; examples of suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin; examples of suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; an example of a luminescent material includes luminol; examples of bioluminescent materials include luciferase, luciferin, and aequorin; and examples of suitable radioactive material include iodine ( 121 1, 123 L, 125 1, 131 I), carbon ( 14 C), sulfur ( 35 S), tritium ( 3 H), indium ( 111 In, 112 In, 113m In, 115m In), technetium ( 99 Tc,
  • albumin fusion proteins of the present invention or fragments or variants thereof are attached to niacrocyclic chelators that associate with radiometal ions, including but not limited to, 177 Lu, 90 Y, 166 Ho, and 153 Sm, to polypeptides.
  • the radiometal ion associated with the macrocyclic chelators is 111 In.
  • the radiometal ion associated with the macrocyclic chelator is 90 Y.
  • the macrocyclic chelator is l,4,7,10-tetraazacyclododecane-N,N',N",N"'-tetraacetic acid (DOTA).
  • DOTA is attached to an antibody of the invention or fragment thereof via linker molecule.
  • linker molecules useful for conjugating DOTA to a polypeptide are commonly known in the art - see, for example, DeNardo et al., Clin Cancer Res. 4(10):2483-90 (1998); Peterson et al., Bioconjug. Chem. 10(4):553-7 (1999); and Zimmerman et al, Nucl. Med. Biol. 2 6 (8):943- 5 0 (1999); which are hereby incorporated by reference in their entirety.
  • albumin fusion proteins of the invention may be modified by either natural processes, such as post-translational processing, or by chemical modification techniques which are well known in the art. It will be appreciated that the same type of modification may be present in the same or varying degrees at several sites in a given polypeptide.
  • Polypeptides of the invention may be branched, for example, as a result of ubiquitination, and they may be cyclic, with or without branching. Cyclic, branched, and branched cyclic polypeptides may result from posttranslation natural processes or may be made by synthetic methods.
  • Modifications include acetylation, acylation, ADP-ribosylation, amidation, covalent attachment of flavin, covalent attachment of a heme moiety, covalent attachment of a nucleotide or nucleotide derivative, covalent attachment of a lipid or lipid derivative, covalent attachment of phosphotidylinositol, cross-linking, cyclization, disulfide bond formation, demethylation, formation of covalent cross-links, formation of cysteine, formation of pyroglutamate, formylation, gamma-carboxylation, glycosylation, GPI anchor formation, hydroxylation, iodination, methylation, myristylation, oxidation, pegylation, proteolytic processing, phosphorylation, prenylation, racemization, selenoylation, sulfation, transfer-RNA mediated addition of amino acids to proteins such as arginylation, and ubiquitination.
  • Albumin fusion proteins of the invention and antibodies that bind a Therapeutic protein or fragments or variants thereof can be fused to marker sequences, such as a peptide to facilitate purification.
  • the marker amino acid sequence is a hexa-histidine peptide, such as the tag provided in a pQE vector (QIAGEN, Inc., 9259 Eton Avenue, Chatsworth, CA, 91311), among others, many of which are commercially available.
  • hexa-histidine provides for convenient purification of the fusion protein.
  • an albumin fusion protein of the invention may be conjugated to a therapeutic moiety such as a cytotoxin, e.g., a cytostatic or cytocidal agent, a therapeutic agent or a radioactive metal ion, e.g., alpha-emitters such as, for example, 213Bi.
  • a cytotoxin or cytotoxic agent ' IL e.g., a cytostatic or cytocidal agent, a therapeutic agent or a radioactive metal ion, e.g., alpha-emitters such as, for example, 213Bi.
  • Examples include paclitaxol, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicin, doxorubicin, daunorubicin, dihydroxy anthracin dione, mitoxantrone, mithramycin, actinomycin D, 1-dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, and puromycin and analogs or homologs thereof.
  • Therapeutic agents include, but are not limited to, antimetabolites (e.g., methotrexate, 6-mercaptopurine, 6-thioguanine, cytarabine, S- fluorouracil decarbazine), alkylating agents (e.g., mechlorethamine, thioepa chlorambucil, melphalan, carmustine (BSNU) and lomustine (CCNU), cyclothosphamide, busulfan, dibromomannitol, streptozotocin, mitomycin C, and cis- dichlorodiamine platinum (II) (DDP) cisplatin), anthracyclines (e.g., daunorubicin (formerly daunomycin) and doxorubicin), antibiotics (e.g., dactinomycin (formerly actinomycin), bleomycin, mithramycin, and anthramycin (AMC)), and anti-mitotic agents (e.g.
  • the conjugates of the invention can be used for modifying a given biological response, the therapeutic agent or drug moiety is not to be construed as limited to classical chemical therapeutic agents.
  • the drug moiety may be a protein or polypeptide possessing a desired biological activity.
  • Such proteins may include, for example, a toxin such as abrin, ricin A, pseudomonas exotoxin, or diphtheria toxin; a protein such as tumor necrosis factor, alpha-interferon, ⁇ -interferon, nerve growth factor, platelet derived growth factor, tissue plasminogen activator, an apoptotic agent, e.g., TNF-alpha, TNF-beta, AIM I (See, International Publication No. WO 97/33899), AIM H (See, International Publication No. WO 97/34911), Fas Ligand (Takahashi et ah, Int.
  • a toxin such as abrin, ricin A, pseudomonas exotoxin, or diphtheria toxin
  • a protein such as tumor necrosis factor, alpha-interferon, ⁇ -interferon, nerve growth factor, platelet derived growth factor, tissue plasminogen activator, an
  • VEGI See, International Publication No. WO 99/231Q5
  • a thrombotic agent or an anti- angiogenic agent e.g., angiostatin or endostatin
  • biological response modifiers such as, for example, lymphokines, interleukin-1 ("IL-I”), interleukin-2 (“IL-2”), interleukin-6 (“IL-6”), granulocyte macrophage colony stimulating factor (“GM-CSF”), granulocyte colony stimulating factor (“G-CSF”), or other growth factors.
  • IL-I interleukin-1
  • IL-2 interleukin-2
  • IL-6 interleukin-6
  • GM-CSF granulocyte macrophage colony stimulating factor
  • G-CSF granulocyte colony stimulating factor
  • Albumin fusion proteins may also be attached to solid supports, which are particularly useful for immunoassays or purification of polypeptides that are bound by, that bind to, or associate with albumin fusion proteins of the invention.
  • solid supports include, but are not limited to, glass, cellulose, polyacrylamide, nylon, polystyrene, polyvinyl chloride or polypropylene.
  • Albumin fusion proteins with or without a therapeutic moiety conjugated to it, administered alone or in combination with cytotoxic factor(s) and/or cytokine(s) can be used as a therapeutic.
  • the albumin fusion protein of the invention comprises only the VH domain of an antibody that binds a Therapeutic protein
  • the albumin fusion protein of the invention comprises only the VL domain of an antibody that binds a Therapeutic protein
  • Some Therapeutic antibodies are bispecific antibodies, meaning the antibody that binds a Therapeutic protein is an artificial hybrid antibody having two different heavy ⁇ ight chain pairs and two different binding sites, to order to create an albumin fusion protein corresponding to that Therapeutic protein, it is possible to create an albumin fusion protein which has an scFv fragment fused to both the N- and C- terminus of the albumin protein moiety.
  • the scFv fused to the N-terminus of albumin would correspond to one of the heavy/light (VH/VL) pairs of the original antibody that binds a Therapeutic protein and the scFv fused to the C-terminus of albumin would correspond to the other heavy ⁇ ight (VH/VL) pair of the original antibody that binds a Therapeutic protein.
  • the chemical moieties for derivitization may be selected from water soluble polymers such as polyethylene glycol, ethylene glycol/propylene glycol copolymers, carboxymethylcellulose, dextran, polyvinyl alcohol and the like.
  • the albumin fusion proteins may be modified at random positions within the molecule, or at predetermined positions within the molecule and may include one, two, three or more attached chemical moieties.
  • the polymer may be of any molecular weight, and may be branched or unbranched.
  • the preferred molecular weight is between about IkDa and about 100 kDa (the term "about” indicating that in preparations of polyethylene glycol, some molecules will weigh more, some less, than the stated molecular weight) for ease in handling and manufacturing.
  • Other sizes may be used, depending on the desired therapeutic profile (e.g., the duration of sustained release desired, the effects, if any on biological activity, the ease in handling, the degree or lack of antigenicity and other known effects of the polyethylene glycol to a Therapeutic protein or analog).
  • the polyethylene glycol may have an average molecular weight of about 200, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, 10,000, 10,500, 11,000, 11,500, 12,000, 12,500, 13,000, 13,500, 14,000, 14,500, 15,000, 15,500, 16,000, 16,500, 17,000, 17,500, 18,000, 18,500, 19,000, 19,500, 20,000, 25,000, 30,000, 35,000, 40,000, 45,000, 50,000, 55,000, 60,000, 65,000, 70,000, 75,000, 80,000, 85,000, 90,000, 95,000, or 100,000 kDa.
  • the polyethylene glycol may have a branched structure. Branched polyethylene glycols are described, for example, in ⁇ • ⁇ " Biochem. Biotechnol. 56:59-12 (1996); Vorobjev et al, Nucleosides Nucleotides 75:2745-2750
  • polyethylene glycol molecules should be attached to the protein with consideration of effects on functional or antigenic domains of the protein.
  • attachment methods available to those skilled in the art, such as, for example, the method disclosed in EP 0 401 384 (coupling PEG to G-CSF), herein incorporated by reference; see also Malik et al., Exp. Hematol. 20:1028-1035 (1992), reporting pegylation of GM-CSF using tresyl chloride.
  • polyethylene glycol may be covalently bound through amino acid residues via reactive group, such as a free amino or carboxyl group.
  • Reactive groups are those to which an activated polyethylene glycol molecule may be bound.
  • the amino acid residues having a free amino group may include lysine residues and the N-terminal amino acid residues; those having a free carboxyl group may include aspartic acid residues glutamic acid residues and the C-terminal amino acid residue.
  • Sulfhydryl groups may also be used as a reactive group for attaching the polyethylene glycol molecules.
  • Preferred for therapeutic purposes is attachment at an amino group, such as attachment at the N-terminus or lysine group.
  • polyethylene glycol may be attached to proteins via linkage to any of a number of amino acid residues.
  • polyethylene glycol can be linked to proteins via covalent bonds to lysine, histidine, aspartic acid, glutamic acid, or cysteine residues.
  • One or more reaction chemistries may be employed to attach polyethylene glycol to specific amino acid residues (e.g., lysine, histidine, aspartic acid, glutamic acid, or cysteine) of the protein or to more than one type of amino acid residue (e.g., lysine, histidine, aspartic acid, glutamic acid, cysteine and combinations thereof) of the protein.
  • polyethylene glycol as an illustration of the present composition, one may select from a variety of polyethylene glycol molecules (by molecular weight, branching, etc.), the proportion of polyethylene glycol molecules to protein (polypeptide) molecules in the reaction mix, the type of pegylation reaction to be performed, and the method of obtaining the selected N-terminally pegylated protein.
  • the method of obtaining the N-terminally pegylated preparation i.e., separating this moiety from other monopegylated moieties if necessary
  • Selective proteins chemically modified at the N-terminus modification may be accomplished by reductive alkylation which exploits differential reactivity of different types of primary amino groups (lysine versus the N-terminal) available for derivatization in a particular protein. Under the appropriate reaction conditions, substantially selective derivatization of the protein at the N-terminus with a carbonyl group containing polymer is achieved.
  • pegylation of the albumin fusion proteins of the invention may be accomplished by any number of means.
  • polyethylene glycol may be attached to the albumin fusion protein either directly or by an intervening linker.
  • Linkerless systems for attaching polyethylene glycol to proteins are described in Delgado et al., Crit. Rev. Thera. Drug Carrier Sys. 9:249-304 (1992); Francis et al., Intern. L of Hematol. 68:1-18 (1998); U.S. Patent No. 4,002,531] U.S. , Patent No, 5,349,Q52; .WO.95/06058; and WO-98/32466, the disclosures of each of which are incorporated herein by reference.
  • One system for attaching polyethylene glycol directly to amino acid residues of proteins without an intervening linker employs rresylated MPEG, which is produced by the modification of monmethoxy polyethylene glycol (MPEG) using tresylchloride (CISO 2 CH2CF 3 ).
  • MPEG monmethoxy polyethylene glycol
  • tresylchloride CISO 2 CH2CF 3
  • polyethylene glycol is directly attached to amine groups of the protein.
  • the invention includes protein- polyethylene glycol conjugates produced by reacting proteins of the invention with a polyethylene glycol molecule having a 2,2,2-trifluoreothane sulphonyl group.
  • Polyethylene glycol can also be attached to proteins using a number of different intervening linkers.
  • U.S. Patent No. 5,612,460 discloses urethane linkers for connecting polyethylene glycol to proteins.
  • Protein-polyethylene glycol conjugates wherein the polyethylene glycol is attached to the protein by a linker can also be produced by reaction of proteins with compounds such as MPEG-succinimidylsuccinate, MPEG activated with l,l'-carbonyldiimidazole, MPEG- 2,4,5-trichloropenylcarbonate, MPEG-p-nitrophenolcarbonate, and various MPEG-succinate derivatives.
  • the number of polyethylene glycol moieties attached to each albumin fusion protein of the invention may also vary.
  • the pegylated proteins of the invention may be linked, on average, to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 15, 17, 20, or more polyethylene glycol molecules.
  • the average degree of substitution within ranges such as 1-3, 2-4, 3-5, 4-6, 5-7, 6-8, 7-9, 8-10, 9-11, 10- 12, 11-13, 12-14, 13-15, 14-16, 15-17, 16-18, 17-19, or 18-20 polyethylene glycol moieties per protein molecule. Methods for determining the degree of substitution are discussed, for example, in Delgado et al., Crit. Rev. Thera. Drug Carrier Sys. 9:249-304 (1992).
  • polypeptides of the invention can be recovered and purified from chemical synthesis and recombinant cell cultures by standard methods which include, but are not limited to, ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite ' "" 1 I""-
  • HPLC high performance liquid chromatography
  • Well known techniques for refolding protein may be employed to regenerate active conformation when the polypeptide is denatured during isolation and/or purification.
  • the presence and quantity of albumin fusion proteins of the invention may be determined using ELISA, a well known immunoassay known in the art.
  • ELISA protocol that would be useful for detecting/quantifying albumin fusion proteins of the invention, comprises the steps of coating an ELISA plate with an anti-human serum albumin antibody, blocking the plate to prevent non-specific binding, washing the ELISA plate, adding a solution containing the albumin fusion protein of the invention (at one or more different concentrations), adding a secondary anti- Therapeutic protein specific antibody coupled to a detectable label (as described herein or otherwise known in the art), and detecting the presence of the secondary antibody.
  • the ELISA plate might be coated with the anti-Therapeutic protein specific antibody and the labeled secondary reagent might be the anti-human albumin specific antibody.
  • polynucleotides of the present invention are useful to produce the albumin fusion proteins of the invention.
  • polynucleotides of the invention encoding albumin fusion proteins
  • Polynucleotides of the present invention are also useful in gene therapy.
  • One goal of gene therapy is to insert a normal gene into an organism having a defective gene, in an effort to correct the genetic defect.
  • the polynucleotides disclosed in the present invention offer a means of targeting such genetic defects in a highly accurate manner.
  • Another goal is to insert a new gene that was not present in the host genome, thereby producing a new trait in the host cell. Additional non-limiting examples of gene therapy methods encompassed by the present invention are more thoroughly described elsewhere herein (see, e.g., the sections labeled "Gene Therapy", and Examples 61 and 62).
  • polypeptides identified herein can be used in numerous ways. The following description should be considered exemplary and utilizes known techniques.
  • Albumin fusion proteins of the invention are useful to provide immunological probes for differential identification of the tissue(s) (e.g., immunohistochemistry assays such as, for example, ABC immunoperoxidase (Hsu et al., J. Histochem. Cytochem. 29: 5 77-580 (19Sl)) or cell type(s) (e.g., immunooytochemistry assays).
  • tissue(s) e.g., immunohistochemistry assays such as, for example, ABC immunoperoxidase (Hsu et al., J. Histochem. Cytochem. 29: 5 77-580 (19Sl)
  • cell type(s) e.g., immunooytochemistry assays.
  • Albumin fusion proteins can be used to assay levels of polypeptides in_a.biological.sample using classical immunohistological methods - known to those of skill in the art (e.g., see Jalkanen, et al., J. Cell. Biol. 101:976-985 (1985); Jalkanen, et al., J. Cell. Biol. 105:3087-3096 (1987)).
  • Other methods useful for detecting protein gene expression include immunoassays, such as the enzyme linked immunosorbent assay (ELISA) and the radioimmunoassay (RIA).
  • Suitable assay labels include enzyme labels, such as, glucose oxidase; radioisotopes, such as iodine ( 131 L 125 I, 123 L 121 I), carbon ( 14 C), sulfur ( 35 S), tritium ( 3 H), indium (" 5111 In, U3m In, " 2 In, “ 1 In), and technetium ( 99 Tc, 99m Tc), thallium ( 201 Ti), gallium ( 68 Ga, 67 Ga), palladium ( 103 Pd), molybdenum ( 99 Mo), xenon ( 133 Xe), fluorine ( 18 F), 153 Sm, " 7 Lu, 159 Gd, 149 Pm, 140 La, 175 Yb, 166 Ho, 90 Y, 47 Sc, 186 Re, 188 Re, 142 Pr, 105 Rh, 97 Ru; luminescent labels, such as luminol; and fluorescent labels, such as fluorescein and rhodamine, and biotin.
  • enzyme labels such
  • Albumin fusion proteins of the invention can also be detected in vivo by imaging.
  • Labels or markers for in vivo imaging of protein include those detectable by X-radiography, nuclear magnetic resonance (NMR) or electron spin relaxtion (ESR).
  • suitable labels include radioisotopes such as barium or cesium, which emit detectable radiation but are not overtly harmful to the subject.
  • suitable markers for NMR and ESR include those with a detectable characteristic spin, such as deuterium, which may be incorporated into the albumin fusion protein by labeling of nutrients given to a cell line expressing the albumin fusion protein of the invention.
  • An albumin fusion protein which has been labeled with an appropriate detectable imaging moiety, such as a radioisotope (for example, 131 L 112 In, 99 Tc, ( 131 1, 125 1, 123 1, 121 I), carbon ( 14 C), sulfur ( 35 S), tritium ( 3 H), indium ( 115m In, 113m In, 112 In, 111 In), and technetium ( 99 Tc, 99m Tc), thallium ( 201 Ti), gallium ( 68 Ga, 67 Ga), palladium ( 103 Pd), molybdenum ( 99 Mo), xenon ( 133 Xe), fluorine ( 18 F, 153 Sm, 177 Lu, 159 Gd, 149 Pm, 140 La, 175 Yb, 166 Ho, 90 Y, 47 Sc, 186 Re, 188 Re, 142 Pr, 105 Rh, 97 Ru), a radio-opaque substance, or a material detectable by nuclear magnetic resonance, is introduced (for example,
  • the size of the subject and the imaging system used will determine the quantity of imaging moiety needed to produce diagnostic images.
  • the quantity of radioactivity injected will normally range from about 5 to 20 millicuries of 99 Tc.
  • the labeled albumin fusion protein will then preferentially accumulate at locations in the body (e.g., organs, cells, extracellular spaces or matrices) where one or more receptors, ligands or substrates (corresponding to that of the Therapeutic protein used to make the albumin fusion protein of the invention) are located.
  • the labeled albumin fusion protein will then preferentially accumulate at the locations in the body (e.g., organs, cells, "" • " txfrac&fiiS' ⁇ Ms'O'r niStri'csS)- ' vtfheiffe ' tne polypeptides/epitopes corresponding to those bound by the Therapeutic antibody (used to make the albumin fusion protein of the invention) are located.
  • locations in the body e.g., organs, cells, "" • " txfrac&fiiS' ⁇ Ms'O'r niStri'csS)- ' vtfheiffe ' tne polypeptides/epitopes corresponding to those bound by the Therapeutic antibody (used to make the albumin fusion protein of the invention) are located.
  • In vivo tumor imaging is described in S.W.
  • the invention provides a method for the specific delivery of albumin fusion proteins of the invention to cells by administering albumin fusion proteins of the invention (e.g., polypeptides encoded by polynucleotides encoding albumin fusion proteins of the invention and/or antibodies) that are associated with heterologous polypeptides or nucleic acids.
  • the invention provides a method for delivering a Therapeutic protein into the targeted cell.
  • the invention provides a method for delivering a single stranded nucleic acid (e.g., antisense or ribozymes) or double stranded nucleic acid (e.g., DNA that can integrate into the cell's genome or replicate episomally and that can be transcribed) into the targeted cell.
  • a single stranded nucleic acid e.g., antisense or ribozymes
  • double stranded nucleic acid e.g., DNA that can integrate into the cell's genome or replicate episomally and that can be transcribed
  • the invention provides a method for the specific destruction of cells (e.g., the destruction of tumor cells) by administering albumin fusion proteins of the invention in association with toxins or cytotoxic prodrugs.
  • toxin is meant one or more compounds that bind and activate endogenous cytotoxic effector systems, radioisotopes, holotoxins, modified toxins, catalytic subunits of toxins, or any molecules or enzymes not normally present in or on the surface of a cell that under defined conditions cause the cell's death.
  • Toxins that may be used according to the methods of the invention include, but are not limited to, radioisotopes known in the art, compounds such as, for example, antibodies (or complement fixing containing portions thereof) that bind an inherent or induced endogenous cytotoxic effector system, thymidine kinase, endonuclease, RNAse, alpha toxin, ricin, abrin, Pseudomonas exotoxin A, diphtheria toxin, saporin, momordin, gelonin, pokeweed antiviral protein, alpha-sarcin and cholera toxin.
  • radioisotopes known in the art
  • compounds such as, for example, antibodies (or complement fixing containing portions thereof) that bind an inherent or induced endogenous cytotoxic effector system, thymidine kinase, endonuclease, RNAse, alpha toxin, ricin, abrin, Pseu
  • Toxin also includes a cytostatic or cytocidal agent, a therapeutic agent or a radioactive metal ion, e.g., alpha-emitters such as, for example, 213 Bi, or other radioisotopes such as, for example, 103 Pd, 133 Xe, 131 1, 68 Ge, 57 Co, 65 Zn, 85 Sr, 32 P, 35 S, 90 Y, 153 Sm, 153 Gd, 169 Yb, 51 Cr, 54 Mn, 75 Se, 113 Sn, 90 Yttrium, 117 Tm, ! ⁇ Rhenium, 166 Holmium, and ⁇ Rhenium; luminescent labels, such as luminol; and fluorescent labels, such as fluorescein and rhodamine, and biotin.
  • alpha-emitters such as, for example, 213 Bi
  • radioisotopes such as, for example, 103 Pd, 133 Xe, 131 1, 68 Ge,
  • the invention provides a method for the specific destruction of cells (e.g., the destruction of tumor cells) by administering polypeptides of the invention or antibodies of the invention in association with the radioisotope 90 Y.
  • the invention provides a method for the specific destruction of cells (e.g., the destruction of tumor cells) by administering polypeptides of the invention or antibodies of the invention in association with the radioisotope " 1 In.
  • the invention provides a method for the specific destruction of cells (e.g., the destruction of tumor cells) by administering polypeptides of the invention or antibodies of the invention in association with the radioisotope 131 I.
  • the invention provides a diagnostic method of a disorder, which involves (a) assaying the expression level of a certain polypeptide in cells or body fluid of an individual using an albumin fusion protein of the invention; and (b) comparing the assayed polypeptide expression level with a standard polypeptide expression level, whereby an increase or decrease in the assayed polypeptide expression level compared to the standard expression level is indicative of a disorder.
  • a diagnostic method of a disorder involves (a) assaying the expression level of a certain polypeptide in cells or body fluid of an individual using an albumin fusion protein of the invention; and (b) comparing the assayed polypeptide expression level with a standard polypeptide expression level, whereby an increase or decrease in the assayed polypeptide expression level compared to the standard expression level is indicative of a disorder.
  • the presence of a relatively high amount of transcript in biopsied tissue from an individual may indicate a predisposition for the development of the disease, or may provide a means for detecting
  • albumin fusion proteins of the present invention can be used to treat or prevent diseases or conditions such as, for example, neural disorders, immune system disorders, muscular disorders, reproductive disorders, gastrointestinal disorders, pulmonary disorders, cardiovascular disorders, renal disorders, proliferative disorders, and/or cancerous diseases and conditions.
  • patients can be administered a polypeptide of the present invention in an effort to replace absent or decreased levels of the polypeptide (e.g., insulin), to supplement absent or decreased levels of a different polypeptide (e.g., hemoglobin S for hemoglobin B, SOD, catalase, DNA repair proteins), to inhibit the activity of a polypeptide (e.g., an oncogene or tumor supressor), to activate the activity of a polypeptide (e.g., by binding to a receptor), to reduce the activity of a membrane bound receptor by competing with it for free ligand (e.g., soluble TNF receptors used in reducing inflammation), or to bring about a desired response (e.g., blood vessel growth inhibition, enhancement of the immune response to proliferative cells or tissues).
  • a polypeptide e.g., insulin
  • a different polypeptide e.g., hemoglobin S for hemoglobin B, SOD, catalase, DNA repair proteins
  • albumin fusion proteins comprising of at least a fragment or variant of a Therapeutic antibody can also be used to treat disease (as described supra, and elsewhere herein).
  • administration of an albumin fusion protein comprising of at least a fragment or variant of a Therapeutic antibody can bind, and/or neutralize the polypeptide to which the Therapeutic antibody used to make the albumin fusion IL M liraiei ⁇ pteMctfll ⁇ bfhd ⁇ han ⁇ or ⁇ fefflce ⁇ ttVerproduction of the polypeptide to which the Therapeutic antibody used to make the albumin fusion protein specifically binds.
  • an albumin fusion protein comprising of at least a fragment or variant of a Therapeutic antibody can activate the polypeptide to which the Therapeutic antibody used to make the albumin fusion protein specifically binds, by binding to the polypeptide bound to a membrane (receptor).
  • the albumin fusion proteins of the invention of the present invention can be used as molecular weight markers on SDS- PAGE gels or on molecular sieve gel filtration columns using methods well known to those of skill in the art.
  • Albumin fusion proteins of the invention can also be used to raise antibodies, which in turn may be used to measure protein expression of the Therapeutic protein, albumin protein, and/or the albumin fusion protein of the invention from a recombinant cell, as a way of assessing transformation of the host cell, or in a biological sample.
  • the albumin fusion proteins of the present invention can be used to test the biological activities described herein.
  • the compounds of the present invention are useful for diagnosis, treatment, prevention and/or prognosis of various disorders in mammals, preferably humans.
  • disorders include, but are not limited to, those described for each Therapeutic protein in the corresponding row of Table 1 and herein under the section headings "Immune Activity,” “Blood Related Disorders,” “Hyperproliferative Disorders,” “Renal Disorders,” “Cardiovascular Disorders,” “Respiratory Disorders,” “Anti-Angiogenesis Activity,” “Diseases at the Cellular Level,” “Wound Healing and Epithelial Cell Proliferation,” “Neural Activity and Neurological Diseases,” “Endocrine Disorders,” “Reproductive System Disorders,” “Infectious Disease,” “Regeneration,” and/or “Gastrointestinal Disorders,” infi-a.
  • substantially altered (increased or decreased) levels of gene expression can be detected in tissues, cells or bodily fluids (e.g., sera, plasma, urine, semen, synovial fluid or spinal fluid) taken from an individual having such a disorder, relative to a "standard" gene expression level, that is, the expression level in tissues or bodily fluids from an individual not having the disorder.
  • a diagnostic method useful during diagnosis of a disorder which involves measuring the expression level of the gene encoding a polypeptide in tissues, cells or body fluid from an individual and comparing the measured gene expression level with a standard gene expression level, whereby an increase or decrease in the gene expression level(s) compared to the standard is indicative of a disorder.
  • diagnostic assays may be performed in vivo or in vitro, such as, for example, on blood samples, biopsy tissue or autopsy tissue.
  • the present invention is also useful as a prognostic indicator, whereby patients exhibiting enhanced or depressed gene expression will experience a worse clinical outcome.
  • saying the expression level of the gene encoding a polypeptide is intended qualitatively or quantitatively measuring or estimating the level of a particular polypeptide (e.g. a polypeptide corresponding to a Therapeutic protein disclosed in Table 1) or the level of the mRNA encoding the polypeptide of the invention in a first biological sample either directly (e.g., by determining or estimating absolute protein level or mRNA level) or relatively (e.g., by comparing to the polypeptide level or mRNA level in a. second biological sample).
  • a particular polypeptide e.g. a polypeptide corresponding to a Therapeutic protein disclosed in Table 1
  • mRNA encoding the polypeptide of the invention in a first biological sample either directly (e.g., by determining or estimating absolute protein level or mRNA level) or relatively (e.g., by comparing to the polypeptide level or mRNA level in a. second biological sample).
  • the polypeptide expression level or mRNA level in the first biological sample is measured or estimated and compared to a standard polypeptide level or mRNA level, the standard being taken from a second biological sample obtained from an individual not having the disorder or being determined by averaging levels from a population of individuals not having the disorder.
  • a standard polypeptide level or mRNA level is known, it can be used repeatedly as a standard for comparison.
  • biological sample any biological sample obtained from an individual, cell line, tissue culture, or other source containing polypeptides of the invention (including portions thereof) or mRNA.
  • biological samples include body fluids (such as sera, plasma, urine, synovial fluid and spinal fluid) and tissue sources found to express the full length or fragments thereof of a polypeptide or mRNA. Methods for obtaining tissue biopsies and body fluids from mammals are well known in the art. Where the biological sample is to include mRNA, a tissue biopsy is the preferred source.
  • Total cellular RNA can be isolated from a biological sample using any suitable technique such as the single-step guanidinium-thiocyanate-phenol-chloroform method described in Chomczynski and Sacchi, Anal. Biochem. 162:156-159 (1987). Levels of mRNA encoding the polypeptides of the invention are then assayed using any appropriate method. These include Northern blot analysis, Sl nuclease mapping, the polymerase chain reaction (PCR), reverse transcription in combination with the polymerase chain reaction (RT-PCR), and reverse transcription in combination with the ligase chain reaction (RT-LCR).
  • PCR polymerase chain reaction
  • RT-PCR reverse transcription in combination with the polymerase chain reaction
  • RT-LCR reverse transcription in combination with the ligase chain reaction
  • the present invention also relates to diagnostic assays such as quantitative and diagnostic assays for detecting levels of polypeptides that bind to, are bound by, or associate with albumin fusion proteins of the invention, in a biological sample (e.g., cells and tissues), including determination of normal and abnormal levels of polypeptides.
  • a diagnostic assay in accordance with the invention for detecting abnormal expression of polypeptides that bind to, are bound by, or associate with albumin fusion proteins compared to normal control tissue samples may be used to detect the presence of tumors.
  • Assay techniques that can be used to determine levels of a polypeptide that bind to, are bound by, or associate with albumin fusion proteins of the present invention in a sample derived from a host are well-known to those of skill in the art. Such assay methods include radioimmunoassays, competitive-binding assays, Western Blot analysis and ELISA assays. Assaying polypeptide levels in a biological sample can occur using any art-known method. ''" ⁇ " "[0'4OfI" 1 ' MgSyiiig'pol
  • polypeptide expression in tissues can be studied with classical immunohistological methods (Jalkanen et al., J. Cell. Biol. 101:976-985 (19S 5 ); Jalkanen, M., et al., J. Cell . Biol. 10 5 :3087-3096 (1987)).
  • Other methods useful for detecting polypeptide gene expression include immunoassays, such as the enzyme linked immunosorbent assay (ELBA) and the radioimmunoassay (RIA).
  • ELBA enzyme linked immunosorbent assay
  • RIA radioimmunoassay
  • Suitable antibody assay labels include enzyme labels, such as, glucose oxidase, and radioisotopes, such as iodine ( 125 I, ' 21 I), carbon ( 14 C), sulfur ( 35 S), tritium ( 3 H), indium (“ 2 In), and technetium ( 99 Tc), and fluorescent labels, such as fluorescein and rhodamine, and biotin.
  • enzyme labels such as, glucose oxidase, and radioisotopes, such as iodine ( 125 I, ' 21 I), carbon ( 14 C), sulfur ( 35 S), tritium ( 3 H), indium (“ 2 In), and technetium ( 99 Tc)
  • fluorescent labels such as fluorescein and rhodamine, and biotin.
  • the tissue or cell type to be analyzed will generally include those which are known, or suspected, to express the gene of interest (such as, for example, cancer).
  • the protein isolation methods employed herein may, for example, be such as those described in Harlow and Lane (Harlow, E. and Lane, D., 1988, “Antibodies: A Laboratory Manual", Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York), which is incorporated herein by reference in its entirety.
  • the isolated cells can be derived fiom cell culture or from a patient.
  • the analysis of cells taken from culture may be a necessary step in the assessment of cells that could be used as part of a cell-based gene therapy technique or, alternatively, to test the effect of compounds on the expression of the gene.
  • albumin fusion proteins may be used to quantitatively or qualitatively detect the presence of polypeptides that bind to, are bound by, or associate with albumin fusion proteins of the present invention. This can be accomplished, for example, by immunofluorescence techniques employing a fluorescently labeled albumin fusion protein coupled with light microscopic, flow cytometric, or fluorimetric detection.
  • albumin fusion proteins comprising at least a fragment or variant of an antibody that specifically binds at least a Therapeutic protein disclosed herein (e.g., the Therapeutic proteins disclosed in Table 1) or otherwise known in the art may be used to quantitatively or qualitatively detect the presence of gene products or conserved variants or peptide fragments thereof. This can be accomplished, for example, by immunofluorescence techniques employing a fluorescently labeled antibody coupled with light microscopic, flow cytometric, or fluorimetric detection.
  • the albumin fusion proteins of the present invention may, additionally, be employed histologically, as in immunofluorescence, immunoelectron microscopy or non-immunological assays, for in situ detection of polypeptides that bind to, are bound by, or associate with an albumin fusion protein of the present invention.
  • In situ detection may be accomplished by removing a histological specimen from a patient, and applying thereto a labeled antibody or polypeptide of the present invention.
  • the albumin fusion proteins are preferably applied by overlaying the labeled albumin fusion proteins onto a biological sample.
  • Immunoassays and non-imrnunoassays that detect polypeptides that bind to, are.bound by, or- associate with albumin fusion proteins TVUT typically comprise incubating a sample, such as a biological fluid, a tissue extract, freshly harvested cells, or lysates of cells which have been incubated in cell culture, in the presence of a detectably labeled antibody capable of binding gene products or conserved variants or peptide fragments thereof, and detecting the bound antibody by any of a number of techniques well-known in the art.
  • the biological sample may be brought hi contact with and immobilized onto a solid phase support or carrier such as nitrocellulose, or other solid support which is capable of immobilizing cells, cell particles or soluble proteins.
  • a solid phase support or carrier such as nitrocellulose, or other solid support which is capable of immobilizing cells, cell particles or soluble proteins.
  • the support may then be washed with suitable buffers followed by treatment with the detectably labeled albumin fusion protein of the invention.
  • the solid phase support may then be washed with the buffer a second time to remove unbound antibody or polypeptide.
  • the antibody is subsequently labeled.
  • the amount of bound label on solid support may then be detected by conventional means.
  • solid phase support or carrier any support capable of binding a polypeptide (e.g., an albumin fusion protein, or polypeptide that binds, is bound by, or associates with an albumin fusion protein of the invention.
  • a polypeptide e.g., an albumin fusion protein, or polypeptide that binds, is bound by, or associates with an albumin fusion protein of the invention.
  • Well-known supports or carriers include glass, polystyrene, polypropylene, polyethylene, dextran, nylon, amylases, natural and modified celluloses, polyacrylamides, gabbros, and magnetite.
  • the nature of the carrier can be either soluble to some extent or insoluble for the purposes of the present invention.
  • the support material may have virtually any possible structural configuration so long as the coupled molecule is capable of binding to a polypeptide.
  • the support configuration may be spherical, as in a bead, or cylindrical, as in the inside surface of a test tube, or the external surface of a rod.
  • the surface may be flat such as a sheet, test strip, etc.
  • Preferred supports include polystyrene beads.
  • albumin fusion proteins of the invention are used to image diseased or neoplastic cells.
  • Labels or markers for in vivo imaging of albumin fusion proteins of the invention include those detectable by X-radiography, NMR, MRI, CAT-scans or ESR.
  • suitable labels include radioisotopes such as barium or cesium, which emit detectable radiation but markers for NMR and ESR include those with a detectable characteristic spin, such as deuterium, which may be incorporated into the albumin fusion protein by labeling of nutrients of a cell line (or bacterial or yeast strain) engineered.
  • albumin fusion proteins of the invention whose presence can be detected, can be administered.
  • albumin fusion proteins of the invention labeled with a radio-opaque or other appropriate compound can be administered and visualized in vivo, as discussed, above for labeled antibodies. Further, such polypeptides can be utilized for in vitro diagnostic procedures.
  • a polypeptide-specific antibody or antibody fragment which has been labeled with an appropriate detectable imaging moiety such as a radioisotope (for example, 131 I, " 2 In, 99 Tc), a radio-opaque substance, or a material detectable by nuclear magnetic resonance, is introduced (for example, parenterally, subcutaneously or intraperitoneally) into the mammal to be examined for a disorder.
  • an appropriate detectable imaging moiety such as a radioisotope (for example, 131 I, " 2 In, 99 Tc), a radio-opaque substance, or a material detectable by nuclear magnetic resonance.
  • the labeled albumin fusion protein will then preferentially accumulate at the locations in the body which contain a polypeptide or other substance that binds to, is bound by or associates with an albumin fusion protein of the present invention.
  • In vivo tumor imaging is described in S.W. Burchiel et al., "Immunopharmacokinetics of Radiolabeled Antibodies and Their Fragments" (Chapter 13 in Tumor Imaging: The Radiochemical Detection of Cancer, S.W. Burchiel and B. A. Rhodes, eds., Masson Publishing Inc. (1982)).
  • an albumin fusion protein of the present invention can be detectably labeled is by linking the same to a reporter enzyme and using the linked product in an enzyme immunoassay (EIA) (Voller, A., "The Enzyme Linked Immunosorbent Assay (ELISA)", 1978, Diagnostic Horizons 2:1-7, Microbiological Associates Quarterly Publication, Walkersville, MD); Voller et al., J. Clin. Pathol. 31:507-520 (1978); Butler, IE., Meth. En ⁇ ymol. 73:482-523 (1981); Maggio, E. (ed.), 1980, Enzyme Immunoassay, CRC Press, Boca Raton, FL,; Ishikawa, E.
  • EIA enzyme immunoassay
  • the reporter enzyme which is bound to the antibody will react with an appropriate substrate, preferably a chromogenic substrate, in such a manner as to produce a chemical moiety which can be detected, for example, by spectrophotometric, fluorimetric or by visual means.
  • Reporter enzymes which can be used to detectably label the antibody include, but are not limited to, malate dehydrogenase, staphylococcal nuclease, delta-5-steroid isomerase, yeast alcohol dehydrogenase, alpha-glycerophosphate, dehydrogenase, triose phosphate isomerase, horseradish peroxidase, alkaline phosphatase, asparaginase, glucose oxidase, beta-galactosidase, ribonuclease, urease, catalase, glucose-6-phosphate dehydrogenase, glucoamylase and acetylcholinesterase. Additionally, the detection can be accomplished by colorimetric methods which employ a chromogenic substrate for the reporter enzyme. Detection may also be accomplished by visual comparison of the extent of enzymatic reaction of a substrate in comparison with similarly prepared standards.
  • Albumin fusion proteins may also be radiolabelled and used in any of a variety of other immunoassays.
  • RIA radioimmunoassay
  • the radioactive isotope can be detected by means including, but not limited to, a gamma counter, a scintillation counter, or autoradiography.
  • chelator molecules are known in the art and can be used to label the Albumin fusion proteins.
  • Chelator molecules may be attached Albumin fusion proteins of the invention to facilitate labeling said protein with metal ions including radionuclides or fluorescent labels.
  • metal ions including radionuclides or fluorescent labels.
  • chelator which can be covalently bound to said Albumin fusion proteins may be used according to the present invention.
  • the chelator may further comprise a linker moiety that connects the chelating moiety to the Albumin fusion protein.
  • the Albumin fusion protein of the invention are attached to an acyclic chelator such as diethylene triamine- N,N,N',N",N"-pentaacetic acid (DPTA), analogues of DPTA, and derivatives of DPTA.
  • acyclic chelator such as diethylene triamine- N,N,N',N",N"-pentaacetic acid (DPTA), analogues of DPTA, and derivatives of DPTA.
  • the chelator may be 2-(p- isothiocyanatobenzyl)-6- methyldiethylenetriaminepentaacetic acid (1B4M-DPTA, also known as MX-DTPA), 2-methyl-6-(rho-nitrobenzyl)-l,4,7- triazaheptane-N,N,N',N",N"-pentaacetic acid (nitro-lB4M-DTPA or nitro-MX-DTPA); 2-(p-isothiocyanatobenzyl)- cyclohexyldiethylenetriaminepentaacetic acid (CHX-DTPA), or N-[2-amino-3-(rho-nitrophenyl)propyl]-trans-cyclohexane-l,2-diamine-N,N',N"- pentaacetic acid (nitro-CHX-A-DTPA).
  • 1B4M-DPTA also known as MX-DTPA
  • the Albumin fusion protein of the invention are attached to an acyclic terpyridine chelator such as 6,6"- bis[[N,N,N",N"- tetra(carboxymethyl)amino]methyl]-4'-(3-amino-4-methoxyphenyl)-2,2':6',2 "- terpyridine (TMT-amine).
  • an acyclic terpyridine chelator such as 6,6"- bis[[N,N,N",N"- tetra(carboxymethyl)amino]methyl]-4'-(3-amino-4-methoxyphenyl)-2,2':6',2 "- terpyridine (TMT-amine).
  • the macrocyclic chelator which is attached to the the Albumin fusion protein of the invention is 1,4,7,10- tetraazacyclododecane-N,N',N",N'"-tetraacetic acid (DOTA).
  • the DOTA is attached to the the Albumin fusion protein of the invention via a linker molecule.
  • linker molecules useful for conjugating DOTA to a polypeptide are commonly known in the art " ' u ]l - &e,M ' Ah$ll ⁇ b£N&''$ ' a$Mr »l &aiicer Res. 4(10):2483-90, 1998; Peterson et al, Bioconjug.
  • Bifunctional chelators based on macrocyclic ligands in which conjugation is via an activated arm, or functional group, attached to the carbon backbone of the ligand can be employed as described by M. Moi et al, J. Amer. Chem. Soc. 49:2639 (1989) (2-p-nitrobenzyl-l,4,7,10- tetraazacyclododecane-N,N',N",N'"-tetraacetic acid); S. V. Deshpande et al, J. Nncl. Med. 31:473 (1990); G. Ruser et al., Bioconj. Chem. 7:345
  • a macrocyclic chelator such as polyazamacrocyclic chelators, optionally containing one or more carboxy, amino, hydroxamate, phosphonate, or phosphate groups, are attached to the Albumin fusion protein of the invention.
  • the chelator is a chelator selected from the group consisting of DOTA, analogues of DOTA, and derivatives of DOTA.
  • suitable chelator molecules that may be attached to the the Albumin fusion protein of the invention include DOXA
  • Suitable chelators include chelating agents disclosed in U.S. Patent Nos.
  • suitable macrocyclic carboxylic acid chelators which can be used in the present invention include 1,4,7,10- tetraazacyclododecane- ⁇ yV v ⁇ '",iV"-tetraacetic acid (DOTA); l,4,8,12-tetraazacyclopentadecane- ⁇ f , ⁇ ?', ⁇ >'', ⁇ !'-tetraacetic acid (1 5 N4); 1,4,7- triazacyclononane-N,N',N"-triacetic acid (9N3); l,5,9-triazacyclododecane-N,N',N"-triacetic acid (12N3); and 6-bromoacetamido-benzyl-l,4,8,ll- tetraazacyclotetradecane- N,ht,N" ⁇ "-tetraacetic acid (BAT).
  • DOTA 1,4,7,10- tetraazacyclododecane- ⁇ yV v
  • a preferred chelator that can be attached to the Albumin Fusion protein of the invention is ⁇ -(5-isothiocyanato- 2-methoxyphenyl)- l,4,7,10-tetraazacyclododecane-l,4,7,10-tetraacetic acid, which is also known as MeO-DOTA-NCS.
  • a salt or ester of ⁇ -(5-isothiocyanato- 2- methoxyphenyl)- l,4,7,10-tetraazacyclododecane-l,4,7,10-tetraacetic acid may also be used.
  • Albumin fusion proteins of the invention to which chelators such as those decribed are covalently attached may be labeled (via the coordination site of the chelator) with radionuclides that are suitable for therapeutic, diagnostic, or both therapeutic and diagnostic purposes.
  • radionuclide used for diagnostic purposes Fe, Gd, 111 In, 67 Ga, or 68 Ga. In another embodiment, the radionuclide used fordiagnostic purposes is
  • radionuclide used for therapeutic purposes are 166 Ho, 165 Dy, 90 Y, I 15m In, 52 Fe, or 72 Ga.
  • the radionuclide used for diagnostic purposes is 166 Ho or 90 Y.
  • the radionuclides used for both therapeutic and diagnostic purposes include
  • the radionuclide is 153 Sm, 177 Lu, 159 Gd, 175 Yb, Or 47 Sc.
  • the radionuclide is 153 Sm, 177 Lu, 175 Yb, or 159 Gd.
  • Preferred metal radionuclides include 90 Y, 99m Tc, 111 In, 47 Sc, 67 Ga, 51 Cr, 177m Sn, 67 Cu, 167 Tm, 97 Ru, 188 Re, 177 Lu, 199 Au, 47 Sc, 67 Ga, 51 Cr,
  • Albumin fusion proteins of the invention to which chelators are covalently attached may be labeled with a metal ion selected from the group consisting Of 90 Y, 111 In, 177 Lu, 166 Ho, 215 Bi, and 225 Ac.
  • ⁇ -emitting radionuclides such as 99 Tc, 111 In, 67 Ga, and 169 Yb have been approved or under investigation for diagnostic imaging, while ⁇ -emitters, such as 07 Cu, 111 Ag, 186 Re, and 90 Y are useful for the applications in tumor therapy.
  • radionuclides include y-emitters, such as 99 Tc, 111 In, 67 Ga, and 169 Yb, and ⁇ -emitters, such as 67 Cu, 111 Ag, 186 Re, 188 Re and 90 Y, as well as other radionuclides of interest such as 211 At, 212 Bi, 177 Lu, 86 Rb , 105 Rh, 153 Sm, 198 Au, 149 Pm, 85 Sr, 142 Pr, 214 Pb, 109 Pd, 166 Ho, 208 Tl, and 44 Sc.
  • Albumin fusion proteins of the invention to which chelators are covalently attached may be labeled with the radionuclides described above.
  • Albumin fusion proteins of the invention to which chelators are covalently attached may be labeled with paramagnetic metal ions including ions of transition and lanthanide metal, such as metals having atomic numbers of 21-29, 42, 43, 44, or 57-71, in particular ions of Cr, V, Mn, Fe, Co, Ni, Cu, La, Ce, Pr, Nd, Pm, Sm, Eu, Gd, Tb, Dy, Ho, Er, Tm, Yb, and Lu.
  • the paramagnetic metals used in compositions for magnetic resonance imaging include the elements having atomic numbers of 22 to 29, 42, 44 and 58-70.
  • Albumin fusion proteins of the invention to which chelators are covalently attached may be labeled with fluorescent metal ions including lanthanides, in particular La, Ce, Pr, Nd, Pm, Sm, Eu (e.g., 152 Eu), Gd, Tb, Dy, Ho, Er, Tm, Yb, and Lu.
  • fluorescent metal ions including lanthanides, in particular La, Ce, Pr, Nd, Pm, Sm, Eu (e.g., 152 Eu), Gd, Tb, Dy, Ho, Er, Tm, Yb, and Lu.
  • Albumin fusion proteins of the invention to which chelators are covalently attached may be labeled with heavy metal-containing reporters may include atoms of Mo, Bi, Si, and W.
  • the albumin fusion protein can also be detectably labeled using fluorescence emitting metals such as ' 52 Eu, or others of the lanthanide series. These metals can be attached to the antibody using such metal chelating groups as diethylenetriaminepentacetic acid (DTPA) or ethylenediaminetetraaeetic acid (EDTA).
  • DTPA diethylenetriaminepentacetic acid
  • EDTA ethylenediaminetetraaeetic acid
  • the albumin fusion proteins can also can be detectably labeled by coupling it to a chemiluminescent compound. The presence of the chemiluminescent-tagged albumin fusion protein is then determined by detecting the presence of luminescence that arises during the course of a chemical reaction.
  • chemiluminescent labeling compounds are luminol, isoluminol, theromatio acridinium ester, imidazole, acridinium salt and oxalate ester.
  • a bioluminescent compound may be used to label albumin fusion proteins of the present invention.
  • Bioluminescence is a type of chemiluminescence found in biological systems in, which a catalytic protein increases the efficiency of the chemiluminescent reaction. The presence of a bioluminescent protein is determined by detecting the presence of luminescence.
  • Important bioluminescent compounds for purposes of labeling are luciferin, luciferase and aequorin.
  • Transgenic organisms that express the albumin fusion proteins of the invention are also included in the invention.
  • Transgenic organisms are genetically modified organisms into which recombinant, exogenous or cloned genetic material has been transferred. Such genetic material is often referred to as a transgene.
  • the nucleic acid sequence of the transgene may include one or more transcriptional regulatory sequences and other nucleic acid sequences such as introns, that may be necessary for optimal expression and secretion of the encoded protein.
  • the transgene may be designed to direct the expression of the encoded protein in a manner that facilitates its recovery from the organism or from a product produced by the organism, e.g. from the milk, blood, urine, eggs, hair or seeds of the organism.
  • the transgene may consist of nucleic acid sequences derived from the genome of the same species or of a different species than the species of the target animal.
  • the transgene may be integrated either at a locus of a genome where that particular nucleic acid sequence is not otherwise normally found or at the normal locus for the transgene.
  • the term "germ cell line transgenic organism” refers to a transgenic organism in which the genetic alteration or genetic information was introduced into a germ line cell, thereby conferring the ability of the transgenic organism to transfer the genetic information to offspring. If such offspring in fact possess some or all of that alteration or genetic information, then they too are transgenic organisms.
  • the alteration or genetic information may be foreign to the species of organism to which the recipient belongs, foreign only to the particular individual recipient, or may be genetic information already possessed by the recipient. In the last case, the altered or introduced gene may be expressed differently than the native gene.
  • a transgenic organism may be a transgenic animal or a transgenic plant.
  • Transgenic animals can be produced by a variety of different methods including transfection, el ⁇ ctroporation, microinjection, gene targeting hxembjryomc.stem. cells and recombinant viral and retroviral infection (see, e.g., U.S. Patent No. 4,736,8 6 6; U.S. Patent No. 5,602,307; Mullins et al (1993) Hypertension 22(4):630-633; Brenin et al. (1997) Surg. Oncol. 6(2)99-110; Tuan (ed.), Recombinant Gene Expression Protocols, Methods in Molecular Biology No. 62, Humana Press (1997)).
  • the method of introduction of nucleic acid fragments into recombination competent mammalian cells can be by any method which favors co-transformation of multiple nucleic acid molecules.
  • Detailed procedures for producing transgenic animals are readily available to one skilled in the art, including the disclosures in U.S. Patent No. 5,489,743 and U.S. Patent No. 5,602,307.
  • a number of recombinant or transgenic mice have been produced, including those which express an activated oncogene sequence (U.S. Patent No. 4,736,866); express simian SV40 T-antigen (U.S. Patent No. 5,728,915); lack the expression of interferon regulatory factor 1 (IRF-I) (U.S. Patent No. 5 ,731,490); exhibit dopaminergic dysfunction (U.S. Patent No. 5 ,723,719); express at least one human gene which participates in blood pressure control (U.S. Patent No. 5,731,489); display greater similarity to the conditions existing in naturally occurring Alzheimer's disease (U.S. Patent No.
  • mice and rats remain the animals of choice for most transgenic experimentation, in some instances it is preferable or even necessary to use alternative animal species.
  • Transgenic procedures have been successfully utilized in a variety of non-murine animals, including sheep, goats, pigs, dogs, cats, monkeys, chimpanzees, hamsters, rabbits, cows and guinea pigs (see, e.g., Kim et al. (1997) MoI. Reprod. Dev. 46(4):515-526; Houdebine (1995) Reprod. Nutr. Dev. 35(6):609-617; Petters (1994) Reprod. Fertil. Dev. 6(5):643-645; Schnieke et al. (1997) Science 278(5346):2130-2133; and Amoah (1997) J. Animal Science 75(2):578-585).
  • transgene-encoded protein of the invention may be put under the control of a promoter that is preferentially activated in mammary epithelial cells.
  • Promoters that control the genes encoding milk proteins are preferred, for example the promoter for casein, beta lactoglobulin, whey acid protein, or lactalbumin (see, e.g., DiTullio (1992) BioTechnology 10:74-77; Clark et al. (1989) BioTechnology 7:487-492; Gorton et al. (1987) BioTechnology 5:1183-1187; and Soulier et al. (1992) FEBS Letts.
  • transgenic mammals of choice would produce large volumes of milk and have long lactating periods, for example goats, cows, camels il LII IO or ,/ s.he yep. ::;:» I , ⁇ ' ⁇ n::::: “all .,:::li "-sit ,l.
  • An albumin fusion protein of the invention can also be expressed in a transgenic plant, e.g. a plant in which the DNA transgene is inserted into the nuclear or plastidic genome.
  • Plant transformation procedures used to introduce foreign nucleic acids into plant cells or protoplasts are known in the art. See, in general, Methods in Enzymology Vol. 1 5 3 ("Recombinant DNA Part D") 1987, Wu and Grossman Eds., Academic Press and European Patent Application EP 693554. Methods for generation of genetically engineered plants are further described in US Patent No. 5,283,184, US Patent No. 5, 482,8 5 2, and European Patent Application EP 693 554, all of which are hereby incorporated by reference.
  • the albumin fusion proteins of the invention or formulations thereof may be administered by any conventional method including parenteral (e.g. subcutaneous or intramuscular) injection or intravenous infusion.
  • the treatment may consist of a single dose or a plurality of doses over a period of time.
  • an albumin fusion protein of the invention While it is possible for an albumin fusion protein of the invention to be administered alone, it is preferable to present it as a pharmaceutical formulation, together with one or more acceptable carriers.
  • the carrier(s) must be "acceptable” in the sense of being compatible with the albumin fusion protein and not deleterious to the recipients thereof.
  • the carriers will be water or saline which will be sterile and pyrogen free.
  • Albumin fusion proteins of the invention are particularly well suited to formulation in aqueous carriers such as sterile pyrogen free water, saline or other isotonic solutions because of their extended shelf-life in solution.
  • compositions of the invention may be formulated well in advance in aqueous form, for instance, weeks or months or longer time periods before being dispensed.
  • formulations containing the albumin fusion protein may be prepared taking into account the extended shelf-life of the albumin fusion protein in aqueous formulations. As discussed above, the shelf-life of many of these Therapeutic proteins are markedly increased or prolonged after fusion to HA.
  • the albumin fusion proteins of the invention can be formulated as aerosols using standard procedures.
  • aerosol includes any gas-borne suspended phase of an albumin fusion protein of the instant invention which is capable of being inhaled into the bronchioles or nasal passages.
  • aerosol includes a gas-borne suspension of droplets of an albumin fusion protein of the instant invention, as may be produced in a metered dose inhaler or nebulizer, or in a mist sprayer.
  • Aerosol also includes a dry powder composition of a compound of the instant invention suspended in air or other carrier gas, which may be delivered by insufflation from an inhaler device, for example.
  • the formulations of the invention are also typically non-immunogenic, in part, because of the use of the components of the albumin fusion protein being derived from the proper species. For instance, for human use, both the Therapeutic protein and albumin portions of the albumin fusion protein will typically be-human.
  • compositions may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. Such methods include the step of bringing into association the albumin fusion protein with the carrier that constitutes one or more accessory ingredients. In general the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product.
  • Formulations suitable for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation appropriate for the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
  • the formulations may be presented in unit-dose or multi-dose containers, for example sealed ampules, vials or syringes, and may be stored in a freeze-dried (lyophilised) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders.
  • Dosage formulations may contain the Therapeutic protein portion at a lower molar concentration or lower dosage compared to the non-fused standard formulation for the Therapeutic protein given the extended serum half-life exhibited by many of the albumin fusion proteins of the invention.
  • an albumin fusion protein of the invention comprises one of the proteins listed in the "Therapeutic Protein:X" column of Table 1 as one or more of the Therapeutic protein regions
  • the dosage form can be calculated on the basis of the potency of the albumin fusion protein relative to the potency of the therapeutic protein alone, while taking into account the prolonged serum half-life and shelf-life of the albumin fusion proteins compared to that of native therapeutic protein. For example, if the therapeutic protein is typically administered at 0.3 to 30.0 IU/kg/week, or 0.9 to 12.0 IU/kg/week, given in three or seven divided doses for a year or more. In an albumin fusion protein consisting of full length HA fused to a therpeutic protein, an equivalent dose in terms of units would represent a greater weight of agent but the dosage frequency can be reduced, for example to twice a week, once a week or less.
  • Formulations or compositions of the invention may be packaged together with, or included in a kit with, instructions or a package insert referring to the extended shelf-life of the albumin fusion protein component.
  • instructions or package inserts may address ⁇ " IU rec'omme ⁇ r ⁇ ed ⁇ torage' ' con'diti ⁇ tis7suc ⁇ i as'time, temperature and light, taking into account the extended or prolonged shelf-life of the albumin fusion proteins of the invention.
  • Such instructions or package inserts may also address the particular advantages of the albumin fusion proteins of the inventions, such as the ease of storage for formulations that may require use in the field, outside of controlled hospital, clinic or office conditions.
  • formulations of the invention may be in aqueous form and may be stored under less than ideal circumstances without significant loss of therapeutic activity.
  • albumin fusion proteins of the invention can also be included in nutraceuticals.
  • certain albumin fusion proteins of the invention may be administered in natural products, including milk or milk product obtained from a transgenic mammal which expresses albumin fusion protein.
  • Such compositions can also include plant or plant products obtained from a transgenic plant which expresses the albumin fusion protein.
  • the albumin fusion protein can also be provided in powder or tablet form, with or without other known additives, carriers, fillers and diluents. Nutraceuticals are described in Scott Hegenhart, Food Product Design, Dec. 1993.
  • the invention also provides methods of treatment and/or prevention of diseases or disorders (such as, for example, any one or more of the diseases or disorders disclosed herein) by administration to a subject of an effective amount of an albumin fusion protein of the invention or a polynucleotide encoding an albumin fusion protein of the invention ("albumin fusion polynucleotide”) in a pharmaceutically acceptable carrier.
  • diseases or disorders such as, for example, any one or more of the diseases or disorders disclosed herein
  • the albumin fusion protein and/or polynucleotide will be formulated and dosed in a fashion consistent with good medical practice, taking into account the clinical condition of the individual patient (especially the side effects of treatment with the albumin fusion protein and/or polynucleotide alone), the site of delivery, the method of administration, the scheduling of administration, and other factors known to practitioners.
  • the "effective amount" for purposes herein is thus determined by such considerations.
  • the total pharmaceutically effective amount of the albumin fusion protein administered parenterally per dose will be in the range of about lug/kg/day to 10 mg/kg/day of patient body weight, although, as noted above, this will be subject to therapeutic discretion. More preferably, this dose is at least 0.01 mg/kg/day, and most preferably for humans between about 0.01 and 1 mg/kg/day for the hormone.
  • the albumin fusion protein is typically administered at a dose rate of about 1 ug/kg/hour to about 50 ug/kg/hour, either by 1-4 injections per day or by continuous subcutaneous infusions, for example, using a mini-pump. An intravenous bag solution may also be employed. The length of treatment needed to observe changes and the interval following treatment for responses to occur appears to vary depending on the desired effect.
  • the albumin fusion protein of the invention has a higher plasma stablity compared to the Therapeutic protein portion (or fragment or variant thereof) alone. This increase in plasma stability should be taken into account when determining the effective amount of the albumin fusion protein to be administered per dose and the dosing administration schedule.
  • higher plasma stability may allow the albumin fusion protein to be administered at a lower dose at the same frequency of administrations, or alternatively, may allow the albumin fusion protein to be administered in fewer dosings.
  • the higher stability allows the albumin fusion protein of the invention to be administered less often in fewer dosings. More preferably, the albumin fusion protein can be administered once every two weeks.
  • the albumin fusion protein can be administered once every three, four, five, or more weeks depending on the pharmacokinetics of the albumin fusion protein.
  • the pharmacokinetics of an IFN-alpha-HSA fusion protein supports a dosing regimen of once every 2-4 weeks or more, and even dosing at intervals of 4 weeks or more than every 4 weeks.
  • the effective amount of the albumin fusion protein to be administered per dose can also be denoted as the total formulated albumin fusion protein concentration given per dose.
  • the total formulated albumin fusion protein concentration administered to a patient per dose is in the range of about 10 ug/dose to about 2000 ug/dose. More preferably, the total concentration is in the range of about 100 ug/dose to about 1000 ug/dose, or alternatively, about 1000 ug/dose to about 1200 ug/dose or about 900 ug/dose to about 1800 ug/dose.
  • an IFN-alpha-HSA fusion protein of the invention (e.g., produced by CIDs 2249, 2343, 2366, 2381, 2382, 2410, 3165, 3422, 3423, 3424, 3476, 3960, 4290, 4291, 4292, 4295, or 4296) is dosed in a total formulated concentration of about 90 ug/dose to about 2000 ug/dose.
  • an IFN-alpha-HSA fusion protein of the invention (e.g., produced by CIDs 2249, 2343, 2366, 2381, 2382, 2410, 3165, 3422, 3423, 3424, 3476, 3960, 4290, 4291, 4292, 4295, or 4296) is dosed in a total formulated concentration of about 900 ug/dose to about 2000 ug/dose, about 900 ug/dose to about 1200 ug/dose, about 900 ug/dose to about 1800 ug/dose and most preferably in a total formulated concentration of about 1200 ug/dose to about 1800 ug/dose.
  • an EFN-alpha-HSA fusion protein of the invention (e.g., produced by CIDs 2249, 2343, 2366, 2381, 2382, 2410, 3165, 3422, 3423, 3424, 3476, 3960, 4290, 4291, 4292, 4295, or 4296) is dosed in a total formulated concentration of 600 ug/dose, 720 ug/dose, 800 ug/dose, 900 ug/dose, 1000 ug/dose, 1200 ug/dose, 1500 ug/dose, 1800 ug/dose, or 2000 ug/dose.
  • the total formulated dose of an IFN-alpha-HSA fusion protein of the invention is administered either alone or in combination with an antiviral compound, such as ribavirin.
  • the total formulated dose of an IFN-alpha-HSA fusion protein of the invention is administered in combination with one or more antiviral compounds, including, but not limited to ribavirin.
  • the total formulated concentration of an IFN-alpha-HSA fusion proteins of the invention is administered to treat a patient infected with HCV.
  • the IFN-alpha-HSA fusion proteins of the invention are administered to a Treatment na ⁇ ve patient with HCV either alone or in combination with an effective amount of an antiviral compound, such as ribavirin, in a total formulated concentration of about 90 ug/dose to about 2000 ug/dose.
  • an antiviral compound such as ribavirin
  • the IFN-alpha-HSA fusion protein of the invention (e.g., produced by CEDs 2249, 2343, 2366, 2381, 2382, 2410, 3165, 3422, 3423, 3424, 3476, 3960, 4290, 4291, 4292, 4295, or 4296) is administered to a Treatment na ⁇ ve patient with HCV either alone or in combination with an effective amount of antiviral compound, such as ribavirin, in a total formulated concentration of about 900 ug/dose to about 2000 ug/dose, about 900 ug/dose to about 1200 ug/dose, about 900 ug/dose to about 1800 ug/dose and most preferably in a total formulated concentration of about 1200 ug/dose to about 1800 ug/dose.
  • an effective amount of antiviral compound such as ribavirin
  • an IFN-alpha-HSA fusion protein of the invention (e.g., produced by CIDs 2249, 2343, 2366, 2381, 2382, 2410, 3165, 3422, 3423, 3424, 3476, 3960, 4290, 4291, 4292, 4295, or 4296) is administered to a Treatment na ⁇ ve patient with HCV either alone or in combination with an effective amount of antiviral compound, such as ribavirin, in a total formulated concentration of 600 ug/dose, 720 ug/dose, 800 ug/dose, 900 ug/dose, 1000 ug/dose, 1200 ug/dose, 1500 ug/dose, 1800 ug/dose, or 2000 ug/dose.
  • an effective amount of antiviral compound such as ribavirin
  • the total formulated concentration of an IFN-alpha-HSA fusion proteins of the invention are administered to a Treatment na ⁇ ve patient with HCV in combination with an effective amount of one or more antiviral compounds, including, for example, ribavirin, in a total formulated concentration of about 90 ug/dose to about 2000 ug/dose.
  • one or more antiviral compounds including, for example, ribavirin
  • the IFN-alpha-HSA fusion protein of the invention (e.g., produced by CIDs 2249, 2343, 2366, 2381, 2382, 2410, 3165, 3422, 3423, 3424, 3476, 3960, 4290, 4291, , 4292, 4295, or 4296) is administered to a Treatment na ⁇ ve patient with HCV in combination with an effective amount of one or more antiviral compounds, including, for example, ribavirin, in a total formulated concentration of about 900 ug/dose to about 2000 ug/dose, about 900 ug/dose to about 1200 ug/dose, about 900 ug/dose to about 1800 ug/dose and most preferably in a total formulated concentration of about 1200 ug/dose to about 1800 ug/dose.
  • an effective amount of one or more antiviral compounds including, for example, ribavirin
  • an IFN-alpha-HSA fusion protein of the invention (e.g., produced by CIDs 2249, 2343, 2366, 2381, 2382, 2410, 3165, 3422, 3423, 3424, 3476, 3960, 4290, 4291, 4292, 4295, or 4296) is administered to a Treatment na ⁇ ve patient with HCV in combination with an effective amount of one or more antiviral compounds, including, for example, ribavirin, in a total formulated concentration of 600 ug/dose, 720 ug/dose, 800 ug/dose, 900 ug/dose, 1000 ug/dose, 1200 ug/dose, 1500 ug/dose, 1800 ug/dose, or 2000 ug/dose.
  • an antiviral compounds including, for example, ribavirin
  • an IFN-alpha-HSA fusion protein of the invention (e.g., produced by CIDs 2249, 2343, 2366, 2381, 2382, 2410, 3165, 3422, 3423, 3424, 3476, 3960, 4290, 4291, 4292, 4295, or 4296) is administered to a Treatment experienced patient with HCV either alone or in combination with an effective amount of antiviral compound, such as ribavirin, in a total formulated concentration of about 90 ug/dose to about 2000 ug/dose.
  • an effective amount of antiviral compound such as ribavirin
  • an IFN-alpha-HSA fusion protein of the invention (e.g., produced by CEDs 2249, 2343, 2366, 2381; 2382, 2410, 3165, 3422, 3423; 3424, 3476, 3960; 4290, 4291, 4292,-4295, or 4296) is administered -to a Treatment experienced patientwith HCV either alone or in combination with an effective amount of antiviral compound, such as ribavirin, in a total formulated concentration of about 900 ug/dose to about 2000 ug/dose, about 900 ug/dose to about 1200 ug/dose, about 900 ug/dose to about 1800 ug/dose and most preferably in a total formulated concentration of about 1200 ug/dose to about 1800 ug/dose.
  • an effective amount of antiviral compound such as ribavirin
  • an EFN-alpha-HSA fusion proteins of the invention (e.g., produced by CIDs 2249, 2343, 2366, 2381, 2382, 2410, 3165, 3422, 3423, 3424, 3476, 3960, 4290, 4291, 4292, 4295, or 4296) is administered to a Treatment experienced patient with HCV either alone or in combination with an effective amount of antiviral compound, such as ribavirin, in a total formulated concentration of 600 ug/dose, 720 ug/dose, 800 ug/dose, 900 ug/dose, 1000 ug/dose, 1200 ug/dose, 1500 ug/dose, 1800 ug/dose, or 2000 ug/dose.
  • an effective amount of antiviral compound such as ribavirin
  • an IFN-alpha-HSA fusion protein of the invention (e.g., produced by CIDs 2249, 2343, 2366, 2381, 2382, 2410, 3165, 3422, 3423, 3424, 3476, 3960, 4290, 4291, 4292, 4295, or 4296) is administered to a Treatment experienced patient with HCV in combination with an effective amount of one or more antiviral compounds, including, for example, ribavirin, in a total formulated concentration of about 90 ug/dose to about 2000 ug/dose.
  • an antiviral compounds including, for example, ribavirin
  • an IFN-alpha-HSA fusion protein of the invention (e.g., produced by CIDs 2249, 2343, 2366, 2381, 2382, 2410, 3165, 3422, 3423, 3424, 3476, 3960, 4290, 4291, 4292, 4295, or 4296) is administered to a Treatment experienced patient with HCV in combination with an effective amount of one or more antiviral compounds, including, for example, ribavirin, in a total formulated concentration of about 900 ug/dose to about 2000 ug/dose, about 900 ug/dose to about 1200 ug/dose, about 900 ug/dose to about 1800 ug/dose and most preferably in a total formulated concentration of about 1200 ug/dose to about 1800 ug/dose.
  • an antiviral compounds including, for example, ribavirin
  • an IFN-alpha-HSA fusion proteins of the invention (e.g., produced by CIDs 2249, 2343, 2366, 2381, 2382, 2410, 3165, 3422, 3423, 3424, 3476, 3960, 4290, 4291, 4292, 4295, or 4296) is administered to a Treatment experienced patient with HCV in combination with an effective amount of one or more antiviral compounds, including, for example, ribavirin, in a total formulated concentration of 600 ug/dose, 720 ug/dose, 800 ug/dose, 900 ug/dose, 1000 ug/dose, 1200 ug/dose, I 5 OO ug/dose, 1800 ug/dose, or 2000 ug/dose.
  • an antiviral compounds including, for example, ribavirin
  • the total formulated concentration of the albumin fusion protein and the dosing interval in which the dosing interval at which the albumin fusion protein will administered will vary depending on the desired effect and the particular therapeutic protein adminstered.
  • the total formulated albumin fusion protein concentration administered to a patient per dose is hi the range of about 10 ug/dose to abd'ut 20OO" " ug/ ⁇ ose once a'Veel ⁇ 'Once every two weeks, once every three weeks, once every four weeks or more.
  • the total concentration is in the range of about 100 ug/dose to about 1000 ug/dose once a week, once every two weeks, once every three weeks, once every four weeks or more, or alternatively, about 1000 ug/dose to about 1200 ug/dose or about 900 ug/dose to about 1800 ug/dose once a week, once every two weeks, once every three weeks, once every four weeks or more.
  • an IFN-alpha-HSA f ⁇ sion protein of the invention (e.g., produced by CIDs 2249, 2343, 2366, 2381, 2382, 2410, 3165, 3422, 3423, 3424, 3476, 3960, 4290, 4291, 4292, 4295, or 4296) is administered at a total formulated concentration of about 90 ug/dose to about 2000 ug/dose once every two, three, four, or five weeks.
  • an IFN-alpha-HSA fusion protein of the invention (e.g., produced by CIDs 2249, 2343, 2366, 2381, 2382, 2410, 3165, 3422, 3423, 3424, 3476, 3960, 4290, 4291, 4292, 4295, or 4296) is dosed in a total formulated concentration of about 900 ug/dose to about 2000 ug/dose once every one, two, three, four or five weeks; about 900 ug/dose to about 1200 ug/dose once every one, two, three, four or five weeks; about 900 ug/dose to about 1800 ug/dose once every one, two, three, four or five weeks; and most preferably in a total formulated concentration of about 1200 ug/dose to 1800 ug/dose once every one, two, three, four or five weeks.
  • an IFN-alpha-HSA fusion protein of the invention (e.g., produced by CIDs 2249, 2343, 2366, 2381, 2382, 2410, 3165, 3422, 3423, 3424, 3476, 3960, 4290, 4291, 4292, 4295, or 4296) is administered at a total formulated concentration of about 600 ug/dose once every one, two, three, four or five weeks; 800 ug/dose once every one, two, three, four or five weeks, 900 ug/dose once every one, two, three, four or five weeks; 1000 ug/dose once every one, two, three, four or five weeks; 1200 ug/dose once every one, two, three, four or five weeks; 1500 ug/dose once every one, two, three, four or five weeks; 1600 ug/dose once every one, two, three, four or five weeks; 1800 ug/dose once every one, two, three, four or five weeks; or 2000 ug/dose
  • the IFN-alpha- HSA fusion protein of the invention (e.g., produced by CIDs 2249, 2343, 2366, 2381, 2382, 2410, 3165, 3422, 3423, 3424, 3476, 3960, 4290, 4291, 4292, 4295, or 4296) is administered at a total formulated concentration of 900 ug/dose once every two weeks, and more preferably at a total concentration of 1200 ug/dose once every two weeks, 1200 ug/dose once every four weeks, or 1800 ug/dose once every four weeks.
  • the total formulated dose of an IFN-alpha-HSA fusion protein of the invention (e.g., produced by CIDs 2249, 2343, 2366, 2381, 2382, 2410, 3165, 3422, 3423, 3424, 3476, 3960, 4290, 4291, 4292, 4295, or 4296) is administered either alone or in combination with an antiviral compound, sueh as ribavirin.
  • the total formulated dose of an IFN-alpha-HSA fusion protein of the invention is administered in combination with one or more antiviral compounds, including, for example, ribavirin.
  • an IFN-alpha-HSA fusion protein of the invention (e.g., produced by CIDs 2249, 2343, 2366, 2381, 2382, 2410, 3165, 3422, 3423, 3424, 3476, 3960, 4290, 4291, 4292, 4295, or 4296) is administered to a Treatment na ⁇ ve HCV patient at a total formulated concentration of about 90 ug/dose to about 2000 ug/dose once every two, three, four, or five weeks either alone or in combination with - an antiviral compound, such as ribavirin.
  • an IFN-alpha-HSA fusion protein of the- invention (e.g., produced by - CIDs 2249, 2343, 2366, 2381, 2382, 2410, 3165, 3422, 3423, 3424, 3476, 3960, 4290, 4291, 4292, 4295, or 4296) is administered to a Treatment na ⁇
  • an IFN-alpha-HSA fusion protein of the invention (e.g., produced by CIDs 2249, 2343, 2366, 2381, 2382, 2410, 3165, 3422, 3423, 3424, 3476, 3960, 4290, 4291, 4292, 429 5 , or 4296) is administered to a Treatment na ⁇ ve HCV patient at a total formulated concentration of about 90 ug/dose to about 2000 ug/dose once every two, three, four, or five weeks in combination with one or more antiviral compounds, including, for example, ribavirin.
  • an IFN-alpha-HSA fusion protein of the invention (e.g., produced by CDDs 2249, 2343, 2366, 2381, 2382, 2410, 3165, 3422, 3423, 3424, 3476, 3960, 4290, 4291, 4292, 4295, or 4296) is administered to a Treatment na ⁇ ve HCV patient in a total formulated concentration of about 900 ug/dose to about 2000 ug/dose once every one, two, three, four or five weeks; about 900 ug/dose to about 1200 ug/dose once every one, two, three, four or five weeks; about 900 ug/dose to about 1800 ug/dose once every one, two, three, four or five weeks; and most preferably in a total formulated concentration of about 1200 ug/dose to about 1800 ug/dose once every one, two, three, four or five weeks in combination with one or more antiviral compounds, including, for example, ribavirin
  • an IFN-alpha-HSA fusion protein of the invention (e.g., produced by CIDs 2249, 2343, 2366, 2381, 2382, 2410, 3165, 3422, 3423, '"" 429 ⁇ Or 4296) is administered to a Treatment na ⁇ ve HCV patient at a total formulated concentration of about 600 ug/dose once every one, two, three, four or five weeks; 800 ug/dose once every one, two, three, four or five weeks, 900 ug/dose once every one, two, three, four or five weeks; 1000 ug/dose once every one, two, three, four or five weeks; 1200 ug/dose once every one, two, three, four or five weeks; 1500 ug/dose once every one, two, three, four or five weeks; 1600 ug/dose once every one, two, three, four or five weeks; 1800 ug/dose once every one, two, three, four or five weeks; or 2000 ug/dose once every
  • an IFN-alpha-HSA fusion protein of the invention (e.g., produced by CIDs 2249, 2343, 2366, 2381, 2382, 2410, 3165, 3422, 3423, 3424, 3476, 3960, 4290, 4291, 4292, 4295, or 4296) is administered to a Treatment na ⁇ ve HCV patient at a total formulated concentration of 900 ug/dose once every two weeks, and more preferably at a total concentration of 1200 ug/dose once every two weeks, 1200 ug/dose once every four weeks, or 1800 ug/dose once every four weeks, either alone or in combination with an antiviral compound, such as ribavirin.
  • an antiviral compound such as ribavirin.
  • an IFN-alpha-HSA fusion protein of the invention (e.g., produced by CIDs 2249, 2343, 2366, 2381, 2382, 2410, 3165, 3422, 3423, 3424, 3476, 3960, 4290, 4291, 4292, 4295, or 4296) is administered to a Treatment na ⁇ ve HCV patient at a total formulated concentration of 900 ug/dose once every two weeks, and more preferably at a total concentration of 1200 ug/dose once every two weeks, 1200 ug/dose once every four weeks, or 1800 ug/dose once every four weeks, in combination with one or more antiviral compounds, including, for example, ribavirin.
  • an IFN-alpha-HSA fusion protein of the invention (e.g., produced by CIDs 2249, 2343, 2366, 2381, 2382, 2410, 3165, 3422, 3423, 3424, 3476, 3960, 4290, 4291, 4292, 4295, or 4296) is administered to a Treatment experienced HCV patient at a total formulated concentration of about 90 ug/dose to about 2000 ug/dose once every two, three, four, or five weeks either alone or in combination with an antiviral compound, such as ribavirin.
  • an antiviral compound such as ribavirin.
  • an IFN-alpha-HSA fusion protein of the invention (e.g., produced by CIDs 2249, 2343, 2366, 2381, 2382, 2410, 3165, 3422, 3423, 3424, 3476, 3960, 4290, 4291, 4292, 4295, or 4296) is administered to a Treatment experienced HCV patient in a total formulated concentration of about 900 ug/dose to about 2000 ug/dose once every one, two, three, four or five weeks; about 900 ug/dose to about 1200 ug/dose once every one, two, three, four or five weeks; about 900 ug/dose to about 1800 ug/dose once every one, two, three, four or five weeks; and most preferably in a total formulated concentration of about 1200 ug/dose to about 1800 ug/dose once every one, three, four or five weeks, or most preferably every two weeks either alone or in combination with an antiviral compound, such as ribavirin
  • an IFN-alpha-HSA fusion proteins of the invention (e.g., produced by CIDs 2249, 2343, 2366, 2381, 2382, 2410, 3165, 3422, 3423, 3424, 3476, 3960, 4290, 4291, 4292, 4295, or 4296) is administered to a Treatment experienced HCV patient at a total formulated concentration of about 600 ug/dose once every one, two, three, four or five weeks; 800 ug/dose once every one, two, three, four or five weeks, 900 ug/dose once every one, two, three, four or five weeks; 1000 ug/dose once every one, two, three, four or five weeks; 1200 ug/dose once - every one, two, three, four or five weeks; 1500 ug/dose once every one, two, three, four or five weeks; 1600 ug/dose once every one, two, three, four or five weeks; 1800 ug/dose once every one, two, three, four or
  • an IFN-alpha-HSA fusion protein of the invention (e.g., produced by CIDs 2249, 2343, 2366, 2381, 2382, 2410, 3165, 3422, 3423, 3424, 3476, 3960, 4290, 4291, 4292, 4295, or 4296) is administered to a Treatment experienced HCV patient at a total formulated concentration of about 90 ug/dose to about 2000 ug/dose once every two, three, four, or five weeks in combination with one or more antiviral compounds, including, for example, ribavirin.
  • an IFN-alpha-HSA fusion protein of the invention (e.g., produced by CIDs 2249, 2343, 2366, 2381, 2382, 2410, 3165, 3422, 3423, 3424, 3476, 3960, 4290, 4291, 4292, 4295, or 4296) is administered to a Treatment experienced HCV patient in a total formulated concentration of about 900 ug/dose to about 2000 ug/dose once every one, two, three, four or five weeks; about 900 ug/dose to about 1200 ug/dose once every one, two, three, four or five weeks; about 900 ug/dose to about 1800 ug/dose once every one, two, three, four or five weeks; and most preferably in a total formulated concentration of about 1200 ug/dose to about 1800 ug/dose once every one, three, four or five weeks, or most preferably every two weeks in combination with one or more antiviral compounds, including, for example, riba
  • Treatment experienced HCV patient at a total formulated concentration of about 600 ug/dose once every one, two, three, four or five weeks; 800 ug/dose once every one, two, three, four or five weeks, 900 ug/dose once every one, two, three, four or five weeks; 1000 ug/dose once every one, two, three, four or five weeks; 1200 ug/dose once every one, two, three, four or five weeks; 1500 ug/dose once every one, two, three, four or five weeks; 1600 ug/dose once every one, two, three, four or five weeks; 1800 ug/dose once every one, two, three, four or five weeks; or 2000 ug/dose once every one, two, three, four or five weeks in combination with one or more antiviral compounds, including, for example, ribavir
  • an IFN-alpha-HSA fusion protein of the invention e.g., produced by CBDs 2249, 2343, 2366, 2381,
  • Treatment experienced HCV patient at a total formulated concentration of 900 ug/dose once every two weeks, and more preferably at a total concentration of 1200 ug/dose once every two weeks, 1200 ug/dose once every four weeks, or 1800 ug/dose once every four weeks, either alone or in combination with an antiviral compound, " sucl ⁇ 'as ribavirin.
  • an IFN-alpha-HSA fusion protein of the invention (e.g., produced by CIDs 2249, 2343, 2366, 2381, 2382, 2410, 3165, 3422, 3423, 3424, 3476, 3960, 4290, 4291, 4292, 4295, or 4296) is administered to a Treatment experienced HCV patient at a total formulated concentration of 900 ug/dose once every two weeks, and more preferably at a total concentration of 1200 ug/dose once every two weeks, 1200 ug/dose once every four weeks, or 1800 ug/dose once every four weeks, in combination with one or more antiviral compounds, including, for example, ribavirin.
  • Albumin fusion proteins and/or polynucleotides can be are administered orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, gels, drops or transdermal patch), bucally, or as an oral or nasal spray.
  • “Pharmaceutically acceptable carrier” refers to a non-toxic solid, semisolid or liquid filler, diluent, encapsulating material or formulation auxiliary of any.
  • parenteral refers to modes of administration which include intravenous, intramuscular, intraperitoneal, intrasternal, subcutaneous and intraarticular injection and infusion.
  • Albumin fusion proteins and/or polynucleotides of the invention are also suitably administered by sustained-release systems.
  • sustained-release albumin fusion proteins and/or polynucleotides are administered orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, gels, drops or transdermal patch), bucally, or as an oral or nasal spray.
  • “Pharmaceutically acceptable carrier” refers to a non-toxic solid, semisolid or liquid filler, diluent, encapsulating material or formulation auxiliary of any type.
  • parenteral refers to modes of administration which include intravenous, intramuscular, intraperitoneal, intrasternal, subcutaneous and intraarticular injection and infusion.
  • sustained-release albumin fusion proteins and/or polynucleotides include suitable polymeric materials (such as, for example, semi-permeable polymer matrices in the form of shaped articles, e.g., films, or mirocapsules), suitable hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, and sparingly soluble derivatives (such as, for example, a sparingly soluble salt).
  • Sustained-release matrices include polylactides (U.S. Pat. No. 3,773,919, EP 58,481), copolymers of L-glutamic acid and gamma-ethyl-L-glutamate (Sidman et al., Biopolymers 22:547-556 (1983)), poly (2- hydroxyethyl methacrylate) (Langer et al., J. Biomed. Mater. Res. 15:167-277 (1981), and Langer, Chem. Tech. 12:98-105 (1982)), ethylene vinyl acetate (Langer et al., Id.) or poly-D- (-)-3-hydroxybutyric acid (EP 133,988).
  • polylactides U.S. Pat. No. 3,773,919, EP 58,481
  • copolymers of L-glutamic acid and gamma-ethyl-L-glutamate (Sidman et al.
  • Sustained-release albumin fusion proteins and/or polynucleotides also include liposomally entrapped albumin fusion proteins and/or polynucleotides of the invention (see generally, Langer, Science 249:1527-1533 (1990); Treat et al., in Liposomes in the ⁇ ierapy of Infectious Disease and Cancer, Lopez-Berestein and Fidler (eds.), Liss, New York, pp. 317 -327 and 353-365 (1989)).
  • Liposomes containing the albumin fusion protein and/or polynucleotide are prepared by methods known per se: DE 3,218,121; Epstein et al., Proc. Natl. Acad. Sci.
  • the liposomes are of the small (about 200-800 Angstroms) unilamellar type in which the lipid content is greater than about 30 mol. percent cholesterol, the selected proportion being adjusted for the optimal Therapeutic.
  • the albumin fusion proteins and/or polynucleotides of the invention are delivered by way of a pump (see Langer, supra; Sefton, CRC Crit. Ref. Biomed. Eng. 14:201 (1987); Buchwald et al., Surgery 88:507 (1980); Saudek et al., N. Engl. J. Med. 321:574 (1989)).
  • the albumin fusion protein and/or polynucleotide is formulated generally by mixing it at the desired degree of purity, in a unit dosage injectable form (solution, suspension, or emulsion), with a pharmaceutically acceptable carrier, i.e., one that is non-toxic to recipients at the dosages and concentrations employed and is compatible with other ingredients of the formulation.
  • a pharmaceutically acceptable carrier i.e., one that is non-toxic to recipients at the dosages and concentrations employed and is compatible with other ingredients of the formulation.
  • the formulation preferably does not include oxidizing agents and other compounds that are known to be deleterious to the Therapeutic.
  • the formulations are prepared by contacting the albumin fusion protein and/or polynucleotide uniformly and intimately with liquid carriers or finely divided solid carriers or both.
  • the carrier is a parenteral carrier, more preferably a solution that is isotonic with the blood of the recipient.
  • carrier vehicles include water, saline, Ringer's solution, and dextrose solution.
  • Non-aqueous vehicles such as fixed oils and ethyl oleate are also useful herein, as well as liposomes.
  • the carrier suitably contains minor amounts of additives such as substances that enhance isotonicity and chemical stability.
  • additives such as substances that enhance isotonicity and chemical stability.
  • Such materials are non-toxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, succinate, acetic acid, and other organic acids or their salts; antioxidants such as ascorbic acid; low molecular weight (less than about ten residues) polypeptides, e.g., polyarginine or tripeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids, such as glycine, glutamic acid, aspartic acid, or arginine; monosaccharides, disaccharides, and other carbohydrates including cellulose or its derivatives, glucose, manose, or dextrins; chelating agents such as EDTA; sugar alcohols such as mannitol or sorbi
  • Any pharmaceutical used for therapeutic administration can be sterile. Sterility is readily accomplished by filtration through sterile filtration membranes (e.g., 0.2 micron membranes). Albumin fusion proteins and/or polynucleotides generally are placed into a container having a sterile access port, for example, an intravenous solution bag or vial having a stopper pierceable by a hypodermic injection needle.
  • Albumin fusion proteins and/or polynucleotides ordinarily will be stored in unit or multi-dose containers, for example, sealed ampoules or vials, as an aqueous solution or as a lyophilized formulation for reconstitution.
  • the Albumin fusion protein formulations comprises 0.01 M sodium phosphate, 0.15 niM sodium chloride, 0.16 micromole sodium octanoate/milligram of fusion protein, 15 micrograms/milliliter polysorbate 80, pH 7.2.
  • the Albumin fusion protein formulations consists 0.01 M sodium phosphate, 0.15 mM sodium chloride, 0.16 micromole sodium octanoate/milligram of fusion protein, 15 micrograms/milliliter polysorbate 80, pH 7.2.
  • the pH and buffer are chosen to match physiological conditions and the salt is added as a tonicifler.
  • Sodium octanoate has been chosen due to its reported ability to increase the thermal stability of the protein in solution.
  • polysorbate has been added as a generic surfactant, which lowers the surface tension of the solution and lowers non-specific adsorption of the albumin fusion protein to the container closure system.
  • the invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the albumin fusion proteins and/or polynucleotides of the invention.
  • a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the albumin fusion proteins and/or polynucleotides of the invention.
  • Associated with such containers can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration, hi addition, the albumin fusion proteins and/or polynucleotides may be employed in conjunction with other therapeutic compounds.
  • albumin fusion proteins and/or polynucleotides of the invention may be administered alone or in combination with adjuvants.
  • Adjuvants that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, alum, alum plus deoxycholate (ImmunoAg), MTP-PE (Biocine Corp.), QS21 (Genentech, Inc.), BCG (e.g., THERACYS®), MPL and nonviable preparations of Corynebacterium parvum.
  • albumin fusion proteins and/or polynucleotides of the invention are administered in combination with alum.
  • albumin fusion proteins and/or polynucleotides of the invention are administered in combination with QS-21.
  • Further adjuvants that may be administered with the albumin fusion proteins and/or polynucleotides of the. invention include, but-are not limited to,- MonophosphoryHipid immunomodulator; AdjuVax-lOOa, QS-21, QS-18, SRLlOO 5 , Aluminum-salts," MF- 5 9, and Virosomal adjuvant technology.
  • Vaccines that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, vaccines directed toward protection against MMR (measles, mumps, rubella), polio, varicella, tetanus/diptheria, hepatitis A, hepatitis B, Haemophilus influenzae B, whooping cough, pneumonia, influenza, Lyme's Disease, rotavirus, cholera, yellow fever, Japanese encephalitis, poliomyelitis, rabies, typhoid fever, and pertussis.
  • MMR measles, mumps, rubella
  • polio varicella
  • tetanus/diptheria hepatitis A
  • hepatitis B Haemophilus influenzae B
  • whooping cough pneumonia, influenza, Lyme's Disease, rotavirus
  • cholera yellow fever
  • Japanese encephalitis poliomyelitis
  • Combinations may be administered either concomitantly, e.g., as an admixture, separately but simultaneously or concurrently; or sequentially. This includes presentations hi which the combined agents are administered together as a therapeutic mixture, and also procedures in which the combined agents are administered separately but simultaneously, e.g., as through separate intravenous lines into the same individual. Administration "in combination” further includes the separate administration of one of the compounds or agents given first, followed by the second.
  • albumin fusion proteins and/or polynucleotides of the invention may be administered alone or in combination with other therapeutic agents.
  • Albumin fusion protein and/or polynucleotide agents that may be administered in combination with the albumin fusion proteins and/or polynucleotides of the invention include but not limited to, chemotherapeutic agents, antibiotics, steroidal and non-steroidal anti-inflammatories, conventional immunotherapeutic agents, and/or therapeutic treatments described below. Combinations may be administered either concomitantly, e.g., as an admixture, separately but simultaneously or concurrently; or sequentially.
  • Administration "in combination” further includes the separate administration of one of the compounds or agents given first, followed by the second.
  • the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with an anticoagulant.
  • Anticoagulants that may be administered with the compositions of the invention include, but are not limited to, heparin, low molecular weight heparin, warfarin sodium (e.g., COUMADIN®), dicumarol, 4-hydroxycoumarin, anisindione (e.g., MERADONTM), acenocoumarol (e.g., nicoumalone, SINTHROMETM), indan-l,3-dione, phenprocoumon (e.g., MARCUMARTM), ethyl biscoumacetate (e.g., TROMEXANTM), and aspirin.
  • heparin low molecular weight heparin
  • warfarin sodium e.g., COUMADIN®
  • dicumarol e.g., 4-hydroxycoumarin
  • anisindione e.g., MERADON
  • compositions of the invention are administered in combination with heparin and/or warfarin. In another specific embodiment, compositions of the invention are administered in combination with warfarin. In another specific ⁇ ⁇ ⁇ emfiodimeritj compositions ' of' tlie invention are administered in combination with warfarin and aspirin. In another specific embodiment, compositions of the invention are administered in combination with heparin. In another specific embodiment, compositions of the invention are i administered in combination with heparin and aspirin.
  • the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with thrombolytic drugs.
  • thrombolytic drugs that may be administered with the compositions of the invention include, but are not limited to, plasminogen, lys-plasminogen, alpha2-antiplasmin, streptokinae (e.g., KABKJNASETM), antiresplace (e.g., EMINASETM), tissue plasminogen activator (t-PA, altevase, ACTTVASETM), urokinase (e.g., ABBOKINASE 1' '' 1 ), sauruplase, (Prourokinase, single chain urokinase), and aminocaproic acid (e.g., AMICARTM).
  • compositions of the invention are administered in combination with tissue plasminogen activator and aspirin.
  • albumin fusion proteins and/or polynucleotides of the invention are administered in combination with antiplatelet drugs.
  • Antiplatelet drugs that may be administered with the compositions of the invention include, but are not limited to, aspirin, dipyridamole (e.g., PERSANTINETM), and ticlopidine (e.g., TICLIDTM).
  • the use of anti-coagulants, thrombolytic and/or antiplatelet drugs in combination with albumin fusion proteins and/or polynucleotides of the invention is contemplated for the prevention, diagnosis, and/or treatment of thrombosis, arterial thrombosis, venous thrombosis, thromboembolism, pulmonary embolism, atherosclerosis, myocardial infarction, transient ischemic attack, unstable angina.
  • the use of anticoagulants, thrombolytic drugs and/or antiplatelet drugs in combination with albumin fusion proteins and/or polynucleotides of the invention is contemplated for the prevention of occulsion of saphenous grafts, for reducing the risk of periprocedural thrombosis as might accompany angioplasty procedures, for reducing the risk of stroke in patients with atrial fibrillation including nonrheumatic atrial fibrillation, for reducing the risk of embolism associated with mechanical heart valves and or mitral valves disease.
  • occlusions in extracorporeal devices e.g., intravascular canulas, vascular access shunts in hemodialysis patients, hemodialysis machines, and cardiopulmonary bypass machines.
  • albumin fusion proteins and/or polynucleotides of the invention are administered in combination with antiretroviral agents, nucleoside/nucleotide reverse transcriptase inhibitors (NRTIs), non-nucleoside reverse transcriptase inhibitors (NNRTIs), and/or protease inhibitors (PIs).
  • NRTIs nucleoside/nucleotide reverse transcriptase inhibitors
  • NRTIs non-nucleoside reverse transcriptase inhibitors
  • PIs protease inhibitors
  • NRTIs that may be administered in combination with the albumin fusion proteins and/or polynucleotides of the invention, include, but are not limited to, RETROVIRTM (zidovudine/AZT), VIDEXTM (didanosine/ddl), HIVIDTM (zalcitabine/ddC), ZERITTM (stavudine/d4T), EPTVIRTM (lamivudine/3TC), and COMBIVIRTM (zidovudine/lamivudine).
  • RETROVIRTM zidovudine/AZT
  • VIDEXTM didanosine/ddl
  • HIVIDTM zalcitabine/ddC
  • ZERITTM stavudine/d4T
  • EPTVIRTM lamvudine/3TC
  • COMBIVIRTM zidovudine/lamivudine
  • NNRTIs that may be administered in combination with the albumin fusion proteins and/or polynucleotides of the invention, include, but are not limited to, VIRAMUNETM (nevirapine), RESCRIPTORTM (delavirdine), and -SUSTIVATM (efavirenz).
  • - Protease inhibitors thatrmay be administered ' in combination " with the albumin fusion proteins and/or polynucleotides of the invention, include, but are not limited to, CRDOVANTM (indinavir), NORVIRTM (ritonavir), INV1RASETM (saquinavir), and VIRACEPTTM (nelf ⁇ navir).
  • antiretroviral agents nucleoside reverse transcriptase inhibitors, non-nucleoside reverse transcriptase inhibitors, and/or protease inhibitors may be used in any combination with albumin fusion proteins and/or polynucleotides of the invention to treat AIDS and/or to prevent or treat HIV infection.
  • Additional NRTIs include LODENOSINETM (F-ddA; an acid-stable adenosine NRTI; Triangle/Abbott; COVIRACILTM (emtricitabine/FTC; structurally related to lamivudine (3TC) but with 3- to 10-fold greater activity in vitro; Triangle/Abbott); dOTC (BCH-10652, also structurally related to lamivudine but retains activity against a substantial proportion of lamivudine-resistant isolates; Biochem Pharma); Adefovir (refused approval for anti-HIV therapy by FDA; Gilead Sciences); PREVEON® (Adefovir Dipivoxil, the active prodrug of adefovir; its active form is PMEA-pp); TENOFOVIRTM (bis-POC PMPA, a PMPA prodrug; Gilead); DAPD/DXG (active metabolite of DAPD; Triangle/Abbot
  • Additional NNRTIs include COACTINONTM (Emivirine/MKC-442, potent NNRTI of the HEPT class; Triangle/Abbott); CAPRAVIRINETM (AG-1549/S-1153, a next generation NNRTI with activity against viruses containing the K103N mutation; Agouron); PNU- 142721 (has 20- to 5 0-fold greater activity than its predecessor delavirdine and is active against K103N mutants; Pharmacia & Upjohn); DPC-961 and DPC-963 (second-generation derivatives of efavirenz, designed to be active against viruses with the K103N mutation; DuPont); GW-420867X (has 2S-fold greater activity than HBY097 and is active against K103N mutants; Glaxo Wellcome); CALANOLIDE A (naturally occurring agent from the latex tree; active against viruses containing either or both the Yl 81C and K103N mutations); and Propolis (WO 99/49830).
  • COACTINONTM Esmivi
  • protease inhibitors include LOPINAVIRTM (ABT378/r; Abbott Laboratories); BMS-232632 (an azapeptide; Bristol-Myres Squibb); TIPRANAVIRTM (PNU-140690, a non-peptic dihydropyrone; Pharmacia & Upjohn); PD-178390 (a nonpeptidic dihydropyrone; Parke- Davis); BMS 232632 (an azapeptide; Bristol-Myers Squibb); L-7 5 6,423 (an indinavir analog; Merck); DMP-450 (a cyclic urea compound; Avid & Di ⁇ Pont); A" ⁇ '-rM'V'pepiiaom ' ffieWViuT;H vitro activity against protease inhibitor-resistant viruses; Agouron); VX-17 5 /GW-433908 (phosphate prodrug of amprenavir; Vertex & Glaxo Welcome); CGP617 55 (Ciba
  • Additional antiretroviral agents include fusion inhibitors/gp41 binders.
  • Fusion inhibitors/gp41 binders include T-20 (a peptide from residues 643-678 of the HIV gp41 transmembrane protein ectodomain which binds to gp41 in its resting state and prevents transformation to the fusogenic state; Trimeris) and T-1249 (a second-generation fusion inhibitor; Trimeris).
  • Additional antiretroviral agents include fusion inhibitors/chemokine receptor antagonists.
  • Fusion inhibitors/chemokine receptor antagonists include CXCR4 antagonists such as AMD 3100 (a bicyclam), SDF-I and its analogs, and ALX40-4C (a cationic peptide), T22 (an 18 amino acid peptide; Trimeris) and the T22 analogs T134 and T140; CCR 5 antagonists such as RANTES (9-68), AOP-RANTES, NNY-RANTES, and TAK-779; and CCRS/CXCR4 antagonists such as NSC 651016 (a distamycin analog). Also included are CCR2B, CCR3, and CCR6 antagonists. Chemokine recpetor agonists such as RANTES, SDF-I, MIP-Ia, MlP-l ⁇ , etc., may also inhibit fusion.
  • Additional antiretroviral agents include integrase inhibitors.
  • Integrase inhibitors include dicaffeoylquinic (DFQA) acids; L-chicoric acid (a dicaffeoyltartaric (DCTA) acid); quinalizarin (QLC) and related anthraquinones; ZINTEVIRTM (AR 177, an oligonucleotide that probably acts at cell surface rather than being a true integrase inhibitor; Arondex); and naphthols such as those disclosed in WO 98/ 5 0347.
  • DFQA dicaffeoylquinic
  • DCTA dicaffeoyltartaric
  • QLC quinalizarin
  • ZINTEVIRTM AR 177, an oligonucleotide that probably acts at cell surface rather than being a true integrase inhibitor
  • Arondex naphthols such as those disclosed in WO 98/ 5 0347.
  • Additional antiretroviral agents include hydroxyurea-like compunds such as BCX-34 (a purine nucleoside phosphorylase inhibitor; Biocryst); ribonucleotide reductase inhibitors such as DIDOXTM (Molecules for Health); inosine monophosphate dehydrogenase (MPDH) inhibitors sucha as VX-497 (Vertex); and mycopholic acids such as CellCept (mycophenolate mofetil; Roche).
  • BCX-34 purine nucleoside phosphorylase inhibitor
  • Biocryst ribonucleotide reductase inhibitors
  • DIDOXTM Diotide reductase inhibitors
  • MPDH inosine monophosphate dehydrogenase
  • VX-497 Verytex
  • mycopholic acids such as CellCept (mycophenolate mofetil; Roche).
  • Additional antiretroviral agents include inhibitors of viral integrase, inhibitors of viral genome nuclear translocation such as arylene bis(methylketone) compounds; inhibitors of HIV entry such as AOP-RANTES, NNY-RANTES, RANTES-IgG fusion protein, soluble complexes of RANTES and glycosaminoglycans (GAG), and AMD-3100; nucleocapsid zinc finger inhibitors such as dithiane compounds; targets of HIV Tat and Rev; and pharmacoenhancers such as ABT-378.
  • inhibitors of viral integrase inhibitors of viral genome nuclear translocation such as arylene bis(methylketone) compounds
  • inhibitors of HIV entry such as AOP-RANTES, NNY-RANTES, RANTES-IgG fusion protein, soluble complexes of RANTES and glycosaminoglycans (GAG), and AMD-3100
  • nucleocapsid zinc finger inhibitors such as dithiane compounds
  • cytokines and lymphokines such as MIP-Ia, MlP-l ⁇ , SDF-l ⁇ , IL-2, PROLEUKINTM (aldesleukin/L2-7001; Chiron), IL-4, IL-10, IL-12, and IL-13; interferons such as IFN-alpha2a, IFN-alpha2b, or IFN-beta; antagonists of TNFs, NFKB, GM-CSF, M-CSF, and IL-10; agents that modulate immune activation such as cyclosporin and prednisone; vaccines such as RemuneTM (HTV Immunogen), APL 400-003 (Apollon), recombinant gpl20 and fragments, bivalent (B/E) recombinant envelope glycoprotein, rgpl20CM235, MN rgpl20, SF-2 rgpl20, gpl20/soluble CD4 complex
  • antibodies such as the anti-CXCR4 antibody 12G 5 , the anti-CCRS antibodies 2D7, ⁇ 5C7, PA8, PA9, PAlO, PAIl, PA12, and 1 PA14, the anti-CD4 antibodies Q4120 and RPA-T4, the anti-CCR3 antibody 7Bl 1, the anti-gpl20 antibodies 17b, 48d, 447- 5 2D, 2 5 7-D, 268-D and 50.1, anti-Tat antibodies, anti-TNF- ⁇ antibodies, and monoclonal antibody 33A; aryl hydrocarbon (AH) receptor agonists and antagonists such as TCDD, 3,3',4,4', 5 -pentachlorobiphenyl, 3,3',4,4'- tetrachlorobiphenyl, and ⁇ -naphthoflavone (WO 98/30213); and antioxidants such as ⁇ -L-glutamyl-L-cysteine ethyl ester (AB 98/30213);
  • the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with one or more antiviral agent.
  • Antiviral agents that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, acyclovir, ribavirin, ribavirin analog, amantadine, remantidine, maxamine, or thymalfasin.
  • interferon albumin fusion protein can be administered in combination with any of these agents.
  • interferon alpha albumin fusion protein can also be admistered with any of these agents, and preferably, interferon alpha 2a or 2b albumin fusion protein can be administered with any of these agents.
  • interferon beta albumin fusion protein can also be admistered with any of these agents.
  • any of the IFN hybrids albumin fusion proteins can be administered in combination with any of these agents.
  • an interferon albumin fusion protein of the invention is administered in combination with ribavirin or a ribavirin analog
  • the ribavirin or ribavirin analogs that may be administered in combination with an interferon albumin fusion protein include but are not limited to COPEGUS ® (Hoffman-La Roche, Nutley, N.J.), REBETOL ® (Schering Corp., Kenilworth, N.J.), VIRAZOLE ® (Valeant, Costa Mesa, CA), RBAVINTM (Lupin, Baltimore, MD), RIBAZIDTM (EpIa, Kirachi, Pakistan), tribavirin, VIRAMIDINETM (Valeant, Costa Mesa, CA), and RBASPHERETM (Three Rivers Pharmaceuticals, Cranberry Township, PA).
  • interferon alpha albumin fusion protein is administered in combination with ribavirin or ribavirin analog
  • interferon alpha 2a albumin fusion protein is administered in combination with ribavirin or ribavirin analog.
  • interferon alpha 2b albumin fusion protein is administered in combination with ribavirin or ribavirin analog.
  • interferon beta albumin fusion protein is administered in combination with ribavirin or ribavirin analog.
  • hybrid interferon albumin fusion protein is administered in combination with ribavirin or ribavirin analog.
  • the albumin fusion proteins and/or polynucleotides of the invention may be administered alone or in " ' "
  • an interferon-albumin fusion protein of the invention may be administered in combination with one or more antiviral agents.
  • the viral infection results from infection with a hepatitis virus.
  • the hepatitis virus is hepatitis C virus (HCV).
  • Antiviral agents that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, small-molecule inhibitors of viral enzymes, small-molecule inhibitors of RNA polymerase, nucleic acid based antiviral agents, antisense oligonucleotide inhibitors, thiazolides, novel immunomodulatory agents, and interferon enhancers.
  • Anti-viral enzyme inhibitors that may be administered in combination with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, VX-9S0 (protease inhibitor, Vertex, Cambridge, MA), VX-497 (merimepodib, oral IMPDH inhibitor, Vertex, Cambridge, MA), BILB 1941 (protease inhibitor, Boehringer Ingelheim, Germany), SCH7 (protease inhibitor, Schering Corp., Kenilworth, N.J.), MX-3253 (glucosidase inhibitor, Migenix, Vancouver, BC), IDN-6556 (caspase inhibitor, Pfizer, New York, NY), UT231B (glucosidase inhibitor, United Therapeutics, Silver Spring, MD), R1626 (viral protease inhibitor, F.
  • Anti-viral polymerase inhibitors that may be administered in combination with the albumin fusion proteins and/or polynucleotides of the invention may be nucleoside analogs or non-nucleoside inhibitors (NNIs).
  • the anti-viral polymerase inhibitors inhibit HCV RNA polymerase.
  • the anti-viral polymerase inhibitors may be nucleoside analogs including, but not limited to, NM283 (oral prodrug of 23'-C-methyl-cytidine, Idenix, Cambride, MA), and 2'-C-methyl nucleosides.
  • the anti-viral polymerase inhibitors may be non-nucleoside inhibitors, including, but not limited to, JTK-103, JTK-003, and JTK-109 (Japan Tabacco, Tokyo, Japan), R8O3 (Rigel, South San Francisco, CA), HCV-371, HCV-086, and HCV-79 ⁇ (ViroPharm, Exton, PA / Wyeth, Madison, NJ), and XTL-2125 (BC2125, XTLbio, New York, NY).
  • Anti-viral nucleic acid based agents that may be administered in combination with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, antisense oligonucleotides, ribozymes, and siRNAs or short hairpin RNAs (shRNA).
  • Anti-viral antisense oligonucleotide inhibitor agents that may be administered in combination with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, NEUGENE ® AVI-4065 (AVI Biopharma, Portland, OR).
  • a thiazolide may be administered in combination with the albumin fusion proteins and/or polynucleotides of the invention.
  • thiazolides that may be administered in combination with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to ALINIA ® (nitazoxanide, Romark Laboratories, L.C., Tampa, FL).
  • Anti-viral immunomodulatory agents that may be administered in combination with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, ZADXIN ® (thymosin alpha 1, thymalfasin, SciClone Pharmaceuticals Int'l, Hong Kong) and toll-like receptor (TLR) agonists, including, but not limited to, ANA24 5 (TLR-7 agonist, Anadys Pharmaceuticals, San Diego, CA), ANA975 (oral prodrug of ANA245, Anadys Pharmaceuticals, San Diego, CA), and. CPG-10101 (ACTJLONTM, TLR-9 agonist, Coley-Pharmaceutical Group, Wellesley, MA).
  • ZADXIN ® thymosin alpha 1, thymalfasin, SciClone Pharmaceuticals Int'l, Hong Kong
  • TLR toll-like receptor
  • ANA24 5 TLR-7 agonist, Anadys Pharmaceuticals, San Diego, CA
  • Interferon enhancers that may be administered in combination with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to EMZ702 (Transition Therapeutics, Toronto, Ontario).
  • anti-viral antibodies that may be administered in combination with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to Tarvacin (humanized monoclonal antibody that targets phosphatidylserine on the surface of tumor endothelial cells, Peregrine Pharmaceuticals, Inc., Tustin, CA).
  • the albumin fusion protein that may be administered alone or in combination with one or more of the antiviral agents encompassed by the invention is an inteferon-albumin fusion protein.
  • the interferon portion of the interferon- albumin fusion protein is an interferon alpha.
  • Non-limiting examples of interferon alpha encompassed by the invention include, but are not limited to, the interferon alpha proteins disclosed in the Therapeutic protein column of Table 1.
  • the interferon alpha portion consists or alternatively comprises interferon alpha-2a, interferon alpha-2b, interferon al ⁇ ha-2c, consensus interferon, interferon alfacon-1, interferon alpha-nl, interferon alpha-n3, any commercially available form of interferon alpha, such as, for example, INTRON ® A (Schering Corp., Kenilworth, NJ.
  • ROFERON ® A Hoffman-La Roche, Nutley, NJ.
  • Berofor alpha inteferon Boehringer Ingelheim Pharmaceutical, Inc., Ridgefied, Conn.
  • OMNIFERONTM (Viragen, Inc., Plantation, FL)
  • MULTIFERONTM (Viragen, Inc., Plantation, FL)
  • WELLFERON ® (GlaxoSmithKline, London, Great Britian)
  • INFERGEN ® Amgen, Inc., Thousands Oaks, CA
  • SUMIFERON ® Sumitomo, Japan
  • BELEROFON ® Nautilus Biotech, France
  • MAXY-ALPHATM Maxygen, Redwood City, CA / Hoffinan-La Roche, Nutley, NJ.
  • MAXY-ALPHATM Maxygen, Redwood City, CA / Hoffinan-La Roche, Nutley, NJ.
  • the interferon alpha portion of the IFN-alpha-HSA fusion protein consists or alternatively comprises interferon alpha modified or formulated for extended or controlled release.
  • the interferon alpha portion consists, or alternatively comprises commercially available extended release or controlled release interferon alpha, including, but not limited to interferon-alpha-XL (Flamel Technologies, France) and LOCTERONTM (BioLex Therapeutics/OctoPlus, Pittsboro, NC).
  • the interferon alpha portion of the IFN-alpha-HSA fusion protein may be modified by the attachment of chemical moieties.
  • the inteferon alpha portion may be modified by pegylation.
  • the interferon alpha portion of the IFN-alpha-HSA fusion protein consists or alternatively comprises pegylated forms of interferon alpha-2a, 2b, or consensus interferon and include, but are not limited to, a commercially available pegylated interferon alpha, such as, for example, PEG-INTRON ® (Schering Corp., Kenilworth, NJ.), PEGASYS ® (Hoffman-La Roche, Nutley "1 NX), PECMMN ⁇ FER ⁇ TM'(V ⁇ ragen, Inc., Plantation, FL) or a fragment thereof.
  • the interferon portion of the albumin fusion protein is interferon alpha 2a or 2b interferon, interferon albumin fusion protein can be administered in combination with any of these agents.
  • the interferon portion of the interferon-albumin fusion protein is an interferon beta or an interferon hybrids.
  • the unfused interferon portion of the inteferon-albumin fusion protein may be used alone or in combination with one or more of the antiviral agents encompassed by the invention.
  • albumin fusion proteins and/or polynucleotides of the invention may be administered in combination with anti- opportunistic infection agents.
  • Anti-opportunistic agents that may be administered in combination with the albumin fusion proteins and/or polynucleotides of the invention, include, but are not limited to, TRIMETHOPRIM-SULFAMETHOXAZOLETM, DAPSONETM, PENTAMIDINETM,
  • albumin fusion proteins and/or polynucleotides of the invention are used in any combination with TRIMETHOPRIM-SULFAMETHOXAZOLETM, DAPSONETM, PENTAMIDINETM, and/or ATOVAQUONETM to prophylactically treat or prevent an opportunistic Pneumocystis carinii pneumonia infection.
  • albumin fusion proteins and/or polynucleotides of the invention are used in any combination with ISONIAZIDTM, RIFAMPINTM, PYRAZINAMIDETM, and/or ETHAMBUTOLTM to prophylactically treat or prevent an opportunistic Mycobacterium avium complex infection.
  • albumin fusion proteins and/or polynucleotides of the invention are used in any combination with RIFABUTINTM, CLARITHROMYCINTM, and/or AZITHROMYCINTM to prophylactically treat or prevent an opportunistic Mycobacterium tuberculosis infection.
  • albumin fusion proteins and/or polynucleotides of the invention are used in any combination with GANCICLOVIRTM, FOSCARNETTM, and/or CIDOFOVIRTM to prophylactically treat or prevent an opportunistic cytomegalovirus infection.
  • albumin fusion proteins and/or polynucleotides of the invention are used in any combination with FLUCONAZOLETM, ITRACONAZOLETM, and/or KETOCONAZOLETM to prophylactically treat or prevent an opportunistic fungal infection.
  • albumin fusion proteins and/or polynucleotides of the invention are used in any combination with ACYCLOVIRTM and/or FAMCICOLVIRTM to prophylactically treat or prevent an opportunistic herpes simplex virus type I and/or type ⁇ infection.
  • albumin fusion proteins and/or polynucleotides of the invention are used in any combination with PYRIMETHAMINETM and/or LEUCOVORINTM to prophylactically treat or prevent an opportunistic Toxoplasma gondii infection.
  • albumin fusion proteins and/or polynucleotides of tire invention are used in any combination with LEUCOVORINTM and/or NEUPOGENTM to prophylactically treat or prevent an opportunistic bacterial infection.
  • the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with an antibiotic agent.
  • Antibiotic agents that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, amoxicillin, beta-lactamases, aminoglycosides, beta-lactam (glycopeptide), beta-lactamases, Clindamycin, chloramphenicol, cephalosporins, ciprofloxacin, erythromycin, fluoroquinolones, macrolides, metronidazole, penicillins, quinolones, rapamycin, rifampin, streptomycin, sulfonamide, tetracyclines, trimethoprim, trimethoprim-sulfamethoxazole, and vancomycin.
  • the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with immunestimulants.
  • Immunostimulants that may be administered in combination with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, levamisole (e.g., ERGAMISOLTM), isoprinosine (e.g. INOSIPLEXTM), interferons (e.g. interferon alpha), and interleukins (e.g., IL-2).
  • albumin fusion proteins and/or polynucleotides of the invention are administered in combination with immunosuppressive agents.
  • Immunosuppressive agents that may be administered in combination with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, steroids, cyclosporine, cyclosporine analogs, cyclophosphamide methylprednisone, prednisone, azathioprine, FK- 5 06, l 5 -deoxyspergualin, and other immunosuppressive agents that act by suppressing the function of responding T cells.
  • immunosuppressive agents that may be administered in combination with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, prednisolone, methotrexate, thalidomide, methoxsalen, rapamycin, leflunomide, mizoribine (BREDININTM), brequinar, deoxyspergualin, and azaspirane (SKF 105685), ORTHOCLONE OKT® 3 (muromonab-CD3), SANDIMMUNETM, NEORALTM, SANGDYATM (cyclosporine), PROGRAF® (FK506, tacrolimus), CELLCEPT® (mycophenolate motefil, of which the active metabolite is mycophenolic acid), IMURANTM (azathioprine), glucocorticosteroids, adrenocortical steroids such as DELTASONETM (prednisone) and HYDELTRASOLTM (predni
  • albumin fusion proteins and/or polynucleotides of the invention are administered alone or in combination with one or more intravenous immune globulin preparations.
  • Intravenous immune globulin preparations that may be administered with the albumin polynucleotides '" of tlie invention include, but not limited to, GAMMARTM, IVEEGAMTM, SANDOGLOBULINTM, GAMMAGARD S/DTM, ATGAMTM (antithymocyte glubulin), and GAMIMUNETM.
  • albumin fusion proteins and/or polynucleotides of the invention are administered in combination with intravenous immune globulin preparations in transplantation therapy (e.g., bone marrow transplant).
  • the albumin fusion proteins and/or polynucleotides of the invention are administered alone or as part of a combination therapy, either in vivo to patients or in vitro to cells, for the treament of cancer.
  • the albumin fusion proteins, particularly IL-2-albumin fusions are administered repeatedly during passive immunotherapy for cancer, such as adoptive cell transfer therapy for metastatic melanoma as described in Dudley et al (Science Express, 19 September 2002., at www.scienceexpress.org, hereby incorporated by reference in its entirety).
  • the albumin fusion proteins and/or polynucleotides of tlie invention are administered alone or in combination with an anti-inflammatory agent.
  • Anti-inflammatory agents that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, corticosteroids (e.g.
  • compositions of the invention are administered alone or in combination with an anti-angiogenic agent.
  • Anti-angiogenic agents that may be administered with the compositions of the invention include, but are not limited to, Angiostatin (Entremed, Rockville, MD), Troponin-1 (Boston Life Sciences, Boston, MA), anti-Invasive Factor, retinoic acid and derivatives thereof, paclitaxel (Taxol), Suramin, Tissue Inhibitor of Metalloproteinase-l, Tissue Inhibitor of Metalloproteinase-2, VEGI, Plasminogen Activator Inhibitor-1, Plasminogen Activator Inhibitor-2, and various forms of the lighter "d group" transition metals.
  • Lighter "d group” transition metals include, for example, vanadium, molybdenum, tungsten, titanium, niobium, and tantalum species. Such transition metal species may form transition metal complexes. Suitable complexes of the above-mentioned transition metal species include oxo transition metal complexes.
  • vanadium complexes include oxo vanadium complexes such as vanadate and vanadyl complexes.
  • Suitable - vanadate complexes include metavanadate and orthovanadate complexes -such as, ' for example, ammonium metavanadate, sodium metavanadate, ' and sodium orthovanadate.
  • Suitable vanadyl complexes include, for example, vanadyl acetylacetonate and vanadyl sulfate including vanadyl sulfate hydrates such as vanadyl sulfate mono- and trihydrates.
  • tungsten and molybdenum complexes also include oxo complexes.
  • Suitable oxo tungsten complexes include tungstate and tungsten oxide complexes.
  • Suitable tungstate complexes include ammonium tungstate, calcium tungstate, sodium tungstate dihydrate, and tungstic acid.
  • Suitable tungsten oxides include tungsten (IV) oxide and tungsten (VI) oxide.
  • Suitable oxo molybdenum complexes include molybdate, molybdenum oxide, and molybdenyl complexes.
  • Suitable molybdate complexes include ammonium molybdate and its hydrates, sodium molybdate and its hydrates, and potassium molybdate and its hydrates.
  • Suitable molybdenum oxides include molybdenum (VT) oxide, molybdenum (VI) oxide, and molybdic acid.
  • Suitable molybdenyl complexes include, for example, molybdenyl acetylacetonate.
  • Other suitable tungsten and molybdenum complexes include hydroxo derivatives derived from, for example, glycerol, tartaric acid, and sugars.
  • anti-angiogenic factors include, but are not limited to, platelet factor 4; protamine sulphate; sulphated chitin derivatives (prepared from queen crab shells), (Murata et al., Cancer Res.
  • Sulphated Polysaccharide Peptidoglycan Complex (SP- PG) (the function of this compound may be enhanced by the presence of steroids such as estrogen, and tamoxifen citrate); Staurosporine; modulators of matrix metabolism, including for example, proline analogs, cishydroxyproline, d,L-3,4-dehydroproline, Thiaproline, alpha,alpha-dipyridyl, aminopropionitrile fumarate; 4-propyl-5-(4-pyridinyl)- 2(3H)-oxazolone; Methotrexate; Mitoxantrone; Heparin; Interferons; 2 Macroglobulin-serum; ChIMP-3 (Pavloff et al., J.
  • SP- PG Sulphated Polysaccharide Peptidoglycan Complex
  • Thalidomide (Celgene, Warren, NJ); Angiostatic steroid; AGM-1470 (H. Brem and J. Folkman J Pediatr. Surg.
  • an integrin alpha v beta 3 antagonist C ' [ St ⁇ rgard et ' al.”'/fc//n:"7 «vbri03 f :47-54 (1999)); carboxynaminolmidazole; Carboxyamidotriazole (CAI) (National Cancer Institute, Bethesda, MD); Conbretastatin A-4 (CA4P) (OXiGENE, Boston, MA); Squalamine (Magainin Pharmaceuticals, Madison Meeting, PA); TNP-470, (Tap Pharmaceuticals, Deerf ⁇ eld, BL); ZD-OlQl AstraZeneca (London, UK); APRA (CT2 5 84); Benefin, Byrostatin-1 (SC3395 55 ); CGP-41251 (PKC 412); CMlOl; Dexrazoxane (ICRF187); DMXAA; Endostatin; Flavopridiol; Genestein; GTE;
  • Anti-angiogenic agents that may be administed in combination with the compounds of the invention may work through a variety of mechanisms including, but not limited to, inhibiting proteolysis of the extracellular matrix, blocking the function of endothelial cell-extracellular matrix adhesion molecules, by antagonizing the function of angiogenesis inducers such as growth factors, and inhibiting integrin receptors expressed on proliferating endothelial cells.
  • anti-angiogenic inhibitors that interfere with extracellular matrix proteolysis and which may be administered in combination with the compositons of the invention include, but are not limited to, AG-3340 (Agouron, La Jolla, CA), BAY-12-9U66 (Bayer, West Haven, CT), BMS-27 5 291 (Bristol Myers Squibb, Princeton, NJ), CGS-27032A (Novartis, East Hanover, NJ), Marimastat (British Biotech, Oxford, UK), and Metastat (Aeterna, St-Foy, Quebec).
  • anti-angiogenic inhibitors that act by blocking the function of endothelial cell-extracellular matrix adhesion molecules and which may be administered in combination with the compositons of the invention include, but are not limited to, EMD-121974 (Merck KcgaA Darmstadt, Germany) and Vitaxin (Ixsys, La Jolla, CA/Medimmune, Gaithersburg, MD).
  • anti-angiogenic agents that act by directly antagonizing or inhibiting angiogenesis inducers and which may be administered in combination with the compositons of the invention include, but are not limited to, Angiozyme (Ribozyme, Boulder, CO), Anti-VEGF antibody (Genentech, S.
  • SU-IOl Sud Francisco, S. San Francisco, CA
  • SU- 5 416 Sugen/ Pharmacia Upjohn, Bridgewater, NJ
  • SU-6668 Sugen
  • Other anti-angiogenic agents act to indirectly inhibit angiogenesis.
  • indirect inhibitors of angiogenesis which may be administered in combination with the compositons of the invention include, but are not limited to, IM-862 (Cytran, Kirkland, WA), Interferon-alpha, IL-12 (Roche, Nutley, NJ), and Pentosan polysulfate (Georgetown University, Washington, DC).
  • compositions of the invention in combination with anti-angiogenic agents is contemplated for the treatment, prevention, and/or amelioration of an autoimmune disease, such as for example, an autoimmune disease described herein.
  • an autoimmune disease such as for example, an autoimmune disease described herein.
  • the use of compositions of the invention in combination with anti-angiogenic agents is contemplated for the treatment, prevention, and/or amelioration of arthritis.
  • the use of compositions of the invention in combination with anti-angiogenic agents is contemplated for the treatment, prevention, and/or amelioration of rheumatoid arthritis.
  • the polynucleotides encoding a polypeptide of the present invention are administered in combination with an angiogenic protein, or polynucleotides encoding an angiogenic protein.
  • angiogenic proteins include, but are not limited to, acidic and basic fibroblast growth factors, " VEGF-l, " VEGF-2; VEGF-3, epidermal growth factor alpha and beta, platelet-derived endothelial cell growth factor, platelet-derived growth factor, tumor necrosis factor alpha, hepatocyte growth factor, insulin-like growth factor, colony stimulating factor, macrophage colony stimulating factor, granulocyte/macrophage colony stimulating factor, and nitric oxide synthase.
  • compositions of the invention are administered in combination with a chemotherapeutic agent.
  • Chemotherapeutic agents that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to alkylating agents such as nitrogen mustards (for example, Mechlorethamine, cyclophosphamide, Cyclophosphamide Ifosfamide, Melphalan (L-sarcolysin), and Chlorambucil), ethylenimines and methylmelamines (for example, Hexamethylmelamine and Thiotepa), alkyl sulfonates (for example, Busulfan), nitrosoureas (for example, Carmustine (BCNU), Lomustine (CCNU), Semustine (methyl-CCNU), and Streptozocin (streptozotocin)), triazenes (for example, dacarbazine (DTIC; dimethyltriazenoimidazo
  • compositions of the invention are administered in combination with one or more of the following drugs: infliximab (also known as RemicadeTM Centocor, Inc.), Trocade (Roche, RO-32-3 555 ), Leflunomide (also known as AravaTM from Hoechst Marion Roussel), KineretTM (an IL-I Receptor antagonist also known as Anakinra from Amgen, Inc.)
  • infliximab also known as RemicadeTM Centocor, Inc.
  • Trocade Roche, RO-32-3 555
  • Leflunomide also known as AravaTM from Hoechst Marion Roussel
  • KineretTM an IL-I Receptor antagonist also known as Anakinra from Amgen, Inc.
  • compositions of the invention are administered in combination with CHOP (cyclophosphamide, doxorubicin, vincristine, and prednisone) or combination of one or more of the components of CHOP.
  • CHOP cyclophosphamide, doxorubicin, vincristine, and prednisone
  • the compositions of the invention are administered in combination with anti-CD20 antibodies, human monoclonal anti-CD20 antibodies.
  • the compositions of the invention are administered in combination with anti-CD20 antibodies and CHOP, or anti-CD20 antibodies and any combination of one or more of the components of CHOP, particularly cyclophosphamide and/or prednisone.
  • compositions of the invention are administered in combination with Rituximab.
  • compositions of the invention are administered with Rituximab and CHOP, or Rituximab and any combination of one or more of the components of CHOP, particularly cyclophosphamide and/or prednisone.
  • compositions of the invention are administered in combination with tositumomab.
  • compositions of the invention are administered with tositumomab and CHOP, or tositumomab and any combination of one or more of the components of CHOP, particularly cyclophosphamide and/or prednisone.
  • the anti-CD20 antibodies may optionally be associated with radioisotopes, toxins or cytotoxic prodrugs.
  • compositions of the invention are administered in combination ZevalinTM.
  • compositions of the invention are administered with ZevalinTM and CHOP, or ZevalinTM and any combination of one or more of the components of CHOP, particularly cyclophosphamide and/or prednisone.
  • ZevalinTM may be associated with one or more radisotopes. Particularly preferred isotopes are 90 Y and 111 In.
  • the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with cytokines.
  • Cytokines that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, IL2, EL3, DL4, EL 5 , IL 6 , IL7, ILlO, EL12, IL13, IL15, anti-CD40, CD40L, IFN-gamma and TNF-alpha.
  • albumin fusion proteins and/or polynucleotides of the invention may be administered with any interleukin, including, but not limited to, IL-lalpha, IL-lbeta, IL-2, IL-3, IL-4, IL-5, IL-6, JL-I, JL-S, JL-9, IL-10, IL-Il, IL-12, IL-13, IL-14, IL-I 5 , IL-16, IL- 17, 1L-18, IL-19, IL-20, and EL-21.
  • the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with members of the TNF family.
  • TNF, TNF-related or TNF-like molecules that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, soluble forms of TNF-alpha, lymphotoxin-alpha (LT-alpha, also known as TNF-beta), LT-beta (found in complex heterotrimer LT-alpha2-beta), OPGL, FasL, CD27L, CD30L, CD40L, 4-1BBL, DcR3, OX40L, TNF-gamma (International Publication No. WO 96/14328), AIM-I (International Publication No. WO 97/33899), endokine-alpha (International Publication No.
  • WO 98/07880 OPG, and ' neutrokine-alpha
  • International Publication No/WO 98/18921, ' OX40, and nerve growth factor (NGF) " arid soluble forms " of Fas " , CD30, " CD27, CD40 and 4-D3B
  • TR2 International Publication No. WO 96/3409 5
  • DR3 International Publication No. WO 97/33904
  • DR4 International Publication No. WO 98/328 5 6
  • TR 5 International Publication No. WO 98/30693
  • TRANK TR9 (International Publication No. WO 98/ 5 6892),TR10 (International Publication No. WO 98/ 5 4202), 312C2 (International Publication No. WO 98/06842), and TR12, and soluble forms CD1 5 4, CD70, and CD1 5 3.
  • the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with angiogenic proteins.
  • Angiogenic proteins that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, Glioma Derived Growth Factor (GDGF), as disclosed in European Patent Number EP-399816; Platelet Derived Growth Factor-A (PDGF-A), as disclosed in European Patent Number EP-682110; Platelet Derived Growth Factor-B (PDGF-B), as disclosed in European Patent Number EP-282317; Placental Growth Factor (PlGF), as disclosed in International Publication Number WO 92/06194; Placental Growth Factor-2 (P1GF-2), as disclosed in Hauser et al., Growth Factors, 4:2 5 9-268 (1993); Vascular Endothelial Growth Factor (VEGF), as disclosed in International Publication Number WO 90/13649; Vascular Endothelial Growth
  • the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with Fibroblast Growth Factors.
  • Fibroblast Growth Factors that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, FGF-I, FGF-2, FGF-3, FGF-4, FGF-5, FGF-6, FGF-7, FGF-8, FGF-9, FGF-IO, FGF-Il, FGF-12, FGF-13, FGF-14, and FGF-15.
  • the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with it Arlington ⁇ • ⁇ .., ⁇ t ;TM_B ii,,.ii Diente ⁇ n::::; "an zi »j- ⁇ ,:J
  • Hematopoietic growth factors that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, granulocyte macrophage colony stimulating factor (GM-CSF) (sargramostim, LEUKINETM, PROKINETM), granulocyte colony stimulating factor (G-CSF) (filgrastim, NEUPOGENTM), macrophage colony stimulating factor (M-CSF, CSF-I) erythropoietin (epoetin alfa, EPOGENTM, PROCRTTTM), stem cell factor (SCF, c-kit ligand, steel factor), megakaryocyte colony stimulating factor, PKY321 (a GMCSF/IL-3 fusion protein), interleukins, especially any one or more of IL-I through IL-12, interferon-gamma, or thrombopoietin.
  • GM-CSF granulocyte macrophage colony stimulating factor
  • albumin fusion proteins and/or polynucleotides of the present invention are administered in combination with adrenergic blockers, such as, for example, acebutolol, atenolol, betaxolol, bisoprolol, carteolol, Iabetalol, metoprolol, nadolol, oxprenolol, penbutolol, pindolol, propranolol, sotalol, and timolol.
  • adrenergic blockers such as, for example, acebutolol, atenolol, betaxolol, bisoprolol, carteolol, Iabetalol, metoprolol, nadolol, oxprenolol, penbutolol, pindolol, propranolol, sotalol, and timolol.
  • the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with an antiarrhythmic drug (e.g., adenosine, amidoarone, bretylium, digitalis, digoxin, digitoxin, diliazem, disopyramide, esmolol, flecainide, lidocaine, mexiletine, moricizine, phenytoin, procainamide, N-acetyl procainamide, propafenone, propranolol, quinidine, sotalol, tocainide, and verapamil).
  • an antiarrhythmic drug e.g., adenosine, amidoarone, bretylium, digitalis, digoxin, digitoxin, diliazem, disopyramide, esmolol, flecainide, lidocaine, mexiletine, moricizine, phenytoin, procainamide,
  • the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with diuretic agents, such as carbonic anhydrase-inhibiting agents (e.g., acetazolamide, dichlorphenamide, and methazolamide), osmotic diuretics (e.g., glycerin, isosorbide, mannitol, and urea), diuretics that inhibit Na + -K + -2C1 " symport (e.g., furosemide, bumetanide, azosemide, piretanide, tripamide, ethacrynic acid, muzolimine, and torsemide), thiazide and thiazide-like diuretics (e.g., bendroflumethiazide, benzthiazide, chlorothiazide, hydrochlorothiazide, hydroflumethiazide, methyclothia
  • diuretic agents such
  • the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with treatments for endocrine and/or hormone imbalance disorders.
  • Treatments for endocrine and/or hormone imbalance disorders include, but are not limited to, 127 I, radioactive isotopes of iodine such as 131 I and 123 I; recombinant growth hormone, such as HUMATROPETM (recombinant somatropin); growth hormone analogs such as PROTROPINTM (somatrem); dopamine agonists such as PARLODELTM (bromocriptine); somatostatin analogs such as SANDOSTATINTM (octreotide); gonadotropin preparations such as PREGNYLTM, A.P.L.TM and PROFASF" (chorionic gonadotropin (CG)), PERGONALTM (menotropins), and METRODMTM (urofollitropin (uFSH)); synthetic human gonadotropin releasing hormone
  • Additional treatments for endocrine and/or hormone imbalance disorders include, but are not limited to, estrogens or congugated estrogens such as ESTRACETM (estradiol), ESTINYLTM (ethinyl estradiol), PREMARINTM, ESTRATABTM, ORTHO-ESTTM, OGENTM and estropipate (estrone), ESTROVISTM (quinestrol), ESTRADERMTM (estradiol), DELESTROGENTM and VALERGENTM (estradiol valerate), DEPO- ESTRADIOL CYPIONATETM and ESTROJECT LATM (estradiol cypionate); antiestrogens such as NOLVADEXTM (tamoxifen), SEROPHENETM and CLOMEDTM (clomiphene); progestins such as DURALUTINTM (hydroxyprogesterone caproate), MPATM and DEPO-PROVERATM (medroxyprogesterone acetate), PROVERATM and
  • Additional treatments for endocrine and/or hormone imbalance disorders include, but are not limited to, testosterone esters such as methenolone acetate and testosterone undecanoate; parenteral and oral androgens such as TESTOJECT- 5 0TM (testosterone), TESTEXTM (testosterone propionate), DELATESTRYLTM (testosterone enanthate), DEPO-TESTOSTERONETM (testosterone cypionate), DANOCRINETM (danazol),
  • testosterone esters such as methenolone acetate and testosterone undecanoate
  • parenteral and oral androgens such as TESTOJECT- 5 0TM (testosterone), TESTEXTM (testosterone propionate), DELATESTRYLTM (testosterone enanthate), DEPO-TESTOSTERONETM (testosterone cypionate), DANOCRINETM (danazol)
  • HALOTESTINTM fluoxymesterone
  • ORETON METHYLTM TESTREDTM and VIRILONTM (methyltestosterone), and OXANDRINTM (oxandrolone)
  • testosterone transdermal systems such as TESTODERMTM
  • androgen receptor antagonist and 5 -alpha-reductase inhibitors such as ANDROCURTM (cyproterone acetate), EULEXINTM (flutamide), and PROSCARTM (finasteride)
  • adrenocorticotropic hormone preparations such as CORTROSYNTM (cosyntropin); adrenocortical steroids and their synthetic analogs such as ACLOVATETM (alclometasone dipropionate), CYCLOCORTTM (ameinonide), BECLOVENTTM and VANCERILTM (beclomethasone dipropionate), CELESTONETM (betamethasone), BENISONETM and UTICORTTM (betamethasone
  • the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with treatments " for uterine motility disorders.
  • Treatments for uterine motility disorders include, but are not limited to, estrogen drugs such as conjugated estrogens (e.g., PREMARIN ® and ESTRATAB ® ), estradiols (e.g., CLMARA ® and ALORA ® ), estropipate, and chlorotrianisene; progestin drugs (e.g., AMEN ® (medroxyprogesterone), MICRONOR ® (norethidrone acetate), PROMETRIUM ® progesterone, and megestrol acetate); and estrogen/progesterone combination therapies such as, for example, conjugated estrogens/medroxyprogesterone (e.g., PREMPROTM and PREMPHASE ® ) and norethindrone acetate/ethinyl estsradio
  • the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with drugs effective in treating iron deficiency and hypochromic anemias, including but not limited to, ferrous sulfate (iron sulfate, FEOSOLTM), ferrous fumarate (e.g., FEOSTATTM), ferrous gluconate (e.g., FERGONTM), polysaccharide-iron complex (e.g., NIFEREXTM), iron dextran injection (e.g., INFEDTM), cupric sulfate, pyroxidine, riboflavin, Vitamin Bi 2 , cyancobalamin injection (e.g., REDISOLTM, RUBRAMIN PCTM), hydroxocobalamin, folic acid (e.g., FOLVITETM), leucovorin (folinic acid, 5 -CHOH4PteGlu, citrovorum factor) or WELLCOVORIN (Cal
  • the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with agents used to treat psychiatric disorders.
  • Psychiatric drugs that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, antipsychotic agents (e.g., chlorpromazine, chlorprothixene, clozapine, fluphenazine, haloperidol, loxapine, mesoridazine, molindone, olanzapine, perphenazine, pimozide, quetiapine, risperidone, thioridazine, thiothixene, trifluoperazine, and triflupromazine), antimanic agents (e.g., carbamazepine, divalproex sodium, lithium carbonate, and lithium citrate), antidepressants (e.g., amitriptyline, amoxa
  • antipsychotic agents
  • the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with agents H .' i
  • Neurological agents that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, antiepileptic agents (e g , carbamazepine, clonazepam, ethosuximide, phenobarbital, phenytoin, primidone, valproic acid, divalproex sodium, felbamate, gabapentin, lamotrigine, Ievetiracetam, oxcarbazepme, tiagabine, topiramate, zonisamide, diazepam, lorazepam, and clonazepam), antiparkinsonian
  • the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with treatments for gastrointestinal disorders
  • Treatments for gastrointestinal disorders that may be administered with the albumin fusion protein and/or polynucleotide of the invention include, but are not limited to, H 2 histamine receptor antagonists (e g , TAGAMETTM (cimetidine), ZANTACTM (ranitidine), PEPCIDTM (famotidine), and AXIDTM (nizatidine)), inhibitors Of H + , K + ATPase (e g , PREVACIDTM (lansoprazole) and PRILOSECTM (omeprazole)), Bismuth compounds (e g , PEPTO-BISMOLTM (bismuth subsalicylate) and DE-NOLTM (bismuth subcitrate)), various antacids, sucralfate, prostaglandin analogs (e g CYTOTECTM (misoprosto)
  • albumin fusion proteins and/or polynucleotides of the invention are administered in combination with other therapeutic or prophylactic regimens, such as, for example, radiation therapy
  • the invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions comprising albumin fusion proteins of the invention
  • a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions comprising albumin fusion proteins of the invention
  • Constructs encoding albumin fusion proteins of the invention can be used as a part of a gene therapy protocol to deliver therapeutically effective doses of the albumin fusion protein
  • a preferred approach for in vivo introduction of nucleic acid into a cell is by use of a viral vector containing nucleic acid, encoding an albumin fusion protein of the invention
  • Infection of cells with a viral vector has the advantage that a large proportion of the targeted cells can receive the nucleic acid
  • molecules encoded within the viral vector, e g by a cDNA contained in the viral vector, are expressed efficiently in cells which have taken up viral vector nucleic acid
  • Retrovirus vectors and adeno-associated virus vectors can be used as a recombinant gene delivery system for the transfer of exogenous nucleic acid molecules encoding albumin fusion proteins in vivo These vectors provide efficient delivery of nucleic acids into cells, and the transferred nucleic acids are stably integrated into the chromosomal DNA of the host.
  • packaging cells which produce only replication-defective retroviruses has increased the utility of retroviruses for gene therapy, and defective retroviruses are characte ⁇ zed for use in gene transfer for gene therapy purposes (for a review see Miller, A D (1990) Blood 7627 1)
  • a replication defective retrovirus can be packaged into virions which can be used to infect a target cell through the use of a helper virus by standard techniques Protocols for producing recombinant retroviruses and for infecting cells in vitro or in vivo with such viruses can be found in Current Protocols in Molecular Biology, Ausubel,
  • introduced adenoviral DNA (and foreign DNA contained therein) is not integrated into the genome of a host cell but remains episomal, thereby avoiding potential problems that can occur as a result of insertional mutagenesis in situations where introduced DNA becomes integrated into the host genome ⁇ e.g., retroviral DNA).
  • the carrying capacity of the adenoviral genome for foreign DNA is large (up to 8 kilobases) relative to other gene delivery vectors (Berkner et al, cited supra; Haj-Ahmand et al., J. Virol. 5 7:267 (1986)).
  • non-viral gene delivery systems of the present invention rely on endocytic pathways for the uptake of the subject nucleotide molecule by the targeted cell.
  • exemplary gene delivery systems of this type include liposomal derived systems, poly-lysine conjugates, and artificial viral envelopes.
  • a nucleic acid molecule encoding an albumin fusion protein of the invention can be entrapped in liposomes bearing positive charges on their surface (e.g., lipofectins) and (optionally) which are tagged with antibodies against cell surface antigens of the target tissue (Mizuno et al (1992) No Shinkei Geka 20:547-55 1; PCT publication W091/06309; Japanese patent application 1047381; and European patent publication EP-A-43075).
  • Gene delivery systems for a gene encoding an albumin fusion protein of the invention can be introduced into a patient by any of a number of methods.
  • a pharmaceutical preparation of the gene delivery system can be introduced systemically, e.g. by intravenous injection, and specific transduction of the protein in the target cells occurs predominantly from specificity of transfection provided by the gene delivery vehicle, cell-type or tissue-type expression due to the transcriptional regulatory sequences controlling expression of the receptor gene, or a combination thereof.
  • initial delivery of the recombinant gene is more limited with introduction into the animal being quite localized.
  • the gene delivery vehicle can be introduced by catheter (see U.S. Patent 5,328,470) or by Stereotactic injection (e.g.
  • the pharmaceutical preparation of the gene therapy construct can consist essentially of the gene delivery system in an acceptable diluent, or can comprise a slow release matrix in which the gene delivery vehicle is imbedded.
  • the albumin fusion protein can be produced intact from recombinant cells, e.g. retroviral vectors, the pharmaceutical preparation can comprise one or more cells which produce the albumin fusion protein.
  • the gene therapy methods relate to the introduction of nucleic acid (DNA, RNA and antisense DNA or RNA) sequences into an animal to achieve expression of an albumin fusion protein of the invention.
  • This method requires a polynucleotide which codes for an albumin fusion protein of the present invention operatively linked to a promoter and any other genetic elements necessary for the expression of the fusion protein by the target tissue.
  • Such gene therapy and delivery techniques are known in the art, see, for example, WO90/11092, which is herein incorporated by reference.
  • cells from a patient may be engineered with a polynucleotide (DNA or RNA) comprising a promoter operably linked to.a polynucleotide-encoding an albumin fusion protein of the present invention ex-vivo, with fhe engineered cells then being provided to a patient to be treated with the fusion protein of the present invention.
  • a polynucleotide DNA or RNA
  • Such methods are well-known in the art. For example, see Belldegrun, A., et al., J. Natl. Cancer Inst. 85: 207-216 (1993); Ferrantini, M. et al., Cancer Research 53: 1107-1112 (1993); Ferrantini, M. et al., J.
  • the cells which are engineered are arterial cells.
  • the arterial cells may be reintroduced into the patient through direct injection to the artery, the tissues surrounding the artery, or through catheter injection.
  • the polynucleotide constructs can be delivered by any method that delivers injectable materials to the cells of an animal, such as, injection into the interstitial space of tissues (heart, muscle, skin, lung, liver, and the like).
  • the polynucleotide constructs may be delivered in a pharmaceutically acceptable liquid or aqueous carrier.
  • polynucleotides encoding the albumin fusion proteins of the present invention is delivered as a naked polynucleotide.
  • naked polynucleotide, DNA or RNA refers to sequences that are free from any delivery vehicle that acts to assist, promote or facilitate entry into the cell, including viral sequences, viral particles, liposome formulations, lipofectin or precipitating agents and the like.
  • polynucleotides encoding the albumin fusion proteins of the present invention can also be delivered in liposome formulations and lipofectin formulations and the like can be prepared by methods well known to those skilled in the art. Such methods are described, for example, in U.S. Patent Nos. 5,593,972, 5,589,466, and 5,580,859, which are herein incorporated by reference.
  • the polynucleotide vector constructs used in the gene therapy method are preferably constructs that will not integrate into the host genome nor will they contain sequences that allow for replication.
  • Appropriate vectors include pWLNEO, pSV2CAT, pOG44, pXTl and pSG available from Stratagene; pSVK3, pBPV, pMSG and pSVL available from Pharmacia; and pEFl/V5, pcDNA3.1, and pRc/CMV2 available from Invitrogen.
  • Other suitable vectors will be readily apparent to the skilled artisan.
  • Suitable promoters include adenoviral promoters, such as the adenoviral major late promoter; or heterologous promoters, such as the cytomegalovirus (CMV) promoter; the respiratory syncytial virus (RSV) promoter; inducible promoters, such as the MMT promoter, the metallothionein promoter; heat shook promoters; the albumin promoter; the ApoAI promoter; human globin promoters; viral thymidine kinase promoters, such as the Herpes
  • CMV cytomegalovirus
  • RSV respiratory syncytial virus
  • inducible promoters such as the MMT promoter, the metallothionein promoter
  • heat shook promoters such as the albumin promoter
  • the ApoAI promoter human globin promoters
  • viral thymidine kinase promoters such as the Herpes
  • Simplex thymidine kinase promoter may be the native promoter for the gene corresponding to the Therapeutic protein portion of the albumin fusion proteins of the invention.
  • nucleic acid sequences Unlike other gene therapy techniques, one major advantage of introducing naked nucleic acid sequences into target cells is the transitory nature of the polynucleotide synthesis in the cells. Studies have shown that non-replicating DNA sequences can be introduced into cells to provide production of the desired polypeptide for periods of up to six months.
  • the polynucleotide construct can be delivered to the interstitial space of tissues within the an animal, including of muscle, skin, brain, lung, liver, spleen, bone marrow, thymus, heart, lymph, blood, bone, cartilage, pancreas, kidney, gall bladder, stomach, intestine, testis, ovary, uterus, rectum, nervous system, eye, gland, and connective tissue.
  • Interstitial space of the tissues comprises the intercellular, fluid, mucopolysaccharide matrix among the reticular fibers of organ tissues, elastic fibers in the walls of vessels or chambers, collagen fibers of fibrous tissues, or that same matrix within connective tissue ensheathing muscle cells or in the lacunae of bone.
  • the space occupied by the plasma of the circulation and the lymph fluid of the lymphatic channels Delivery to the interstitial space of muscle tissue is preferred for the reasons discussed below. They may be conveniently delivered by injection into the tissues comprising these cells. They are preferably delivered to and expressed in persistent, non-dividing cells which are differentiated, although delivery and expression may be achieved in non-differentiated or less completely differentiated cells, such as, for example, stem cells of blood or skin fibroblasts. In vivo muscle cells are particularly competent in their ability to take up and express polynucleotides.
  • an effective dosage amount of DNA or RNA will be in the range of from about 0.05 mg/kg body weight to about 50 mg/kg body weight. Preferably the dosage will be from about 0.005 mg/kg to about 20 mg/kg and more preferably from about 0.05 mg/kg to about 5 mg/kg. Of course, as the artisan of ordinary skill will appreciate, this dosage will vary according to the tissue site of injection. The appropriate and effective dosage of nucleic acid sequence can readily be determined by those of ordinary skill in the art and may depend on the condition being treated and the route of administration.
  • the preferred route of administration is by the parenteral route of injection into the interstitial space of tissues.
  • parenteral routes may also be used, such as, inhalation of an aerosol formulation particularly for delivery to lungs or bronchial tissues, throat or mucous membranes of the nose.
  • naked DNA constructs can be delivered to arteries during angioplasty by the catheter used in the procedure.
  • the naked polynucleotides are delivered by any method known in the art, including, but not limited to, direct needle injection at the delivery site, intravenous injection, topical administration, catheter infusion, and so-called "gene guns”. These delivery methods are known in the art.
  • The-constructs may also be delivered with delivery vehicles such as viral sequences; viral particles, liposome formulations, lipofectin, precipitating agents, etc. Such methods of delivery are known in the art.
  • the polynucleotide constructs are complexed in a liposome preparation.
  • Liposomal preparations for use in the instant invention include cationic (positively charged), anionic (negatively charged) and neutral preparations.
  • cationic liposomes are particularly preferred because a tight charge complex can be formed between the cationic liposome and the polyanionic nucleic acid.
  • Cationic liposomes have been shown to mediate intracellular delivery of plasmid DNA (Feigner et al., Proc. Natl. Acad. Sci. USA (1987) 84:7413-7416, which is herein incorporated by reference); mRNA (Malone et al., Proc. Natl.
  • Cationic liposomes are readily available.
  • N[l-2,3-dioleyloxy)propyl]-N,N,N-triethylammonium (DOTMA) liposomes are particularly useful and are available under the trademark Lipofectin, from GIBCO BRL, Grand Island, N. Y. (See, also, Feigner et al., Proc. Natl
  • cationic liposomes can be prepared from readily available materials using techniques well known in the art. See, e.g. PCT
  • DOTMA liposomes Preparation of DOTMA liposomes is explained in the literature, see, e.g., P. Feigner et al., Proc. Natl.
  • anionic and neutral liposomes are readily available, such as from Avanti Polar Lipids (Birmingham, Ala.), or can be easily prepared using readily available materials.
  • Such materials include phosphatidyl, choline, cholesterol, phosphatidyl ethanolamine, dioleoylphosphatidyl choline (DOPC), dioleoylphosphatidyl glycerol (DOPG), dioleoylphoshatidyl ethanolamine (DOPE), among others.
  • DOPC dioleoylphosphatidyl choline
  • DOPG dioleoylphosphatidyl glycerol
  • DOPE dioleoylphoshatidyl ethanolamine
  • DOPC dioleoylphosphatidyl choline
  • DOPG dioleoylphosphatidyl glycerol
  • DOPE dioleoylphosphatidyl ethanolamine
  • DOPG/DOPC vesicles can be prepared by drying 5 0 mg each of DOPG and DOPC under a stream of nitrogen gas into a sonication vial. The sample is placed under a vacuum pump overnight and is hydrated the following day with deionized water.
  • the sample is then sonicated for 2 hours in a capped vial, using a Heat Systems model 350 sonicator equipped with an inverted cup (bath type) probe at the maximum setting while the bath is circulated at I 5 degrees celcius.
  • negatively charged vesicles can be prepared without sonication to produce multilamellar vesicles or by extrusion through nucleopore membranes to produce unilamellar vesicles of discrete size.
  • Other methods are known and available to those of skill in the art.
  • the liposomes can comprise multilamellar vesicles (MLVs), small unilamellar vesicles (SUVs), or large unilamellar vesicles (LUVs), with SUVs being preferred.
  • MLVs multilamellar vesicles
  • SUVs large unilamellar vesicles
  • the various liposome-nucleic acid complexes are prepared using methods well known in the art. See, e.g., Straubinger et al., Methods of Immunology (1983), 101:512-527, which is herein incorporated by reference.
  • MLVs containing nucleic acid can be prepared by depositing a thin film of phospholipid on the walls of a glass tube and subsequently hydrating with a solution of the material to be encapsulated.
  • SUVs are prepared by extended sonication of MLVs to produce a homogeneous population of unilamellar liposomes.
  • the material to be entrapped is added to a suspension of preformed MLVs and then sonicated.
  • liposomes containing cationic lipids the dried lipid film is resuspended in an appropriate solution such as sterile water or an isotonic buffer solution such as 10 mM Tris/NaCl, sonicated, and then the preformed liposomes are mixed directly with the DNA.
  • the liposome and DNA form a very stable complex due to binding of the positively charged liposomes to the cationic DNA.
  • SUVs find use with small nucleic acid fragments.
  • LUVs are prepared by a number of methods, well known in the art. Commonly used methods include Ca 2+ -EDTA chelation (Papahadjopoulos et al., Biochim. Biophys. Acta (1975) 394:483; Wilson et al., Cell 17:77 (1979)); ether injection (Deamer, D. and Bangham, A., Biochim. Biophys. Acta 443:629 (1976); Ostro et al., Biochem. Biophys. Res. Commun. 76:836 (1977); Fraley et al., Proc. Natl. Acad. Sci. USA 76:3348 (1979)); detergent dialysis (Enoch, H.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Medicinal Chemistry (AREA)
  • Virology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Oncology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Communicable Diseases (AREA)
  • Pulmonology (AREA)
  • Zoology (AREA)
  • Epidemiology (AREA)
  • Toxicology (AREA)
  • Peptides Or Proteins (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Medicinal Preparation (AREA)

Abstract

L'invention concerne des protéines de fusion avec l'albumine, des molécules d'acide nucléique codant ces protéines de fusion avec l'albumine, des vecteurs contenant ces acides nucléiques, des cellules hôtes transformées par ces vecteurs d'acide nucléique et des procédés pour produire lesdites protéines de fusion avec l'albumine et pour utiliser ces acides nucléiques, ces vecteurs et/ou ces cellules hôtes. La présente invention porte également sur des compositions pharmaceutiques contenant les protéines de fusion avec l'albumine de l'invention et sur des méthodes de traitement, de prévention ou d'amélioration de maladies, de troubles ou d'états au moyen desdites protéines de fusion avec l'albumine.
EP06813242A 2005-08-12 2006-07-31 Protéines de fusion avec l'albumine Withdrawn EP1924596A4 (fr)

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US70752105P 2005-08-12 2005-08-12
US71238605P 2005-08-31 2005-08-31
US73272405P 2005-11-03 2005-11-03
US77691406P 2006-02-28 2006-02-28
US78136106P 2006-03-13 2006-03-13
US81018206P 2006-06-02 2006-06-02
US81368206P 2006-06-15 2006-06-15
PCT/US2006/029391 WO2007021494A2 (fr) 2005-08-12 2006-07-31 Proteines de fusion avec l'albumine

Publications (2)

Publication Number Publication Date
EP1924596A2 true EP1924596A2 (fr) 2008-05-28
EP1924596A4 EP1924596A4 (fr) 2009-07-29

Family

ID=37758042

Family Applications (1)

Application Number Title Priority Date Filing Date
EP06813242A Withdrawn EP1924596A4 (fr) 2005-08-12 2006-07-31 Protéines de fusion avec l'albumine

Country Status (13)

Country Link
EP (1) EP1924596A4 (fr)
JP (1) JP2009504157A (fr)
KR (1) KR20080071119A (fr)
AU (1) AU2006280312A1 (fr)
BR (1) BRPI0614761A2 (fr)
CA (1) CA2618476A1 (fr)
EC (1) ECSP088262A (fr)
IL (1) IL189246A0 (fr)
MA (1) MA29836B1 (fr)
MX (1) MX2008001865A (fr)
NO (1) NO20081233L (fr)
TN (1) TNSN08064A1 (fr)
WO (1) WO2007021494A2 (fr)

Families Citing this family (67)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB9526733D0 (en) 1995-12-30 1996-02-28 Delta Biotechnology Ltd Fusion proteins
US20040010134A1 (en) 2000-04-12 2004-01-15 Rosen Craig A. Albumin fusion proteins
DK1463751T3 (da) 2001-12-21 2013-08-26 Human Genome Sciences Inc Albuminfusionsproteiner.
US20080194481A1 (en) 2001-12-21 2008-08-14 Human Genome Sciences, Inc. Albumin Fusion Proteins
EP1729795B1 (fr) 2004-02-09 2016-02-03 Human Genome Sciences, Inc. Proteines hybrides d'albumine
EP1816201A1 (fr) 2006-02-06 2007-08-08 CSL Behring GmbH Facteur de coagulation VIIa modifié ayant une stabilité 'half-life' améliorée
CA2654055A1 (fr) 2006-06-07 2007-12-21 Human Genome Sciences, Inc. Proteines de fusion d'albumine
US20100254944A1 (en) * 2006-09-14 2010-10-07 Mani Subramanian Albumin Fusion Proteins
EP2423221B1 (fr) 2007-01-30 2015-04-29 Epivax, Inc. Épitopes de lymphocytes t régulateurs, compositions et utilisations de ceux-ci
RU2567667C2 (ru) 2008-06-13 2015-11-10 Проекто Де Биомедисина Сима, С.Л. Конъюгаты для введения биологически активных соединений
EA023344B1 (ru) 2009-01-16 2016-05-31 Тева Фармасьютикал Индастриз Лтд. Способ лечения или предотвращения нейтропении или лейкопении или снижения частоты возникновения инфекции, проявляющейся фебрильной нейтропенией
CN106986933A (zh) 2009-02-11 2017-07-28 阿尔布梅迪克斯医疗公司 白蛋白变体和缀合物
KR20100100254A (ko) * 2009-03-05 2010-09-15 (주)바이오큐어팜 인간 혈청 알부민-팀프-2 융합 단백질과 항암제를 포함하는암의 예방 또는 치료용 조성물
BR112012004094A2 (pt) 2009-08-24 2016-03-08 Amunix Operating Inc composições de fator vii de coagulação e métodos para fazer e usar as mesmas
RU2607374C2 (ru) * 2009-10-30 2017-01-10 Новозаймс Байофарма Дк А/С Варианты альбумина
KR20130070576A (ko) 2010-04-09 2013-06-27 노보자임스 바이오파마 디케이 에이/에스 알부민 유도체 및 변이체
BR112012029611A2 (pt) 2010-05-21 2017-07-25 Merrimack Pharmaceuticals Inc proteína de fusão biespecífica, composição farmacêutica, método de tratamento de dano ao tecido em um indivíduo, método de promoção da regeração ou sobrevivência do tecido em um indivíduo e molécula de ácido nucleico
KR101969526B1 (ko) * 2011-02-28 2019-04-17 고쿠리츠켄큐카이하츠호진 고쿠리츠쥰칸키뵤 겐큐센터 악성 종양 전이 억제용 의약
CN104011072B (zh) 2011-05-05 2018-10-12 阿尔布梅迪克斯医疗有限公司 白蛋白变体
US10201584B1 (en) 2011-05-17 2019-02-12 Abbvie Inc. Compositions and methods for treating HCV
WO2012170969A2 (fr) 2011-06-10 2012-12-13 Biogen Idec Ma Inc. Composés pro-coagulants et leurs procédés d'utilisation
ES2527510T1 (es) 2011-10-21 2015-01-26 Abbvie Inc. Métodos para el tratamiento del VHC que comprenden al menos dos agentes antivirales de acción directa, ribavirina pero no interferón
DE112012003457T5 (de) 2011-10-21 2015-03-12 Abbvie Inc. Kombinationsbehandlung (z.B. mit ABT-072 oder ABT-333 von DAAs zur Verwendung in der Behandlung von HCV)
US8466159B2 (en) 2011-10-21 2013-06-18 Abbvie Inc. Methods for treating HCV
US8492386B2 (en) 2011-10-21 2013-07-23 Abbvie Inc. Methods for treating HCV
US20140315817A1 (en) 2011-11-18 2014-10-23 Eleven Biotherapeutics, Inc. Variant serum albumin with improved half-life and other properties
MY201293A (en) 2012-01-12 2024-02-15 Bioverativ Therapeutics Inc Chimeric factor viii polypeptides and uses thereof
RS63870B1 (sr) 2012-02-15 2023-01-31 Bioverativ Therapeutics Inc Sastavi faktora viii i postupci za pravljenje i upotrebu istih
JP6383666B2 (ja) 2012-02-15 2018-08-29 バイオベラティブ セラピューティクス インコーポレイテッド 組換え第viii因子タンパク質
CA2861592A1 (fr) 2012-03-16 2013-09-19 Novozymes Biopharma Dk A/S Variants d'albumine
EP2863940A4 (fr) 2012-06-08 2016-08-10 Biogen Ma Inc Facteurs de coagulation chimériques
EP2858659B1 (fr) 2012-06-08 2019-12-25 Bioverativ Therapeutics Inc. Composés pro-coagulants
CN103525695B (zh) * 2012-07-04 2015-05-20 长沙中生众捷生物技术有限公司 便携式肾功能检测系统
DK2882450T3 (da) 2012-07-11 2020-02-24 Bioverativ Therapeutics Inc Faktor viii-kompleks med xten og von willebrand-faktorprotein samt anvendelser deraf
WO2014063108A1 (fr) 2012-10-18 2014-04-24 Biogen Idec Ma Inc. Procédés d'utilisation d'une dose fixe d'un facteur de coagulation
WO2014072481A1 (fr) 2012-11-08 2014-05-15 Novozymes Biopharma Dk A/S Variants d'albumine
HRP20231183T1 (hr) 2013-02-15 2024-01-05 Bioverativ Therapeutics Inc. Optimizirani gen faktora viii
ES2648591T3 (es) 2013-02-16 2018-01-04 Albumedix A/S Modelo animal farmacocinético
SG10201707600XA (en) 2013-03-15 2017-11-29 Biogen Ma Inc Factor ix polypeptide formulations
CA2913078A1 (fr) 2013-06-28 2014-12-31 Biogen Ma Inc. Lieur pouvant etre fendu par thrombine ayant un xten et ses utilisations
US10548953B2 (en) 2013-08-14 2020-02-04 Bioverativ Therapeutics Inc. Factor VIII-XTEN fusions and uses thereof
WO2015048330A2 (fr) 2013-09-25 2015-04-02 Biogen Idec Ma Inc. Procédés d'inactivation de virus sur colonne
WO2015070014A1 (fr) 2013-11-08 2015-05-14 Biogen Idec Ma Inc. Composé de fusion procoagulant
AU2015204646B2 (en) 2014-01-10 2020-08-27 Bioverativ Therapeutics Inc. Factor VIII chimeric proteins and uses thereof
EP3160478A4 (fr) 2014-06-30 2018-05-16 Bioverativ Therapeutics Inc. Gène du facteur ix optimisé
CA3203273A1 (fr) 2014-10-14 2016-04-21 Halozyme, Inc. Compositions d'adenosine deaminase-2, variants et methodes d'utilisation
MA40835A (fr) 2014-10-23 2017-08-29 Biogen Ma Inc Anticorps anti-gpiib/iiia et leurs utilisations
MA40861A (fr) 2014-10-31 2017-09-05 Biogen Ma Inc Anticorps anti-glycoprotéines iib/iiia
MX2018001497A (es) 2015-08-03 2018-05-15 Bioverativ Therapeutics Inc Proteinas de fusion de factor ix y metodos para producirlas y usarlas.
EP3337816B1 (fr) 2015-08-20 2024-02-14 Albumedix Ltd Variants de l'albumine et leurs conjugués
CA3000742A1 (fr) 2015-10-02 2017-04-06 Silver Creek Pharmaceuticals, Inc. Proteines therapeutiques bispecifiques pour la reparation de tissus
CN105254766B (zh) * 2015-10-26 2018-10-16 中国航天员科研训练中心 机械生长因子MGF E domain肽在调控记忆相关基因和miRNA表达中的应用
SI3411478T1 (sl) 2016-02-01 2022-10-28 Bioverativ Therapeutics Inc. Optimirani geni dejavnika VIII
WO2017189978A1 (fr) 2016-04-28 2017-11-02 Emory University Compositions thérapeutiques à base de nucléotides et nucléosides contenant un alcyne et utilisations associées
EP3535585A1 (fr) 2016-11-04 2019-09-11 Aarhus Universitet Identification et traitement de tumeurs caractérisées par une surexpression du récepteur fc néonatal
MX2019006444A (es) 2016-12-02 2019-10-30 Bioverativ Therapeutics Inc Métodos de tratamiento de artropatía hemofílica utilizando factores de coagulación quiméricos.
BR112019011198A2 (pt) 2016-12-02 2019-12-17 Bioverativ Therapeutics Inc métodos de indução de tolerância imune a fatores de coagulação
WO2018112362A1 (fr) 2016-12-16 2018-06-21 Biogen Ma Inc. Facteur 11 de différenciation de croissance activé protéolytiquement stabilisé
TW201831521A (zh) 2017-01-31 2018-09-01 美商生物化學醫療公司 因子ix融合蛋白以及其製備方法及使用方法
WO2018193033A1 (fr) * 2017-04-20 2018-10-25 Novo Nordisk A/S Procédés de purification de protéines de fusion d'albumine
US20210163986A1 (en) 2017-08-09 2021-06-03 Bioverativ Therapeutics Inc. Nucleic acid molecules and uses thereof
SG11202007114VA (en) 2018-02-01 2020-08-28 Bioverativ Therapeutics Inc Use of lentiviral vectors expressing factor viii
CA3108799A1 (fr) 2018-08-09 2020-02-13 Bioverativ Therapeutics Inc. Molecules d'acide nucleique et leurs utilisations pour une therapie genique non virale
JP2022514465A (ja) 2018-12-06 2022-02-14 バイオベラティブ セラピューティクス インコーポレイテッド 第ix因子を発現するレンチウイルスベクターの使用
WO2021067389A1 (fr) 2019-09-30 2021-04-08 Bioverativ Therapeutics Inc. Formulations de vecteur lentiviral
JPWO2022030580A1 (fr) * 2020-08-06 2022-02-10
JP2024056667A (ja) * 2022-10-11 2024-04-23 Jcrファーマ株式会社 血清アルブミンと生理活性を有する蛋白質との融合蛋白質

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003060071A2 (fr) * 2001-12-21 2003-07-24 Human Genome Sciences, Inc. Proteines hybrides d'albumine
WO2005003296A2 (fr) * 2003-01-22 2005-01-13 Human Genome Sciences, Inc. Proteines hybrides d'albumine
WO2005077042A2 (fr) * 2004-02-09 2005-08-25 Human Genome Sciences, Inc. Proteines hybrides d'albumine

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060051859A1 (en) * 2004-09-09 2006-03-09 Yan Fu Long acting human interferon analogs

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003060071A2 (fr) * 2001-12-21 2003-07-24 Human Genome Sciences, Inc. Proteines hybrides d'albumine
WO2005003296A2 (fr) * 2003-01-22 2005-01-13 Human Genome Sciences, Inc. Proteines hybrides d'albumine
WO2005077042A2 (fr) * 2004-02-09 2005-08-25 Human Genome Sciences, Inc. Proteines hybrides d'albumine

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of WO2007021494A2 *

Also Published As

Publication number Publication date
EP1924596A4 (fr) 2009-07-29
MA29836B1 (fr) 2008-10-03
KR20080071119A (ko) 2008-08-01
BRPI0614761A2 (pt) 2009-05-19
CA2618476A1 (fr) 2007-02-22
WO2007021494A3 (fr) 2007-07-26
WO2007021494A2 (fr) 2007-02-22
ECSP088262A (es) 2008-05-30
IL189246A0 (en) 2008-08-07
NO20081233L (no) 2008-05-09
AU2006280312A1 (en) 2007-02-22
JP2009504157A (ja) 2009-02-05
MX2008001865A (es) 2008-04-15
TNSN08064A1 (en) 2009-07-14

Similar Documents

Publication Publication Date Title
US8287859B2 (en) Methods of reducing toxicity and effects of cocaine by administering a butyrylcholinesterase (BChE)-albumin fusion protein
US8143026B2 (en) Albumin fusion proteins
US8969538B2 (en) Albumin fusion proteins
US7521424B2 (en) Albumin fusion proteins
US7238660B2 (en) Albumin fusion proteins
US20100254944A1 (en) Albumin Fusion Proteins
US20080004206A1 (en) Albumin fusion proteins
EP2277889A2 (fr) Protéines chimériques d'albumine et interféron beta
WO2007021494A2 (fr) Proteines de fusion avec l'albumine
ZA200605871B (en) Albumin fusion proteins

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20080312

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC NL PL PT RO SE SI SK TR

REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 1113581

Country of ref document: HK

A4 Supplementary search report drawn up and despatched

Effective date: 20090626

17Q First examination report despatched

Effective date: 20100108

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20120201

DAX Request for extension of the european patent (deleted)
REG Reference to a national code

Ref country code: HK

Ref legal event code: WD

Ref document number: 1113581

Country of ref document: HK