CA2513213C - Albumin fusion proteins - Google Patents

Albumin fusion proteins Download PDF

Info

Publication number
CA2513213C
CA2513213C CA2513213A CA2513213A CA2513213C CA 2513213 C CA2513213 C CA 2513213C CA 2513213 A CA2513213 A CA 2513213A CA 2513213 A CA2513213 A CA 2513213A CA 2513213 C CA2513213 C CA 2513213C
Authority
CA
Canada
Prior art keywords
hsa
protein
albumin fusion
therapeutic
seq
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.)
Expired - Fee Related
Application number
CA2513213A
Other languages
French (fr)
Other versions
CA2513213A1 (en
Inventor
William A. Haseltine
Craig A. Rosen
Steven M. Ruben
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 CA2513213A1 publication Critical patent/CA2513213A1/en
Application granted granted Critical
Publication of CA2513213C publication Critical patent/CA2513213C/en
Anticipated expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/38Albumins
    • 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]
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/31Fusion polypeptide fusions, other than Fc, for prolonged plasma life, e.g. albumin

Abstract

The present invention encompasses albumin fusion proteins. Nucleic acid molecules encoding the albumin fusion proteins of the invention are also encompassed by the invention, as are vectors containing these nucleic acids, host cells transformed with these nucleic acids vectors, and methods of making the albumin fusion proteins of the invention and using these nucleic acids, vectors, and/or host cells. Additionally the present invention encompasses pharmaceutical compositions comprising albumin fusion proteins and methods of treating, preventing, or ameliorating diseases, disordrs or conditions using albumin fusion proteins of the invention.

Description

DEMANDES OU BREVETS VOLUMINEUX
LA PRESENTE PARTIE DE CETTE DEMANDE OU CE BREVETS
COMPREND PL US D'UN TOME.

NOTE: Pour les tomes additionels, veillez contacter le Bureau Canadien des Brevets.
JUMBO APPLICATIONS / PATENTS
THIS SECTION OF THE APPLICATION / PATENT CONTAINS MORE
THAN ONE VOLUME.

NOTE: For additional volumes please contact the Canadian Patent Office.

Albumin Fusion Proteins BACKGROUND OF THE INVENTION
[0001] 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.
[0002]
Human serum albumin (HSA, or HA), a protein of 585 amino acids in its mature form (as shown in Figure 1 (SEQ ID NO:1)), is responsible for a significant proportion of the osmotic pressure of serum and also functions as a carrier of endogenous and exogenous ligands. At present, HA for clinical use is produced by extraction from human blood. The production of recombinant HA (rHA) in microorganisms has been disclosed in EP 330 451 and EP 361 991.
[0003]
Therapeutic proteins in their native state or when recombinantly produced, such as interferons and growth hormones, are typically labile molecules exhibiting short shelf-lives, particularly when formulated in aqueous solutions. The instability in these molecules when formulated for administration dictates that many of the molecules must be lyophilized and refrigerated at all times during storage, thereby rendering the molecules difficult to transport and/or store.
Storage problems are particularly acute when pharmaceutical formulations must be stored and dispensed outside of the hospital environment.
[0004] Few practical solutions to the storage problems of labile protein molecules have been proposed. Accordingly, there is a need for stabilized, long lasting formulations of proteinaceous therapeutic molecules that are easily dispensed, preferably with a simple formulation requiring minimal post-storage manipulation.

SUMMARY OF THE INVENTION
An object of the present invention is to provide albumin fusion proteins. In accordance with an aspect of the present invention, there is provided an albumin fusion protein comprising a member selected from the group consisting of:
(a) a Therapeutic protein:X and albumin comprising the amino acid sequence of SEQ ID NO:1;
(b) a Therapeutic protein:X and a fragment or a variant of the amino acid sequence of SEQ ID NO:1, wherein said fragment or variant has albumin activity;
(c) a Therapeutic protein:X and a fragment or a variant of the amino acid sequence of SEQ L1D NO:1, wherein said fragment or variant has albumin activity, and further wherein said albumin activity is the ability to prolong the shelf life of the Therapeutic protein:X compared to the shelf-life of the Therapeutic protein:X in an tmfused state;
(d) a Therapeutic protein:X and a fragment or a variant of the amino acid sequence of SEQ ID NO:1, wherein said fragment or variant has albumin activity, and further wherein the fragment or variant comprises the amino acid sequence of amino acids 1-387 of SEQ LD NO:1;
(e) a fragment or variant of a Therapeutic protein:X and albumin comprising the amino acid sequence of SEQ ID NO:1, wherein said fragment or variant has a biological activity of the Therapeutic protein:X;
(f) a Therapeutic protein:X, or fragment or variant thereof, and albumin, or fragment or variant thereof, of (a) to (e), wherein the Therapeutic protein:3C, or fragment or variant thereof, is fused to the N-terminus of albumin, or the N-terminus of the fragment or variant of albumin;
(g) a Therapeutic protein:X, or fragment or variant thereof, and albumin, or fragment or variant thereof, of (a) to (e), wherein the Therapeutic protein:X, or fragment or variant thereof, is fused to the C-terminus of albumin, or the C-terminus of the fragment or variant of albumin;
(h) a Therapeutic protein:X, or fragment or variant thereof and albumin, or fragment or variant thereof, of (a) to (e), wherein the Therapeutic protein:X, or fragment or variant thereof, is fused to the N- terminus and C-terminus of albumin, or the N-terminus and the C-terminus of the fragment or variant of albumin;
(i) a Therapeutic protein:X, or fragment or variant thereof, and albumin, or fragment or variant thereof, of (a) to (e), which comprises a first Therapeutic protein:X, or fragment or variant thereof, and a second Therapeutic protein:X, or fragment or variant thereof, wherein said first Therapeutic protein:X, or fragment or variant thereof, is different from said second Therapeutic protein:X, or fragm' ent or variant thereof;
(j) a Therapeutic protein:X, or fragment or variant thereof, and albumin, or fragment or variant thereof; of (a) to (i), wherein the Therapeutic protein:X, or fragment or variant thereof, is separated from the albumin or the fragment or variant of albumin by a linker, and (k) a Therapeutic protein:X, or fragment or variant thereof, and albumin, or fragment or variant thereof, of (a) to (j), wherein the albumin fusion protein has the following formula:
R1-L-R2; R2-L-R1; or R1-L-R2-L-R1, and further wherein RI is Therapeutic protein:X, or fragment or variant thereof; L is a peptide linker, and R2 is albumin comprising the amino acid sequence of SEQ ID
NO:1 or a fragment or variant of albumin.
In accordance with another aspect of the invention, there is provided an albumin fusion protein comprising a Therapeutic piotein:X, or fragment or variant thereof;
inserted into an albumin, or fragment or variant thereof, comprising the amino acid sequence of SEQ ID NO:1 or fragment or variant thereof.
In accordance with another aspect of the invention, there is provided an albumin fusion protein comprising a Therapeutic protein:X, or fragment or variant thereof;
inserted into an albumin, or fragment or variant thereof, comprising an amino acid sequence selected from the group consisting of:
(a) amino acids 54 to 61 of SEQ 1D NO:1;
(b) amino acids 76 to 89 of SEQ 1D NO:1;
(c) amino acids 92 to100 of SEQ ID NO:1;
(d) amino acids 170 to 176 of SEQ ID NO:1;
(e) amino acids 247 to 252 of SEQ ID NO:1;
(1) amino acids 266 to 277 of SEQ ID NO:1;
(g) amino acids 280 to 288 of SEQ ID NO:1;
(h) amino acids 362 to 368 of SEQ ID NO:1;
(i) amino acids 439 to 447 of SEQ ID NO:1;
2a (i) amino acids 462 to 475 of SEQ ID NO: I;
(k) amino acids 478 to 486 of SEQ ID NO:1; and (1) amino acids 560 to 566 of SEQ NO:l.
In accordance with another aspect of the invention, there is provided a method of extending the shelf life of Therapeutic protein:X, or fragment or variant thereof, comprising the step of fusing the Therapeutic protein:X, or fragment or variant thereof, to albumin, or fragment or variant thereof, sufficient to extend the shelf-life of the Therapeutic protein:X, or fragment or variant thereof, compared to the shelf-life of the Therapeutic protein:X, or fragment or variant thereof, in an unfused state.
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. 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, and/or stabilize the Therapeutic protein and/or its activity in solution (or in a pharmaceutical composition) in vitro and/or in vivo. 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.
In a preferred aspect of the invention, 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 an'd a pharmaceutically acceptable diluent or carrier.
Such formulations may be in a kit or container. Such 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 2b 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 In other embodiments, the present invention encompasses methods of preventing, treating, or ameliorating a disease or disorder. In preferred embodiments, 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 =
2c 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.
10009] In one embodiment, an albumin fusion protein described in Table 1 or 2 has extended shelf life.
[0010] In a second embodiment, an albumin fusion protein described in Table 1 or 2 is more stable than the corresponding unfused Therapeutic molecule described in Table 1.
[0011] 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.
BRIEF DESCRIPTION OF THE FIGURES
[0012]
Figure 1A-D shows the amino acid sequence of the mature form of human albumin (SEQ ID NO:1) and a polynucleotide encoding it (SEQ ID NO:2).
[0013]
Figure 2 shows the restriction map of the pPPC0005 cloning vector ATCC
deposit PTA-3278.
[0014]
Figure 3 shows the restriction map of the pSAC35 yeast S. cerevisiae expression vector (Sleep etal., BioTechnology 8:42 (1990)).
[0015]
Figure 4 shows the effect, of various dilutions of BNP albumin fusion proteins encoded by DNA comprised in Construct ID Nos. (hereinafter CID) 3448 (BNP/HSA) and 3477 (BNP2X/HSA) versus BNP alone on cGMP induction in RFL-6 lung fibroblasts.
Cells were cultured overnight in a 12-well plate. The culture medium was replaced with 400 ill pre stimulation buffer for 10 minutes at room temperature to stop endogenous phosphodiesterase. Serial dilutions of BNP or BNP-HSA fusion proteins were applied to the cells. The cells were incubated on a plate shaker at 37 C for 15 minutes. The cells were then lysed in 100 I lysis buffer and the cGMP levels were determined by CatchPoint cGMP
Assay Kit (Molecular Devices). (.) BNP; (0) BNP/HSA CID 3448; (.) BNP2x/HSA
CID
3477.
[0016]
Figure 5 shows the effect of various dilutions of IFNb albumin fusion proteins encoded by DNA comprised in CID 2011 and 2053 on SEAP activity in the ISRE-SEAP/293F reporter cells (see Example 77). Proteins were serially diluted from 5e-7 to le-14 g/ml in DMEM/10% FBS and used to treat ISRE-SEAP/293F reporter cells. After hours supernatants were removed from reporter cells and assayed for SEAP
activity. IFNb albumin fusion protein was purified from three stable clones: 293F/#2011, CH0/#2011 and NSO/#2053. Mammalian derived IFNb, Avonex, came from Biogen and was reported to have a specific activity of 2.0e5 IU/ug.
[0017]
Figure 6 compares the anti-proliferative activity of IFN albumin fusion protein encoded by CID 3165 (CID 3165 protein) and recombinant IFNa (rIFNa) on Hs294T
melanoma cells. The cells were cultured with varying concentrations of either 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. =

protein, (6) = rIFNa.
[0018]
Figure 7 shows the effect of various dilutions of IFNa albumin fusion proteins on SEAP activity in the ISRE-SEAP/293F reporter cells. One preparation of IFNa fused upstream of albumin (6) was tested, as well as two different preparations of IFNa fused downstream of albumin (a) and ( = ).
[0019]
Figure 8 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 79). Per time point: first bar =
Vehicle control, 2nd bar = 30 ug/kg CID 2249 protein day 1 iv, third bar = 30 ug/kg CID 2249 protein day 1 Sc, 4th bar = 300 ug/kg CID 2249 protein day 1 Sc, 5th bar = 40 ug/kg recombinant IFNa day 1, 3 and 5 Sc.
DETAILED DESCRIPTION
Definitions [0020] The following definitions are provided to facilitate understanding of certain terms used throughout this specification.
[0021] As used herein, "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 ID NO:Z; a nucleic acid molecule having a nucleotide sequence encoding an albumin fusion protein of the invention generated as described in Table 2 or in the Examples; a nucleic acid molecule having a nucleotide sequence encoding a Therapeutic albumin fusion protein of the invention, 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).
[0022] As used herein, "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 encoding at least one molecule of a Therapeutic protein (or fragment or variant thereof), further comprising, for example, one or more of the following elements: (1) a functional self-replicating vector (including but not limited to, a shuttle vector, an expression vector, an integration vector, and/or a replication system), (2) a region for initiation of transcription (e.g., a promoter region, such as for example, a regulatable or inducible promoter, a constitutive promoter), (3) a region for termination of transcription, (4) a leader sequence, and (5) a selectable marker. The polynucleotide encoding the Therapeutic protein and albumin protein, once part of the albumin fusion construct, may each be referred to as a "portion," "region" or "moiety" of the albumin fusion construct.
[0023] 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. As used herein, "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"). In a highly preferred embodiment, 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).
[0024] In a further preferred embodiment, 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 0- linked glycosylation); specific proteolytic cleavages; and assembly into multimeric proteins. An albumin fusion protein of the invention is preferably in the processed form. In a most preferred embodiment, 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".
[0025] In several instances, 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.
[0026] In one embodiment, the invention provides a polynucleotide encoding an albumin fusion protein comprising, or alternatively consisting of, a Therapeutic protein and a serum albumin protein. In a further embodiment, the invention provides an albumin fusion protein comprising, or alternatively consisting of, a Therapeutic protein and a serum albumin protein. In a preferred embodiment, 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. In a further preferred embodiment, the invention provides a polynucleotide encoding an albumin fusion protein whose sequence is shown as SEQ ID NO:Y in Table 2. In other embodiments, 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. In other embodiments, 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. In preferred embodiments, 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.
[0027] In further embodiments, 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. In further embodiments, 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. In preferred embodiments, the Therapeutic protein portion of the albumin fusion protein is the mature portion of the Therapeutic protein. In a further preferred embodiment, the Therapeutic protein portion of the albumin fusion protein is the extracellular soluble domain of the Therapeutic protein. In an alternative embodiment, 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.
[0028] In further embodiments, 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. In preferred embodiments, 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.

Therapeutic proteins [0029] As stated above, 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.
[0030] 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.
[0031] As used herein, "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 biologics. (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. Thus 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.
[0032] By 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. As a non-limiting example, a "Therapeutic protein" is a protein that is useful to treat, prevent or ameliorate a disease, condition or disorder. As a non-limiting example, 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.
[0033] For example, 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, GLP-1, GLP-2, PACAP-27, PACAP-28, VIP, CD4M33, secretin, glicentin, oxyntomodulin, PHM, IFNa, IFN13, ANP, BNP, NGF, BDNF, GDNF, and somatostatin.

[0034] 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, 251:543-545 (1974); and Johnson, Texas Reports Biol Med, 35:357-369 (1977)). Each interferon hybrid albumin fusion protein can be used to treat, prevent, or ameliorate viral infections (e.g., hepatitis (e.g., HCV); or HIV), multiple sclerosis, or cancer.
[0035] In one embodiment, 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). For example, 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. In a further embodiment, the AID hybrid is fused at the common BgIII
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. For example, this AID hybrid would comprise the amino acid sequence:
CDLPQTHSLGSRRTLMLLAQMRXIISLFSCLKDRHDFGFPQEEFGNQFQKAETIPVLHE

SILAVKKYFRRITLYLTEKKYSPCAWEVVRAEIMRSLSLSTNLQERLRRKE (SEQ ID
NO:99), wherein the X1 is R or K and the X2 is A or V (see, for example, Construct ID
#2875). In an additional embodiment, the AID hybrid is fused at the common PvuIII
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. For example, this AID hybrid would comprise the amino acid sequence:
CDLPQTHSLGSRRTLMLLAQMRXIISLFSCLKDRHDFGFPQEEFGNQFQKAETIPVLHE

SILAVICKYFRRITLYLTEKKYSPCAWEVVRAEIMRSLSLSTNLQERLRRKE (SEQ ID
NO:100), wherein the X1 is R or K and the second X2 is A or V (see, for example, Construct ID #2872). These hybrids are further described in U.S. Patent No. 4,414,510.
[0036] In an additional embodiment, 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. In a further embodiment, the A/F hybrid is fused at the common PvuIII
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. For example, this A/F hybrid would comprise the amino acid sequence:
CDLPQTHSLGSRRTLMLLAQMRXISLFSCLKDRHDFGFPQEEFGNQFQKAETIPVLHE
MIQQIFNLFSTKDS SAAWDETLLDKFYTELYQ QLNDMEA CVIQEV GVEETPLMNVD SI
LAVKICYFQRITLYLTEKKYSPCAWEVVRAEIMRSFSLSKIFQERLRRICE (SEQ ID
NO:101), wherein X is either R or K (see, for example, Construct ID #2874).
These hybrids are further described in U.S. Patent No. 4,414,510.
In a further embodiment, 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. In an additional embodiment, the A/B hybrid is fused at the common Pvulli 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. For example, this A/B hybrid would comprise an amino acid sequence:
CDLPQTHSLGSRRTLMLLAQMRXIISLFSCLKDRHDFGFPQEEFGNQFQKAETIPVLHE

DSILAVRKYFQRITLYLTEKKYSSCAWEVVRAEIMRSFSLSINLQKRLKSKE (SEQ ID
NO:102), wherein the X1 is R or K and X2 through X5 is SCVM or VLCD (see, for example, Construct ID #2873). These hybrids are further described in U.S. Patent No.
4,414,510.
[0037] In another embodiment, 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). For example, 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). In a further embodiment, the beta-1/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. For example, this beta-1/alpha D hybrid would comprise an amino acid sequence:
MSYNLLGFLQRSSNFQCQKLLWQLNGRLEYCLKDRMNFDIPEEIKQLQQFQKEDAAL
TIYEMLQNIFAlFRQDSSAAWDEDLLDKFCTELYQQLNDLEACVMQEERVGETPLMN
XDSILAVKKYFRRITLYLTEKKYSPCAWEVVRAEIMRSLSLS TNLQERLRRKE (SEQ

ID NO:103), wherein X is A or V. These hybrids are further described in U.S.
Patent No.
4,758,428.
[00381 In another embodiment, 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). For example, the alpha-beta hybrid portion of the interferon hybrid albumin fusion protein consists, or alternatively comprises, of interferon alpha D (also referred to as interferon alpha-1) fused to interferon beta-1. In a further embodiment, 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. For example, this alpha D/beta-1 hybrid would have an amino acid sequence:
MCDLPETHSLDNRRTLMLLAQMSRISPSSCLMDRHDFGFPQEEFDGNQFQKAPAISVL

MSSLHLKRYYGRILHYLKAKEYSHCAWTIVRVEILRNFYFINRLTGYLRN (SEQ ID
NO:104). These hybrids are further described in U.S. Patent No. 4,758,428.
[00391 In another embodiment, IFN-beta-HSA fusions are used to effectively inhibit antiviral activity against Ebola virus and the SARS virus (Toronto-2 strains).
The in vitro antiviral activity of IFN-beta fused upstream of mature HSA (CID 2053 protein) was evaluated against Ebola virus and SARS virus in Vero cells. These cells were used to assess the protective effects of CID 2053 protein based on inhibition of cytopathic effect (CPE) and the neutral red assay of cell viability. In vitro signal transduction was assessed by analysis of gene expression. The pharrnacolcineties and pharmacodynamics of CID 2053 protein were evaluated in rhesus monkeys. The results indicate that potent in vitro antiviral activity was achieved with a favorable safety index. The IC50 for CID 2053 protein was 0.4 rig/ml against Ebola and 2 ng/ml against the SARS virus. Array analysis showed that CID 2053 protein and IFN-beta induce the expression of a similar set of genes and trigger the IFN-stimulated response element (ISRE) signal transduction pathway. In rhesus monkeys administered a dose of 50 ug/kg IV or SC or 300 ug/kg SC, CID 2053 protein demonstrated favorable phannacokinetic properties. The terminal half-life was 36-40 hours and induced sustained increases in serum neopterin levels and OAS1 mRNA expression.
[0040] In a further embodiment, IFN-alpha-HSA fusions are used to effectively inhibit viral agents classified under Category A- Fib o (Ebola), Arena (Pichende), Category B-Toga (VEE) or Category C- Bunya (Punto toro), Flavi (Yellow fever, West Nile).
CPE
inhbition, neural red staining and virus yield assays were employed to evaluate the anti-viral activities of INF-alpha fused downstream of HSA (CID 3165 protein). The pharmacokinetics and pharmacodynamic activity of CID 3165 protein in cynomolgus monkeys and human subjects were evaluated. The results indicate that potent antiviral activity was achieved against all the RNA viruses evaluated with a favorable safety index. The IC50 values ranged from <0.1 ng/ml (Punta Toro A) to 19 ng/ml (VEE) in the CPE assay. In cynomolgus monkeys, the half-life of CID 3165 protein was 90 hours and was detectable up to 14 days post-dose. In human subjects, CID 3165 protein was safe and well tolerated.
Cma, following single injection doses was dose-proportional. The mean Cm ax in the 500 ug cohort was 22 ng/ml, and the mean ti/2 of 150 hours. Dosing once every 2-4 weeks is supported by the pharmacokinetics. Antiviral response against Hepatitis C was observed in 58%
of subjects in the single injection cohorts (120-500 ug).
[0041] In further embodiments, 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. In additional embodiments, 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.
[0042] The above-described interferon hybrid albumin fusion proteins are encompassed by the invention, as are host cells and vectors containing polynucleotides encoding the polypeptides. In one embodiment, a interferon hybrid albumin fusion protein encoded by a polynucleotide as described above has extended shelf life. In an additional embodiment, 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.
[0043] In another non-limiting example, 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 HIV-1 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).
[0044] As used herein, "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.
As an example, when BNP is the Therapeutic protein, the effects of BNP on cGMP
induction as shown in Figure 4 may be used as the endpoint for which therapeutic activity is measured.
Such 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.
[0045] Therapeutic proteins corresponding to a Therapeutic protein portion of an albumin fusion protein of the invention, such as cell surface and secretory proteins, 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. There are usually two major types of glycosylation:
glycosylation characterized by 0-linked oligosaccharides, which are attached to serine or threonine residues; and 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) is usually the terminal residue of both N-linked and 0-linked oligosaccharides. 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.
[0046] Therapeutic proteins corresponding to a Therapeutic protein portion of an albumin fusion protein of the invention, as well as analogs and variants thereof, 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. For example, 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. coil or glycosylation-deficient yeast. These approaches are described in more detail below and are known in the art.
[0047] 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). 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)). 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 NO: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.
[0048] 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 variable thereof [0049] 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. Column 3, "Exemplary Activity Assay,"
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.
The fourth column, "Preferred Indication: Y," 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.

Table 1 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic Protein:X Assay Protein:Z
CD4M33 Inhibits infection of Inhibition of HIV
HIV, AIDS, viral infection. __ 3583, 3584. __ X INLHFCQL
cells by HIV-1 by infection into cultured RCKSLGLLG
binding to the CD4 cells can be measured binding site on the using methods known CV (SEQ ID
HIV-1 exterior in the art, for example, NO:549) envelope and as described in Martin wherein X1 =
inhibiting HIV-1 cell et al. Nature thiopropionic entry. Biotechnology 21:71-acid (Tpa) and 76 (2003).

biphenylalanin e (Bip) (Martin et al.

Nature Biotechnology 21:71-76 (2003)).

GLP-2 Stimulates Intestinal epithelial cell Most preferred:
Gastrointestinal 3518, 3519. __ See Table 2, (Glucagon- proliferation and proliferation can be disorders including, but not limited to: __ SEQ ID
NO:Z
Like Peptide- inhibits apoptosis of measured using gastrointestinal epithelial injury; for particular 2) intestinal epithelial methods known in the recovery from bowel resection; enteritis; construct; See cells; reduces art, including the cell colitis; gastritis; chemotherapy-induced Endocrine epithelial proliferation assays mucositis; short bowel syndrome; Reviews permeability; described in Dig. Dis. intestinal atrophy;
inflammatory bowel 21(6):619-670 decreases gastric acid Sci. 47(5):1135-40 disease; Crolm's disease;
Ulcerative (2000).
secretion and (2002). colitis; acid reflux;
peptic ulcers;

Table 1 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic Protein:X _Assay Protein:Z
gastrointestinal diabetes-associated bowel growth;
motility. Protection of intestinal intestinal ischemia syndromes;
epithelium can be maintenance of gut integrity after major evaluated using burn trauma; regulation of intestinal methods known in the permeability and nutrient absorption.
art, including the in vitro intestinal injury Also preferred:
Hyperglycemia;
model described in J. Diabetes; Diabetes Insipidus; Diabetes Surg. Res 107(1):44-9 mellitus; Type 1 diabetes; Type 2 (2002). diabetes; Insulin resistance; Insulin deficiency; Hyperlipidemia;
Hyperketonemia; Non-insulin dependent Diabetes Mellitus (NIDDM); Insulin-dependent Diabetes Mellitus (IDDM); A
Condition Associated With Diabetes Including, But Not Limited To Obesity, Heart Disease, Hyperglycemia, Infections, Retinopathy, And/Or Ulcers;
Metabolic Disorders; Immune Disorders;
Obesity; Vascular Disorders;
Suppression of Body Weight;
Suppression of Appetite; Syndrome X.
GLP-2 analog Stimulates Intestinal epithelial cell Most preferred:
Gastrointestinal 3535, 3536. See Table 2, ALX-0600 proliferation and proliferation can be disorders including, but not limited to: SEQ ID
NO:Z
(G1y2GLP-2) inhibits apoptosis of measured using gastrointestinal epithelial injury; for particular intestinal epithelial methods known in the recovery from bowel resection; enteritis; construct.
cells; reduces art, including the cell colitis;_gastritis; chemotherapy-induced Table 1 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic t..) o o Protein:X Assay Protein: Z u, epithelial proliferation proliferation assays mucositis; short bowel syndrome;
,...) t..) permeability; described in Dig. Dis. intestinal atrophy;
inflammatory bowel o o decreases gastric acid Sci. 47(5):1135-40 disease; Crohn's disease; Ulcerative secretion and (2002). colitis; acid reflux;
peptic ulcers;
gastrointestinal diabetes-associated bowel growth;
motility. Protection of intestinal intestinal ischemia syndromes;
epithelium can be maintenance of gut integrity after major evaluated using burn trauma; regulation of intestinal n methods known in the permeability and nutrient absorption.

I.) art, including the in Ul H
vitro intestinal injury Also preferred:
Hyperglycemia; us, I.) 00 model described in J.
Diabetes; Diabetes Insipidus;
Diabetes us, Surg. Res 107(1):44-9 mellitus; Type 1 diabetes; Type 2 I.) (2002). diabetes; Insulin resistance; Insulin , deficiency; Hyperlipidemia;

H
Hyperketonemia; Non-insulin dependent I.) Diabetes Mellitus (NIDDM); Insulin-dependent Diabetes Mellitus (IDDM); A
Condition Associated With Diabetes Including, But Not Limited To Obesity, Heart Disease, Hyperglycemia, n Infections, Retinopathy, And/Or Ulcers;
Metabolic Disorders; Immune Disorders;
cp t..) Obesity; Vascular Disorders;
=
o .6.
Suppression of Body Weight;
O-o Suppression of Appetite; Syndrome X.
.
,...) o Table 1 ,0 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic t..) o Protein:X Assay Protein:Z o u, (Pituitary secretion; enhances 27 on glucose uptake Diabetes; Diabetes Insipidus; Diabetes SEQ ID NO:Z
,...) Adenylate insulin-induced can be measured using mellitus; Type 1 diabetes; Type 2 for particular t..) o o Cylease glucose uptake; methods known in the diabetes; Insulin resistance; Insulin construct;
Activating stimulates gastric art, including the [3-H]- deficiency;
Hyperlipidemia; See Endocrine ' Polypeptide- acid secretion; glucose uptake assay. (J Hyperketonemia; Non-insulin dependent Reviews 27) stimulates adenylate Biol Chem 1999 Oct Diabetes Mellitus (NIDDM); Insulin- 21(6):619-cyclase. Protects 22; 274(43):30864- dependent Diabetes Mellitus (IDDM); A (2000), neurons from gp120- 30873). Condition Associated With Diabetes incorporated n mediated toxicity. Including, But Not Limited To Obesity, by reference.

Induces Insulin secretion can be Heart Disease, Hyperglycemia, N) Ul H
brochodilation and measured by methods Infections, Retinopathy, And/Or Ulcers;
us, .
I.) vasodilation. known in the art, Metabolic Disorders;
Immune Disorders; H
UJ
including the MIN6 cell Obesity; Vascular Disorders;
N) assay described in Ann. Suppression of Body Weight;

NY Acad. Sci. 805:44- Suppression of Appetite; Syndrome X.

51 (1996).
H
I.) Also preferred: Prevention of Gp120 neuroprotection neurotoxicity, such as, for example, can be measured using gp120-mediated neurotoxicity;
the neuroprotection Cardiovascular disorders, including but assays described in not limited to hypertension, stroke, and Neuropeptides 36(4): congestive heart failure; pulmonary n ,-i 271-80. disorders, including but not limited to cp asthma and allergy.
t..) o o Bronchodilation can be .6.
measured using, using, for o ,...) example, the isolated o o _ _ Table 1 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic Protein:X Assay Protein:Z
, _ rabbit tracheal smooth muscle assay described in Res Commun Chem Pathol Pharmacol 79(1):11-22 (1993).
Vasodilation can be measured using, for 1.) example, the 1-, vasodilation assay w 1.) 1-, N.) described in Pharmacol w Res. 39(3):217-20 1.) 1-, (1999).
, PACAP-38 Stimulates insulin The effect of PACAP- Most preferred:
Hyperglycemia; Obesity; 3539, 3540. See Table 2, 0 i (Pituitary secretion; enhances 38 on glucose uptake Diabetes; Diabetes Insipidus; Diabetes SEQ ID NO:Z "

Adenylate insulin-induced can be measured using mellitus; Type 1 diabetes; Type 2 for particular Cylcase glucose uptake; methods known in the diabetes; Insulin resistance; Insulin construct;
Activating stimulates gastric art, including the [3-H]- deficiency;
Hyperlipidemia; See Endocrine Polypeptide- acid secretion; glucose uptake assay. (J Hyperketonemia;
Non-insulin dependent Reviews 38) stimulates adenylate Biol Chem 1999 Oct Diabetes Mellitus (NIDDM); Insulin- 21(6):619-670 cyclase. Protects 22; 274(43):30864- dependent Diabetes Mellitus (IDDM); A (2000).
neurons from gp120- 30873). Condition Associated With Diabetes mediated toxicity. Including, But Not Limited To Obesity, Induces Insulin secretion can be Heart Disease, Hyperglycemia, brochodilation and measured by methods Infections, Retinopathy, And/Or Ulcers;
vasodilation. known in the art, Metabolic Disorders;
Immune Disorders;
including the MIN6 cell,Obesity; Vascular Disorders;

Table!
Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic Protein:X Assay Protein:Z
assay described in Ann. Suppression of Body Weight;
NY Acad. Sci. 805:44- Suppression of Appetite; Syndrome X.
51 (1996).
Also preferred: Prevention of Gp120 neuroprotection neurotoxicity, such as, for example, can be measured using gp120-mediated neurotoxicity;
the neuroprotection Cardiovascular disorders, including but assays described in not limited to hypertension, stroke, and Neuropeptides 36(4): congestive heart failure; pulmonary 271-80. disorders, including but not limited to UJ
asthma and allergy.
Bronchodilation can be us, measured using, for example, the isolated rabbit tracheal smooth muscle assay described in Res Commun Chem Pathol Pharmacol 79(1):11-22 (1993).
Vasodilation can be measured using, for example, the vasodilation assay described in Pharmacol Res. 39(3):217-20 (1999).

Table 1 o Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic t..) o Protein:X Assay Protein:Z o u, VIP Stimulates insulin The effect of VIP on Most preferred: Hyperglycemia; Obesity; 3568, 3569.
See Table 2, O-o ,...) (vasoactive secretion; stimulates insulin secretion can be Diabetes;
Diabetes Insipidus; Diabetes SEQ ID NO:Z
o, intestinal glycogenolysis; measured by methods mellitus; Type 1 diabetes; Type 2 for particular peptide) Induces known in the art, diabetes; Insulin resistance; Insulin construct;
brochodilation and including the MIN6 cell deficiency;
Hyperlipidemia; See Endocrine vasodilation. assay described in Ann. Hyperketonemia; Non-insulin dependent Reviews NY Acad. Sci. 805:44- Diabetes Mellitus (NIDDM); Insulin-21(6):619-670 51 (1996). dependent Diabetes Mellitus (IDDM); A (2000), n Condition Associated With Diabetes incorporated 0 Bronchodilation can be Including, But Not Limited To Obesity, by reference. "
Ul H
measured using, for Heart Disease, Hyperglycemia, us, tµ.) I.) t\.) example, the isolated Infections, Retinopathy, And/Or Ulcers; H
UJ
rabbit tracheal smooth Metabolic Disorders; Immune Disorders;
"

muscle assay described Obesity; Vascular Disorders;

in Res Commun Chem Suppression of Body Weight;

Pathol Pharmacol Suppression of Appetite;
Syndrome X. H
I.) 79(1):11-22 (1993).
Also preferred: Cardiovascular disorders, Vasodilation can be including but not limited to measured using, for hypertension, stroke, and congestive , example, the heart failure; pulmonary disorders, vasodilation assay including but not limited to asthma and n ,-i described in Pharmacol allergy.
Res. 39(3):217-20 t..) o o (1999).
.6.
O-secretin Stimulates insulin The effect of secretin Most preferred:
Hyperglycemia; Obesity; 3570, 3571. SEQ ID NO :Z
,...) secretion; stimulates on insulin secretion can Diabetes; Diabetes Insipidus;
Diabetes for particular o, ,z Table 1 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic t..) o Protein:X Assay Protein:Z
u, cAMP production; be measured-by mellitus; Type 1 diabetes; Type 2 construct; -o-o ,...) stimulates secretion methods known in the diabetes; Insulin resistance;
Insulin See Endocrine tt,' o of bicarbonate-rich art, including the MIN6 deficiency; Hyperlipidemia;
Reviews fluid from pancreas; cell assay described in Hyperketonemia; Non-insulin dependent 21(6):619-670 stimulates gastric Ann. NY Acad. Sci. Diabetes Mellitus (NIDDM); Insulin- (2000), pepsin secretion; 805:44-51 (1996). dependent Diabetes Mellitus (IDDM); A incorporated inhibits gastric acid Condition Associated With Diabetes by reference.
secretion and gut cAMP accumulation Including, But Not Limited To Obesity, n motility. Stimulates can be measured using Heart Disease, Hyperglycemia, bile production. methods known in the Infections, Retinopathy, And/Or Ulcers; I.) Ul H
art, including the in Metabolic Disorders;
Immune Disorders; us, IQ
I.) 4) H
vitro assay described in Obesity; Vascular Disorders;
us, Br J Pharmacol Suppression of Body Weight; I.) 138(4):660-70 (2003). Suppression of Appetite; Syndrome X.

u-, I
Also preferred: Autism; Gastrointestinal H
I.) disorders, including but not limited to Inflammatory Bowel Disease; Crohn's disease; and ulcerative colitis.
glicentin Stimulates insulin The effect of glicentin Most preferred:
Hyperglycemia; Obesity; 3577, 3578. SEQ ID NO:Z
secretion; stimulates on insulin secretion can Diabetes; Diabetes Insipidus;
Diabetes for particular cAMP production; be measured by mellitus; Type 1 diabetes; Type 2 construct. n ,-i inhibits meal- methods known in the diabetes; Insulin resistance; Insulin cp stimulated gastric art, including the MIN6 deficiency;
Hyperlipidemia; t..) o o acid secretion; cell assay described in Hyperketonemia; Non-insulin dependent .6.
regulates gutgut Ann. NY Acad. Sci. Diabetes Mellitus (NIDDM); Insulin-,...) motility; inhibits 805:44-51 (1996). dependent Diabetes Mellitus (IDDM); A o o Table 1 .
.o Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic t..) o Protein:X Assay Protein:Z
u, --food intake. Condition Associated With Diabetes o =
(.#4 cAMP accumulation Including, But Not Limited To Obesity, t..) o o can be measured using Heart Disease, Hyperglycemia, methods known in the Infections, Retinopathy, And/Or Ulcers;
art, including the in Metabolic Disorders;
Immune Disorders; , vitro assay described in Obesity; Vascular Disorders;
Br J Pharmacol Suppression of Body Weight;
138(4):660-70 (2003). Suppression of Appetite; Syndrome X.
n oxyntomodulin Stimulates insulin The effect of Most preferred:
Hyperglycemia; Obesity; 3579, 3580. SEQ ID NO:Z "
Ul H
secretion; stimulates oxyntomodulin on Diabetes; Diabetes Insipidus; Diabetes for particular IQ
I.) cAMP production; insulin secretion can be mellitus; Type 1 diabetes; Type 2 construct.
inhibits meal- measured by methods diabetes; Insulin resistance; Insulin "

stimulated gastric known in the art, deficiency;
Hyperlipidemia;

acid secretion; including the MIN6 cell Hyperketonemia; Non-insulin dependent 0 regulates gut assay described in Ann. Diabetes Mellitus (NIDDM); Insulin- H
I.) motility; inhibits NY Acad. Sci. 805:44- dependent Diabetes Mellitus (IDDM); A
food intake. 51 (1996). Condition Associated With Diabetes Including, But Not Limited To Obesity, cAMP accumulation Heart Disease, Hyperglycemia, can be measured using Infections, Retinopathy, And/Or Ulcers;
methods known in the Metabolic Disorders; Immune Disorders;
n ,-i art, including the in Obesity; Vascular Disorders;
cp vitro assay described in Suppression of Body Weight;
t..) o o Br J Pharmacol Suppression of Appetite;
Syndrome X. .6.
O-138(4):660-70 (2003).
,...) PHM (Peptide Stimulates insulin The effect of PHM on Most preferred:
Hyperglycemia; Obesity; 3581, 3582. SEQ ID NO:Z o o Table 1 o Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic t..) =
Protein:X Assay ,Protein:Z =
u, 7a3 Histidine secretion; stimulates insulin secretion can be Diabetes;
Diabetes Insipidus; Diabetes for particular o t...) Methionine) glycogenolysis; measured by methods mellitus; Type 1 diabetes; Type 2 construct. t..) ,z o, Induces known in the art, diabetes; Insulin resistance; Insulin See Endocrine brochodilation and including the MIN6 cell deficiency; Hyperlipidemia;
Reviews vasodilation. assay described in Ann. Hyperketonemia; Non-insulin dependent 21(6):619-670 NY Acad. Sci. 805:44- Diabetes Mellitus (NIDDM); Insulin-(2000), 51 (1996). dependent Diabetes Mellitus (IDDM); A incorporated Condition Associated With Diabetes by reference. n Bronchodilation can be Including, But Not Limited To Obesity, measured using, for Heart Disease, Hyperglycemia, I.) Ul H
example, the isolated Infections, Retinopathy, And/Or Ulcers;
us, t.) I.) (J) rabbit tracheal smooth Metabolic Disorders;
Immune Disorders; H
UJ
muscle assay described Obesity; Vascular Disorders;
N) in Res Commun Chem Suppression of Body Weight;

Pathol Pharmacol Suppression of Appetite;
Syndrome X. 0 79(1):11-22 (1993).
H
I.) Also preferred: Cardiovascular disorders, Vasodilation can be including but not limited to measured using, for hypertension, stroke, and congestive example, the heart failure; pulmonary disorders, vasodilation assay including but not limited to asthma and described in Pharmacol allergy.
n ,-i Res. 39(3):217-20 (1999).
t..) o o (Interferon cellular cellular responses Rubinstein S, Familletti Respiratory Syndrome (SARS) and other 2381, 2382, 2410, SEQ ID NO:Z
t...) alfa-2b; including antiviral, PC, Pestka S. (1981) coronavirus infections; filoviruses, 3165, 3422, 3423, for particular o, ,z Table 1 , ,o Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic t..) o Protein:X Assay Protein:Z
u, recombinant; antiproliferative, Convenient assay for including but not limited to Ebola 3424. construct. O-o ,...) Interferon alfa- antitumor and interferons. J. Virol.
viruses and Marburg virus;
Arenaviruses, t..) o o n1; Interferon immunomodulatory 37(2):755-8; Anti- including but not limited to Pichende alfa-n3; activities; stimulate proliferation assay: virus, Lassa virus, Junin virus, Machupo Peginterferon production of two Gao Y, et al (1999) virus, Guanarito virus; and lymphocytic alpha-2b; enzymes: a protein Sensitivity of an choriomeningitis virus (LCMV);
Ribavirin and kinase and an epstein-barr virus- Bunyaviruses, including but not limited interferon alfa- oligoadenylate positive tumor line, to Punta toro virus, Crimean-Congo n 2b; Interferon synthetase. Daudi, to alpha hemorrhagic fever virus, sandfly fever alfacon-1; interferon correlates viruses, Rift Valley fever virus, La I.) Ul H
interferon with expression of a Crosse virus, and hantaviruses; L.., t..) I.) (:7H
consensus; GC-rich viral Flaviviruses, including but not limited to L.., YM 643; transcript. Mol Cell Yellow Fever, Banzi virus, West Nile I.) CIFN; Biol. 19(11):7305-13. virus, Dengue viruses, Japanese 0 u-, interferon - Encephalitis virus, Tick-borne 0 alpha encephalitis, Omsk Hemorrhagic Fever, H
IV
consensus; and Kyasanur Forest Disease virus;
recombinant Togaviruses, including but not limited to methionyl Venezuelan, eastern, and western equine consensus encephalitis viruses, Ross River virus, interferon; and Rubella virus;
Orthopox viruses, ,-o recombinant including but not limited to Vaccinia, n ,-i consensus Cowpox, Smallpox, and Monkeypox;
cp interferon; Herpesviruses; FluA/B;
Respiratory t..) o CGP 35269; Sincytial virus (RSV);
paraflu; measles; o .6.
O-RO 253036; rhinoviruses;
adenoviruses; Semliki =
RO 258310; Forest virus; Viral Hemorrhagic fevers; ,...) o o Table 1 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic a) Protein:X Assay Protein:Z
INTRON A; Rhabdoviruses;
Paramyxoviruses, PEG- including but not limited to Nipah virus INTRON; and Hendra virus; and other viral agents OIF; identified by the U.S.
Centers for OMNIFERON Disease Control and Prevention as high-PEG- priority disease agents (i.e., Category A, OMNIFERON B, and C agents; see, e.g., Moran, ; VELDONA; Emerg. Med. Clin. North.
Am. 2002;
PEG- 20(2):311-30 and Darling et al., Emerg. 0 REBETRON; Med. Clin. North Am.
2002;20(2):273-ROFERON A; 309).
UJ
WELLFERON

; ALFERON

N/LDO;
REBETRON;
ALTEMOL;
VIRAFERON
PEG;
PEGASYS;
VIRAFERON;
VIRAFON;
AMPLIGEN;
INFERGEN;
INFAREX;
ORAGEN) Interferon beta Modulates MHC Anti-viral assay: Viral infections include Severe Acute 1778, 1779, 2011, See Table 2, Table 1 o Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic t..) o Protein:X Assay Protein:Z o u, (Interferon antigen expression, Rubinstein S, Familletti Respiratory Syndrome (SARS) and other 2013, 2053, 2054,O-SEQ ID NO:Z =
,...) beta- 1 a; NK cell activity and PC, Pestka S. (1981) coronavirus infections; filoviruses, 2492, 2580, 2795, for particular t..) o o Interferon beta IFNg production and Convenient assay for including but not limited to Ebola 2796, 2797. construct.
lb; Interferon- IL12 production in interferons. J. Virol. viruses and Marburg virus; Arenaviruses, beta-serine; monocytes. 37(2):755-8; Anti- including but not limited to Pichende SH 579; ZK proliferation assay: virus, Lassa virus, Junin virus, Machupo 157046; Gao Y, et al (1999) virus, Guanarito virus; and lymphocytic BCDF; beta-2 Sensitivity of an choriomeningitis virus (LCMV); n IF; Interferon- epstein-barr virus- Bunyaviruses, including but not limited beta-2; rhIL-6; positive tumor line, to Punta toro virus, Crimean-Congo "
u-, SJ0031; DL N) Daudi, to alpha hemorrhagic fever virus, sandfly fever H
UJ IV
' 8234; FERON; interferon correlates viruses, Rift Valley fever virus, La H
UJ
IFNbeta; with expression of a Crosse virus, and hantaviruses; "

BETASERON GC-rich viral Flaviviruses, including but not limited to 0 u-, ; AVONEX; transcript. Mol Cell Yellow Fever, Banzi virus, West Nile 0 REBIF; Biol. 19(11):7305-13. virus, Dengue viruses, Japanese H
I.) BETAFERON Encephalitis virus, Tick-borne ; SIGOSIX) encephalitis, Omsk Hemorrhagic Fever, and Kyasanur Forest Disease virus;
Togaviruses, including but not limited to Venezuelan, eastern, and western equine encephalitis viruses, Ross River virus, n ,-i and Rubella virus; Orthopox viruses, cp including but not limited to Vaccinia, t..) o o Cowpox, Smallpox, and Monlceypox;
.6.
O-Herpesviruses; FluA/B; Respiratory ' ,...) Sincytial virus (RSV); paraflu; measles;
o o -Table 1 t..) Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic =
=
Protein:X Assay Protein:Z u, , 7a3 =
rhinoviruses; adenoviruses; Semliki ,...) t..) ,z Forest virus; Viral Hemorrhagic fevers;
c, Rhabdoviruses; Paramyxoviruses, including but not limited to Nipah virus and Hendra virus; and other viral agents identified by the U.S. Centers for Disease Control and Prevention as high-priority disease agents (i.e., Category A, n B, and C agents; see, e.g., Moran, I.) Emerg. Med. Clin. North. Am. 2002;
Ul H
UJ
20(2):311-30 and Darling et al., Emerg.
I.) I..) H
vo Med. Clin. North Am.
2002;20(2):273- us, I.) 309).

u-, _ Atrial ANP is diuretic Renin and aldosterone Hypertension; salt-sensitive 3484 See Table 2, -1 I
H
natriuretic (natriuretic), levels can be measured hypertension;
congestive heart failure; SEQ ID NO:Z I.) peptide (ANP; hypotensive, and has using methods known kidney failure; excess fluid in tissues; for particular atrial an inhibitory effect in the art, for example, hypotension;
cardiac volume overload; construct.
natriuretic on renin and in Yamato et al., Circ J cardiac decompensation; left ventricular factor; ANF) aldosterone secretion. 2003 May;67(5):384- dysfunction; dyspnea;
treatment for Involved in 90. Blood pressure can elevated aldosterone levels, which can n regulation of blood be measured with a lead to vasoconstriction, impaired pressure and salt and sphygmomanometer or cardiac output and/or hypertension;
cp t..) water balance/ using other methods cardiovascular disease; cardiac failure; =
=
.6.
electrolyte known in the art, such myocardial reperfusion injury; left 7a3 =
homeostasis in body as in Reddy et al., ventricular remodeling.
.
,...) c, fluids. Ultrasound Med Biol ,z _ Table 1 o Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic t..) o Protein:X Assay Protein:Z
u, 2003 Mar;29(3):379-O-o ,...) ,z o, B-type stimulates smooth Inhibition of Congestive heart failure; cardiac volume 3404, 3448, 3477, See Table 2, natriuretic muscle relaxation angiotensin can be overload; cardiac decompensation; 3513, 3514, 3516, SEQ ID NO :Z
peptide (BNP, and vasodilation, determined using Cardiac Failure; Left Ventricular 3517, 3524, 3525, for particular brain natriuresis,and assays known in the art, Dysfunction;
Dyspnea. 3526, 3616, 3617, construct.
natriuretic suppression of renin- for example using an in 3618, 3619.
peptide) angiotensin and vitro proliferation assay n endothelin. with rat cardiac fibroblasts as described "
Ul H
LiJ in Naunyn us, I.) C) H
Schmiedebergs Arch us, Pharmacol 1999 "

May;359(5):394-9.

u-, Vasodilation can be I
measured in animals by H
I.) measuring the myogenic responses of small renal arteries in an isobaric arteriograph system (see Am J
Physiol Regul Integr n ,-i Comp Physiol 2002 cp Aug;283(2):R349-t..) o o R355). Natriuesis is .6.
determined by by o ,...) measuring the amounto, ,z __ _. _ Table 1 , , _______________________________________________________________________________ ____________________________________ Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic o t..) =
Protein:X Assay Protein:Z =
u, of sodium in the urine.
O-o NGF (nerve Neurotrophic factor NGF activity may be Pain; Neuropathic pain; Complex 3572, 3573. See Table 2, ,...) t..) o growth factor) that promotes assayed by regional pain syndrome I;
Reflex SEQ ID NO:Z o neuronal growth, measurement of CREB sympathetic dystrophy;
Trigeminal for particular differentiation, and transcription factor neuralgia; Allodynia;
Primary and/or construct.
survival; maintains activation in Secondary hyperalgesia;
Causalgia;
the survival of sympathetic neurons in Phantom limb pain;
Post-surgical pain;
sympathetic and culture (Science 1999 Burning feet syndrome;
Guillain-Barre n sensory neurons Dec syndrome; Thalamic syndrome; Post-during development 17;286(5448):2358- stroke pain; Vasculitic/
angiopathic pain; 0 I.) u-, w and in vitro; 2361). The ability of Idiopathic pain; Pain associated with any H
us, ,¨
I.) normalizes elevated NGF to affect pain of the following:
Entrapment H
UJ
tailflick threshold (a perception can be neuropathy, Nerve transection, Spinal I.) test that assesses assayed using the cord injury, Scar formation, Alcoholic 0 u-, perception of pain) in Tailflick test. neuropathy, Pellagra, Beriberi, Post- 0 rodent models, herpetic neuralgia, HIV/AIDS pain, H
IV
indicating reduction Vincristine neurotoxicity, Cisplatin of sensed/perceived neurotoxicity, Taxol neurotoxicity, pain. Thallium neurotoxicity, Arsenic neurotoxicity, Radiation therapy, Diabetes, Malignancies, Multiple ,-o sclerosis, Fabry's disease, Tangier n ,-i disease, or Amyloid.
NGF beta Neurotrophic factor NGF activity may be Pain; Neuropathic pain; Complex 3574, 3575. See Table 2, cp t..) o (nerve growth that promotes assayed by regional pain syndrome I;
Reflex SEQ ID NO:Z 4a factor beta; beta; neuronal growth, measurement of CREI3 sympathetic dystrophy;
Trigeminal for particular o NGF-B) differentiation, and transcription factor neuralgia; Allodynia; Primary and/or construct.
,...) o o Table 1 .

Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic . t..) o Protein:X Assay Protein:Z
, survival; maintains activation in Secondary hyperalgesia;
Causalgia; o ,...) the survival of sympathetic neurons in Phantom limb pain;
Post-surgical pain; t..) o sympathetic and culture (Science 1999 Burning feet syndrome;
Guillain-Barre sensory neurons Dec syndrome; Thalamic syndrome; Post-during development 17;286(5448):2358- stroke pain; Vasculitic/
angiopathic pain;
and in vitro; 2361). The ability of Idiopathic pain; Pain associated with any normalizes elevated NGF to affect pain of the following:
Entrapment tailflick threshold (a perception can be neuropathy, Nerve transection, Spinal n test that assesses assayed using the cord injury, Scar formation, Alcoholic perception of pain) in Tailflick test. neuropathy, Pellagra, Beriberi, Post- I.) u-, rodent models, herpetic neuralgia, HIV/AIDS pain, H
UJ
t.0-) IV
I.) indicating reduction Vincristine neurotoxicity, Cisplatin H
L.., of sensed/perceived neurotoxicity, Taxol neurotoxicity, I.) pain. Thallium neurotoxicity, Arsenic neurotoxicity, Radiation therapy, I
Diabetes, Malignancies, Multiple H
IV
sclerosis, Fabry's disease, Tangier disease, or Amyloid.
BDNF isoform Neurotrophic factor BDNF activity on Pain; Neuropathic pain;
Complex 3541, 3542. See Table 2, a (brain- that promotes neuronal growth can be regional pain syndrome I; Reflex SEQ ID NO:Z
derived neuronal growth, measured using sympathetic dystrophy;
Trigeminal for particular neurotrophic differentiation, and neuronal growth and neuralgia; Allodynia;
Primary and/or construct. n ,-i factor) survival; maintains synaptic activity assays, Secondary hyperalgesia; Causalgia;
cp the survival of such as those described Phantom limb pain;
Post-surgical pain; t..) o o subsets of peripheral in Bartrup et al (1997) Burning feet syndrome; Guillain-Barre .6.
and central central neurons Neuroreport syndrome; Thalamic syndrome; Post- o ,...) during development; 1:8(17):3791-4; BDNF stroke pain; Vasculitic/ angiopathic pain; o o Table 1 o Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic t..) =
=
Protein:X Assay Protein:Z u, 7a3 plays a role in a adult activity on pain Idiopathic pain; Pain associated with any =
,...) t..) nervous system reception can be of the following:
Entrapment o o function; may assayed by measuring neuropathy, Nerve transection, Spinal contribute to the nociceptive behaviors, cord injury, Scar formation, Alcoholic nociceptive hyperalgesia, and/or neuropathy, Pellagra, Beriberi, Post-responses. allodynia as described herpetic neuralgia, HIV/AIDS pain, in Shu et al Pain (1999) Vincristine neurotoxicity, Cisplatin 80:463-470 and in neurotoxicity, Taxol neurotoxicity, n Zhou et al Eur. J. Thallium neurotoxicity, Arsenic Neurosci. (2000) neurotoxicity, Radiation therapy, 0 I.) u-, 12:100-105. Diabetes, Malignancies, Multiple H
w us, wI.) sclerosis, Fabry's disease, Tangier H
UJ
disease, or Amyloid.
I.) BDNF isoform Neurotrophic factor BDNF activity on Pain; Neuropathic pain;
Complex 3543, 3544. SEQ ID NO:Z 0 u-, b (brain- that promotes neuronal growth can be regional pain syndrome I; Reflex for particular 0 I
derived neuronal growth, measured using sympathetic dystrophy;
Trigeminal construct. H
IV
neurotrophic differentiation, and neuronal growth and neuralgia; Allodynia;
Primary and/or factor) survival; maintains synaptic activity assays, Secondary hyperalgesia; Causalgia;
the survival of such as those described Phantom limb pain;
Post-surgical pain;
subsets of peripheral in Bartrup et al (1997) Burning feet syndrome; Guillain-Barre and central neurons Neuroreport syndrome; Thalamic syndrome; Post-n during development; 1:8(17):3791-4; BDNF stroke pain; Vasculitic/ angiopathic pain;
plays a role in a adult activity on pain Idiopathic pain; Pain associated with any cp t..) nervous system reception can be of the following:
Entrapment =
o function; may assayed by measuring neuropathy, Nerve transection, Spinal .6.
O-o contribute to the nociceptive behaviors, cord injury, Scar formation, Alcoholic .
,...) nociceptive hyperalgesia, and/or neuropathy, Pellagra, Beriberi, Post- o o Table 1 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic t..) o Protein:X Assay Protein:Z o u, responses. allodynia allodynia as described herpetic neuralgia, HIV/AIDS pain, =
,...) t..) in Shu et al Pain (1999) Vincristine neurotoxicity, Cisplatin o o 80:463-470 and in neurotoxicity, Taxol neurotoxicity, Zhou et al Eur. J. Thallium neurotoxicity, Arsenic Neurosci. (2000) neurotoxicity, Radiation therapy, 12:100-105. Diabetes, Malignancies, Multiple sclerosis, Fabry's disease, Tangier disease, or Amyloid.
n BDNF isoform Neurotrophic factor BDNF activity on Pain; Neuropathic pain;
Complex 3545, 3550. See Table 2, c (brain- that promotes neuronal growth can be regional pain syndrome I; Reflex SEQ ID NO:Z Ul"
H
W derived neuronal growth, measured using sympathetic dystrophy;
Trigeminal for particular us, I.) 4=, H
neurotrophic differentiation, and neuronal growth and neuralgia; Allodynia;
Primary and/or construct. us, factor) survival; maintains synaptic activity assays, Secondary hyperalgesia; Causalgia; I.) the survival of such as those described Phantom limb pain;
Post-surgical pain;

subsets of peripheral in Bartrup et al (1997) Burning feet syndrome; Guillain-Barre -1 I
H
and central neurons Neuroreport syndrome; Thalamic syndrome; Post- I.) during development; 1:8(17):3791-4; BDNF stroke pain; Vasculitic/ angiopathic pain;
plays a role in a adult activity on pain Idiopathic pain; Pain associated with any nervous system reception can be of the following:
Entrapment function; may assayed by measuring neuropathy, Nerve transection, Spinal contribute to the nociceptive behaviors, cord injury, Scar formation, Alcoholic n nociceptive hyperalgesia, and/or neuropathy, Pellagra, Beriberi, Post-responses. allodynia as described herpetic neuralgia, HIV/AIDS pain, cp in Shu et al Pain (1999) Vincristine neurotoxicity, Cisplatin t..) o o 80:463-470 and in neurotoxicity, Taxol neurotoxicity, .6.
O-o Zhou et al Eur. J. Thallium neurotoxicity, Arsenic .
,...) Neurosci. (2000) neurotoxicity, Radiation therapy, o o Table 1 o Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic t..) o Protein:X Assay Protein:Z
u, 12:100-105. Diabetes, Malignancies, Multiple O-o ,...) sclerosis, Fabry's disease, Tangier t..) ,z o, disease, or Amyloid.
NT3 (NT-3; Neurotrophic factor NT3 activity on Pain; Neuropathic pain;
Complex 3555, 3556. See Table 2, neurotrophin- that promotes neuronal growth can be regional pain syndrome I; Reflex SEQ ID NO:Z
3) neuronal growth, assayed in vitro by sympathetic dystrophy; Trigeminal for particular differentiation, and measuring its ability to neuralgia; Allodynia; Primary and/or construct.
survival; essential for proliferate cultured NC Secondary hyperalgesia;
Causalgia; n the survival of progenitor cells grown Phantom limb pain;
Post-surgical pain; 0 sensory and in a serum-free defined Burning feet syndrome; Guillain-Barre I.) u-, H
4.) sympathetic neurons medium (PNAS 1992 syndrome; Thalamic syndrome; Post- us, I.) (J1 during development; Marl; 89(5):1661- stroke pain; Vasculitic/
angiopathic pain; H
us, may play a role in 1665); activity on pain Idiopathic pain;
Pain associated with any I.) neuropathic pain, perception can be of the following:
Entrapment assayed by measuring neuropathy, Nerve transection, Spinal substance P release in cord injury, Scar formation, Alcoholic H
IV
response to C-fiber neuropathy, Pellagra, Beriberi, Post-stimulation in an in herpetic neuralgia, HIV/AIDS pain, vitro spinal cord Vincristine neurotoxicity, Cisplatin preparation, as in neurotoxicity, Taxol neurotoxicity, Malcangio et al Eur. J. Thallium neurotoxicity, Arsenic Neurosci (2000) neurotoxicity, Radiation therapy, n ,-i 12:139-144. Diabetes, Malignancies, Multiple cp sclerosis, Fabry's disease, Tangier t..) o o disease, or Amyloid.
.6.
O-GDNF (Glial- Neurotrophic factor Activity on neurons can Pain; Neuropathic pain; Complex 3551, 3552. See Table 2, o ,...) derived that promotes be assayed by regional pain syndrome I; Reflex SEQ ID NO:Z o, ,z Table 1 g Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic t..) o Protein:X Assay Protein:Z
u, O-neurotrophic neuronal growth, measuring increases in sympathetic dystrophy; Trigeminal for particular o ,...) factor) differentiation, and Ret tyrosine neuralgia; Allodynia;
Primary and/or construct. t..) ,z o, survival; a potent phosphorylation in Secondary hyperalgesia; Causalgia;
survival factor for response to GDNF Phantom limb pain; Post-surgical pain;
midbrain dopamine treatment; pain Burning feet syndrome;
Guillain-Barre neurons, perception activity can syndrome; Thalamic syndrome; Post-motorneurons, be measured using L5 stroke pain;
Vasculitic/ angiopathic pain;
noradrenergic spinal nerve ligation Idiopathic pain; Pain associated with any n neurons, as well as and partial sciatic nerve of the following:
Entrapment for sympathetic, ligation models of neuropathy, Nerve transection, Spinal I.) Ul H
parasympathetic and neuropathic pain, as in cord injury, Scar formation, Alcoholic us, I.) cA
H
sensory neurons; Boucher et al Science neuropathy, Pellagra, Beriberi, Post- us, promotes ureteric (2000) 290:124-127 herpetic neuralgia, HIV/AIDS pain, I.) branching in kidney and in Boucher et al Vincristine neurotoxicity, Cisplatin development and (2001) Curr. Opin. neurotoxicity, Taxol neurotoxicity, 0 regulates Pharmacol. 1:66-72. Thallium neurotoxicity, Arsenic H
I.) spermatogenesis; neurotoxicity, Radiation therapy, normalizes Diabetes, Malignancies, Multiple nociceptive sensory sclerosis, Fabry's disease, Tangier neuron physiology disease, or Amyloid.
following injury.
Neurturin Neurotrophic factor Activity on neurons can Pain; Neuropathic pain; Complex 3553, 3554. See Table 2, n ,-i (NTN; NRTN) that promotes be assayed by regional pain syndrome I; Reflex SEQ ID NO:Z
cp neuronal growth, measuring increases in sympathetic dystrophy; Trigeminal for particular t..) o o differentiation, and Ret tyrosine neuralgia; Allodynia;
Primary and/or construct. .6.
O-survival; a potent phosphorylation in Secondary hyperalgesia; Causalgia; o ,...) survival factor for response to NTN Phantom limb pain; Post-surgical pain; o, ,z Table 1 t..) Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic =
=
Protein:X Assay Protein:Z u, _ =
midbrain dopamine treatment; pain Burning feet syndrome;
Guillain-Barre ,...) t..) o neurons, perception activity can syndrome; Thalamic syndrome; Post- o motorneurons, be measured using L5 stroke pain;
Vasculitic/ angiopathic pain;
noradrenergic spinal nerve ligation Idiopathic pain;
Pain associated with any neurons, as well as and partial sciatic nerve of the following:
Entrapment for sympathetic, ligation models of neuropathy, Nerve transection, Spinal parasympathetic and neuropathic pain, as in cord injury, Scar formation, Alcoholic sensory neurons; Boucher et al Science neuropathy, Pellagra, Beriberi, Post- n promotes ureteric (2000) 290:124-127 herpetic neuralgia, HIV/AIDS pain, 0 I.) branching in kidney and in Boucher et al Vincristine neurotoxicity, Cisplatin Ul H
UJ
development and (2001) Curr. Opin. neurotoxicity, Taxol neurotoxicity, I.) t4-1 H
regulates Pharmacol. 1:66-72. Thallium neurotoxicity, Arsenic I.) spermatogenesis; neurotoxicity, Radiation therapy, 0 u-, normalizes Diabetes, Malignancies, Multiple 0 nociceptive sensory sclerosis, Fabry's disease, Tangier 1 H
neuron physiology disease, or Amyloid.
I.) following injury.
Persephin Neurotrophic factor Activity on neurons can Pain; Neuropathic pain; Complex 3557, 3558. See Table 2, (PSPN; PSP) that promotes be assayed by regional pain syndrome I; Reflex SEQ ID NO:Z
neuronal growth, measuring increases in sympathetic dystrophy; Trigeminal for particular differentiation, and Ret tyrosine neuralgia; Allodynia;
Primary and/or construct.
n survival; a potent phosphorylation in Secondary hyperalgesia; Causalgia;
survival factor for response to persephin Phantom limb pain;
Post-surgical pain; t..) o midbrain dopamine treatment; pain Burning feet syndrome;
Guillain-Barre =
.6.
neurons, perception activity can syndrome; Thalamic syndrome; Post- O-o motorneurons, be measured using L5 stroke pain;
Vasculitic/ angiopathic pain; ,...) o noradrenergic spinal nerve ligation Idiopathic pain; Pain associated with any o Table 1 , i Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic :F.) - o Protein:X Assay Protein:Z
u, neurons, as well as and partial sciatic nerve of the following:
Entrapment . o for sympathetic, ligation models of neuropathy, Nerve transection, Spinal : VD
: CJ`
parasympathetic and neuropathic pain, as in cord injury, Scar formation, Alcoholic sensory neurons; Boucher et al Science neuropathy, Pellagra, Beriberi, Post-promotes ureteric (2000) 290:124-127 herpetic neuralgia, HIV/AIDS pain, 1 branching in kidney and in Boucher et al Vincristine neurotoxicity, Cisplatin I
development and (2001) Curr. Opin. neurotoxicity, Taxol neurotoxicity, .
:
, regulates Pharmacol. 1:66-72. Thallium neurotoxicity, Arsenic = n spermatogenesis; neurotoxicity, Radiation therapy, normalizes Diabetes, Malignancies, Multiple I.) Ul H
nociceptive sensory sclerosis, Fabry's disease, Tangier L..., L...) I.) H
neuron physiology disease, or Amyloid.
L..., following injury.
I.) u-, isoform 1 that promotes be assayed by regional pain syndrome I; Reflex 1 and 2; SEQ 0 (neublastin; neuronal growth, measuring increases in sympathetic dystrophy; Trigeminal ID NO:Z for H
I.) enovin) differentiation, and Ret tyrosine neuralgia; Allodynia;
Primary and/or particular survival; a potent phosphorylation in Secondary hyperalgesia; Causalgia; construct.
survival factor for response to artemin Phantom limb pain;
Post-surgical pain;
midbrain dopamine treatment; pain Burning feet syndrome;
Guillain-Barre neurons, perception activity can syndrome; Thalamic syndrome; Post-motorneurons, be measured using L5 stroke pain;
Vasculitic/ angiopathic pain; n ,-i noradrenergic spinal nerve ligation Idiopathic pain;
Pain associated with any cp neurons, as well as and partial sciatic nerve of the following:
Entrapment t..) o o for sympathetic, ligation models of neuropathy, Nerve transection, Spinal .6.
parasympathetic andand neuropathic pain, as in cord injury, Scar formation, Alcoholic =
,...) sensory neurons; Boucher et al Science neuropathy, Pellagra, Beriberi, Post- o o Table 1 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic t..) o o Protein:X Assay Protein:Z u, promotes ureteric ureteric (2000) 290:124-127 herpetic neuralgia, HIV/AIDS pain, =
,...) t..) branching in kidney and in Boucher et al Vincristine neurotoxicity, Cisplatin o o development and (2001) Curr. Opin. neurotoxicity, Taxol neurotoxicity, regulates Pharmacol. 1:66-72. Thallium neurotoxicity, Arsenic spermatogenesis; neurotoxicity, Radiation therapy, normalizes Diabetes, Malignancies, Multiple nociceptive sensory sclerosis, Fabry's disease, Tangier neuron physiology disease, or Amyloid.
n following injury.

Artemin- Neurotrophic factor Activity on neurons can Pain; Neuropathic pain; Complex 3562, 3563. Splice variant "
Ul H
l.k.) isoform 2 that promotes be assayed by regional pain syndrome I; Reflex 3; SEQ ID us, I.) ' (neublastin; neuronal growth, measuring increases in sympathetic dystrophy;
Trigeminal NO:Z for H
UJ
enovin) differentiation, and Ret tyrosine neuralgia; Allodynia;
Primary and/or particular I.) survival; a potent phosphorylation in Secondary hyperalgesia; Causalgia; construct.

survival factor for response to artemin Phantom limb pain;
Post-surgical pain; -1 H
midbrain dopamine treatment; pain Burning feet syndrome;
Guillain-Barre I.) neurons, perception activity can syndrome; Thalamic syndrome; Post-motorneurons, be measured using L5 stroke pain;
Vasculitic/ angiopathic pain;
noradrenergic spinal nerve ligation Idiopathic pain;
Pain associated with any neurons, as well as and partial sciatic nerve of the following:
Entrapment for sympathetic, ligation models of neuropathy, Nerve transection, Spinal n parasympathetic and neuropathic pain, as in cord injury, Scar formation, Alcoholic sensory neurons; Boucher et al Science neuropathy, Pellagra, Beriberi, Post-promotes ureteric (2000) 290:124-127 herpetic neuralgia, HIV/AIDS pain, t..) o o branching in kidney and in Boucher et al Vincristine neurotoxicity, Cisplatin .6.
development -development and (2001) Curr. Opin. neurotoxicity, Taxol neurotoxicity, o ,...) regulates Pharmacol. 1:66-72. Thallium neurotoxicity, Arsenic o o Table 1 t..) Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic =
o u, Protein:X Assay Protein:Z -a , o spermatogenesis; neurotoxicity, Radiation therapy, ,...) t..) ,z normalizes Diabetes, Malignancies, Multiple c, nociceptive sensory sclerosis, Fabry's disease, Tangier neuron physiology disease, or Amyloid.
following injury.
Artemin- Neurotrophic factor Activity on neurons can Pain; Neuropathic pain; Complex 3564, 3565. Splice variant isoform 3 that promotes be assayed by regional pain syndrome I; Reflex 4; SEQ ID
(neublastin; neuronal growth, measuring increases in sympathetic dystrophy; Trigeminal NO:Z for n enovin) differentiation, and Ret tyrosine neuralgia; Allodynia;
Primary and/or particular 0 I.) survival; a potent phosphorylation in Secondary hyperalgesia; Causalgia; construct.
, us, H
C) UJ
midbrain dopamine treatment; pain Burning feet syndrome;
Guillain-Barre I.) neurons, perception activity can syndrome; Thalamic syndrome; Post- 0 u-, motorneurons, be measured using L5 stroke pain;
Vasculitic/ angiopathic pain; 0 noradrenergic spinal nerve ligation Idiopathic pain; Pain associated with any H
IV
neurons, as well as and partial sciatic nerve of the following:
Entrapment for sympathetic, ligation models of neuropathy, Nerve transection, Spinal parasympathetic and neuropathic pain, as in cord injury, Scar formation, Alcoholic sensory neurons; Boucher et al Science neuropathy, Pellagra, Beriberi, Post-promotes ureteric (2000) 290:124-127 herpetic neuralgia, HIV/AIDS pain, branching in kidney and in Boucher et al Vincristine neurotoxicity, Cisplatin n development and (2001) Curr. Opin. neurotoxicity, Taxol neurotoxicity, regulates Pharmacol. 1:66-72. Thallium neurotoxicity, Arsenic cp t..) =
spermatogenesis; neurotoxicity, Radiation therapy, =
.6.
normalizes Diabetes, Malignancies, Multiple 7a3 =
nociceptive sensory sclerosis, Fabry's disease, Tangier ,...) c, ,z neuron physiology _disease, or Amyloid.

Table 1 o Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic t..) o Protein:X Assay Protein:Z
u, O-following injury.
o ,...) NT5 (NT-5; Neurotrophic factor Activity on pain Pain; Neuropathic pain; Complex 3566, 3567. See Table 2, t..) o o neurotrophin- that promotes perception can be regional pain syndrome I; Reflex SEQ ID NO:Z
5; neuronal growth, assayed by measuring sympathetic dystrophy; Trigeminal for particular neurotrophic differentiation, and substance P release in neuralgia; Allodynia;
Primary and/or construct.
factor 5; survival; may play a response to C-fiber Secondary hyperalgesia; Causalgia;
NT4/5; NTF5) role in neuropathic stimulation in an in Phantom limb pain;
Post-surgical pain;
pain, vitro spinal cord Burning feet syndrome;
Guillain-Barre n preparation, as in syndrome; Thalamic syndrome; Post-Malcangio et al Eur. J. stroke pain; Vasculitic/ angiopathic pain;
I.) u-, .4. Neurosci (2000) Idiopathic pain; Pain associated with any H
UJ
IV
1--, 12:139-144. of the following:
Entrapment H
UJ
neuropathy, Nerve transection, Spinal "

cord injury, Scar formation, Alcoholic neuropathy, Pellagra, Beriberi, Post-.

herpetic neuralgia, HIV/AIDS pain, H
I.) Vincristine neurotoxicity, Cisplatin neurotoxicity, Taxol neurotoxicity, Thallium neurotoxicity, Arsenic neurotoxicity, Radiation therapy, Diabetes, Malignancies, Multiple sclerosis, Fabry's disease, Tangier n ,-i disease, or Amyloid.
cp Human Involved in Chemokine activities Autoimmune disorders; Immunity; 3373, 3374, 3375. See Table 2, t..) o o chemokine inflammation, can be determined Vascular and Inflammatory disorders; SEQ ID .6.
-o-HCC-1 allergy, tissue using assays known in HIV; AIDS;
infectious diseases. NO:Z for ' ,...) (ckBeta-1; rejection, viral the art: Methods in particular o o Table 1 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic t..) o Protein:X Assay Protein:Z o u, HWFBD) infection, and Molecular Biology, construct. O-o ,...) tumor biology; 2000, vol. 138:
t..) o o enhances Chemokine Protocols.
proliferation of Edited by: A.E.I.
CD34+ myeloid Proudfoot, T.N.C.
progenitor cells. Wells, and C.A.
Power. Humana Press Inc., Totowa, NJ
n Green The green fluorescent Cells and tissues (with Flourescent tag for gene expression 3409. See Table 2, Fluorescent protein (GFP) is the exception of detection.
SEQ ID NO:Z "
Ul H
I.) H
t\-) EGFP; red- green from transgenic mice construct. us, shifted GFP) bioluminescence of expressing this gene are I.) the jellyfish green under excitation Aequorea victoria. light.

Glucagon- Stimulates the GLP1 activity may be Hyperglycemia;
Diabetes; Diabetes 3430, 3438, 3446, See Table 2, H
I.) Like-Peptide 1 synthesis and release assayed in vitro using a Insipidus;
Diabetes mellitus; Type 1 3447, 3458, 3459, SEQ ID NO:Z
(GLP1; GLP- of insulin; enhances [3-1-1]-glucose uptake diabetes; Type 2 diabetes; Insulin 3460, 3461, 3462, for particular 1; the sensitivity of assay. (J Biol Chem resistance; Insulin deficiency; 3479 3480, 3481, construct.
Insulinotropin) adipose, muscle, and 1999 Oct 22; Hyperlipidemia;
Hyperketonemia; Non- 3482, 3493, 3494, liver tissues towards 274(43):30864-30873). insulin dependent Diabetes Mellitus 3495.
n insulin; stimulates GLP-1 effects on (NIDDM); Insulin-dependent Diabetes glucose uptake; slows learning can be Mellitus (IDDM); A
Condition cp t..) the digestive process; investigated using the Associated With Diabetes Including, But =
o suppresses appetite; possive avoidance and Not Limited To Obesity, Heart Disease, .6.
O-o blocks the secretion Morris water maze Hyperglycemia, Infections, Retinopathy, .
,...) of glucagon. (MWM) paradigms in And/Or Ulcers; Metabolic Disorders; o o Table 1 o Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic t..) o Protein:X AssayProtein:Z
=
u, rats (Brain Res. 1996, Immune Disorders; Obesity; Vascular O-o ,...) 716:29-38 and Nature Disorders; Suppression of Body Weight;
t,.) o 1982, 297:681-683). Suppression of Appetite;
Syndrome X; o Cognitive Impairment; Memory Loss.
Use to enhance learning, memory, , associative learning, spatial learning, hippocampal plasticity, cognition, and/or neuroprotection.
n (Octreotide; hormone, glucagons hormone release in Vasoactive Intestinal Peptide secreting SEQ ID NO:Z 0 I.) u-, .4, octreotide and insulin; humans by adenomas; Diarrhea and Flushing; for particular H
UJ
t.A.) I.) acetate; Suppresses LF somatostatin can be Prostatic disorders and cancers; Breast construct. H
UJ
Sandostating response to GnRH; measured as described cancer; Gastrointestinal disorders and I.) LARCI) Decreases splanchnic in J. Clin. Endocrinol. cancers; Cancers of the endocrine 0 u-, blood flow; Inhibits Metab. (1973) Oct; system; Head and neck paragangliomas; 0 I
release of serotonin, 37(4):632-4. Liver disorders and cancers; H
I.) gastrin, vasoactive Inhibition of insulin Nasopharyngeal cancers; Thyroid intestinal peptide, secretion by disorders and cancers;
Acromegaly;
secretin, motilin, and somatostatin can be Carcinoid Syndrome;
Gallbladder pancreatic measured as described disorders, such as gallbladder polypeptide. in the Lancet (1973) contractility diseases and abnormal bile ,-o Dec. 8; 2(7841):1299- secretion; Psoriasis; Diabetes; Diabetes n ,-i 1301. Insipidus; Diabetes mellitus; Type 1 cp diabetes; Type 2 diabetes; Insulin t,.) o o resistance; Insulin deficiency;
O-Hyperlipidemia; Hyperketonemia; Non-=
,...) insulin dependent Diabetes Mellitus o o Table 1 o Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic t..) o Protein:X Assay Protein:Z
u, O-(NIDDM); Insulin-dependent Diabetes o ,...) Mellitus (IDDM); A Condition t..) ,z o, Associated With Diabetes Including, But Not Limited To Obesity, Heart Disease, Hyperglycemia, Infections, Retinopathy, And/Or Ulcers; Metabolic Disorders;
Immune Disorders; Obesity; Vascular Disorders; Suppression of Body Weight;
n Suppression of Appetite; Syndrome X;

Kidney disorders; Neurological disorders "
Ul H
and diseases, including Alzheimers us, -p.
I.) Disease, Parkinson's disease and us, dementia; Neuropsychotic disorders, "

including Bipolar affective disorder;

Rheumatoid arthritis; Hypertension;

Intracranial hypertension; Esophageal H
I.) varices; Graves' disease; Seizures;
Epilepsy; Gastritis; Angiogenesis;
PYY (Peptide Decreases appetite; Appetite and food Most preferred:
Treatment of Obesity; 3510, 3515. See Table 2, YY), including increases satiety; intake can be can be treatment of Diabetes; suppression of SEQ ID NO:Z
PYY3-36 decreases food measured by methods body weight gain;
suppression of for particular (amino acid intake. known in the art appetite.
construct. n ,-i residues 31-64 (Batterham et al. Hyperglycemia;
Diabetes; Diabetes cp of full length Nature 2002; Insipidus; Diabetes mellitus; Type 1 t..) o o PYY, amino 418:650654) diabetes; Type 2 diabetes; Insulin .6.
acid residues residues resistance; Insulin deficiency; o ,...) 3-36 of mature Hyperlipidemia;
Hyperketonemia; Non- o, ,z Table 1 o Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic t..) o Protein:X Assay _Protein:Z o u, PYY) insulin dependent Diabetes Mellitus O-o ,...) (NIDDM); Insulin-dependent Diabetes t..) o o Mellitus (IDDM); A Condition Associated With Diabetes Including, But Not Limited To Obesity, Heart Disease, Hyperglycemia, Infections, Retinopathy, And/Or Ulcers; Metabolic Disorders;
Immune Disorders; Obesity; Vascular n Disorders; Suppression of Body Weight;

Suppression of Appetite; Syndrome X.
"
Ul H
Other indications for antibodies, us, .4.
I.) (II
H
antagonists: treatment of weight loss;
us, treatment of AIDS wasting; appetite "

stimulant; treatment of cachexia.

Interferon Confers a range of Anti-viral assay:
Viral infections include Severe Acute 2875, 2872, 2876, See Table 2, 0 Hybrids, cellular responses Rubinstein S, Familletti Respiratory Syndrome (SARS) and other 2874, 2873. SEQ ID NO:Z H
I.) specifically including antiviral, PC, Pestka S. (1981) coronavirus infections; filoviruses, for particular preferred: antiproliferative, Convenient assay for including but not limited to Ebola construct.
antitumor and interferons. J. Virol. viruses and Marburg virus; Arenaviruses, IFNalpha A/D immunomodulatory 37(2):755-8; Anti- including but not limited to Pichende hybrid (BgIII activities; stimulate proliferation assay: virus, Lassa virus, Junin virus, Machupo version) production of two Gao Y, et al (1999) virus, Guanarito virus; and lymphocytic n ,-i IFNalpha A/D enzymes: a protein Sensitivity of an choriomeningitis virus (LCMV);
cp hybrid (PvuII kinase and an epstein-barr virus- Bunyaviruses, including but not limited t..) o o version) oligoadenylate positive tumor line, to Punta toro virus, Crimean-Congo .6.
O-IFNalpha A/F synthetase. Also, Daudi, to alpha hemorrhagic fever virus, sandfly fever o ,...) ,hybrid modulates MHC interferon correlates viruses, Rift Valley fever virus, La o o Table 1 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic Protein:X Assay Protein:Z
IFNalpha A/B antigen expression, with expression of a Crosse virus, and hantaviruses;
hybrid NK cell activity and GC-rich viral Flaviviruses, including but not limited to IFNbeta IFNg production and transcript. Mol Cell Yellow Fever, Banzi virus, West Nile 1/alpha D IL12 production in Biol. 19(11):7305-13. virus, Dengue viruses, Japanese hybrid monocytes. Encephalitis virus, Tick-borne (IFNbeta- encephalitis, Omsk Hemorrhagic Fever, 1/alpha-1 and Kyasanur Forest Disease virus;
hybrid) Togaviruses, including but not limited to IFNalpha/beta Venezuelan, eastern, and western equine hybrid encephalitis viruses, Ross River virus, and Rubella virus; Orthopox viruses, cr, including but not limited to Vaccinia, UJ
Cowpox, Smallpox, and Monkeypox;

Herpesviruses; FluA/B; Respiratory Sincytial virus (RSV); paraflu; measles;
rhinoviruses; adenoviruses; Semliki Forest virus; Viral Hemorrhagic fevers;
Rhabdoviruses; Paramyxoviruses, including but not limited to Nipah virus and Hendra virus; and other viral agents identified by the U.S. Centers for Disease Control and Prevention as high-priority disease agents (i.e., Category A, B, and C agents; see, e.g., Moran, Emerg. Med. Clin. North. Am. 2002;
20(2):311-30 and Darling etal., Emerg.
Med. Clin. North Am. 2002;20(2):273-Table 1 , Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic o Protein:X Assay Protein:Z
u, 309).
O-o ,...) t..) o IL-2 Promotes the growth T cell proliferation Cancer; Solid Tumors; Pancreatic 1757, 1758, 1812, See Table 2, (Aldesleukin; of B and T cells and assay "Biological Cancer; Colon Cancer;
Liver Cancer; T- 1813, 1952, 1954, SEQ ID NO:Z
interleukin-2 augments NK cell activity of recombinant lymphomas; Graft-versus-host disease 2030, and 2031. for particular fusion toxin; T and CTL cell killing human interleukin-2 (GVH); Autoimmune diseases; construct.
cell growth activity, produced in Autoimmune disorders; IL-2 receptor factor; Escherichia coli." positive malignancies.
n PROLEUKIN; Science 223: 1412-IMMUNACE; 1415, 1984. natural "
u-, -o. CELEUK; killer (NK) cell and H
UJ
IV
-4 ONCOLIPIN CTL cytotoxicity assay H
us, 2; "Control of "

MACROLIN) homeostasis of CD8+

u-, memory T cells by opposing cytokines.
H
I.) Science 288: 675-678, 2000; CTLL-2 Proliferation : Gillis et al (1978) J. Immunol.
120, 2027 Salusin-a Causes rapid, Blood pressure can be Impaired cardiac output and/or 3702, 3703. See Table 2, n ,-i profound measured with a hypertension;
cardiovascular disorders, SEQ ID NO:Z
cp hypotension and sphygmomanometer or including but not limited to stroke, for particular t..) o o bradycardia; using other methods congestive heart failure, myocardial construct. .6.
increases intracellular intracellular that are well known in infarction.
,...) calcium; induces the art, such as in o o Table 1 o Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic t..) o Protein:X Assay Protein:Z
, , o expression of growth- Reddy et al., o ,...) associated genes; Ultrasound Med Biol k...) ,z c., stimulates 2003 Mar;29(3):379-proliferation of 85; cardiac cell vascular smooth proliferation can be muscle cells and assessed using methods fibroblasts. known in the art, for I
=
example, the n cardiogenesis assay as described in Eisenberg I.) u-, -1. et al., Dev Dyn 1999 H
UJ
IV
00 Sep;216(1):45-58.
H
us, Salusin-a(29G) Causes rapid, Blood pressure can be Impaired cardiac output and/or 3704, 3705. See Table 2, "

profound measured with a hypertension;
cardiovascular disorders, SEQ ID NO:Z 0 u-, hypotension and sphygmomanometer or including but not limited to stroke, for particular 0 bradycardia; using other methods congestive heart failure, myocardial construct. H
I.) increases intracellular that are well known in infarction.
calcium; induces the art, such as in expression of growth- Reddy et al., associated genes; Ultrasound Med Biol stimulates 2003 Mar;29(3):379-proliferation of 85; cardiac cell n ,-i vascular smooth proliferation can be cp muscle cells and assessed using methods t..) o o fibroblasts. known in the art, for .6.
example, the the ,...) cardiogenesis assay as ,z Table 1 Therapeutic Biological Activity Exemplary Activity Preferred Indication:Y
Construct ID Therapeutic Protein:X Assay Protein:Z
7a3 described in Eisenberg etal., Dev Dyn 1999 Sep;216(1):45-58.
Salusin-I3 Causes rapid, Blood pressure can be Impaired cardiac output and/or 3706, 3707. See Table 2, profound measured with a hypertension;
cardiovascular disorders, SEQ ID NO:Z
hypotension and sphygmomanometer or including but not limited to stroke, for particular bradycardia; using other methods congestive heart failure, myocardial construct.
increases intracellular that are well known in infarction.
calcium; induces the art, such as in expression of growth- Reddy et al., associated genes; Ultrasound Med Biol us, stimulates 2003 Mar;29(3):379-us, proliferation of 85; cardiac cell vascular smooth proliferation can be muscle cells and assessed using methods fibroblasts; known in the art, for stimulates release of example, the arginine-vasopressin cardiogenesis assay as from pituitary, may described in Eisenberg regulate water et al., Dev Dyn 1999 homeostasis Sep;216(1):45-58.

Table 2 Fusion Construct Construct Name Description Expressio SEQ SEQ SEQ SEQ SEQ Leader No. M n Vector ID ID ID ID ID Sequence a NO:Y NO: NO:Z NO: NO:B O-o X
A
t..) o 1 3373 pSAC:INV.CKB1.S26- S26-N93 of CKB1 linked to the N-pSAC35 198 111 285 372 373 Invertase c:
N93.DAHICHSA terminus of HSA through a 16aa linker peptide -- derived from the N-terminus of HSA (D25-E40), Invertase signal peptide, in yeast expression vector 2 3374 pSAC:CKB1.S26- S26-N93 of CKB1 linked to the N-pSAC35 199 112 286 374 375 HSA/kex2 N93.DAHK.HSA terminus of HSA through a 16aa linker n peptide -- derived from the N-terminus of HSA (D25-E40), HSA/kex2 signal peptide, I.) in in yeast expression vector H
CA
CA
cp 3 3375 pSACINV.HA.CKB1.G G28-N93 of CKB1 fused to the N-terminus pSAC35 200 113 287 376 377 Invertase I.) H
u.) 28-N93.HSA of HSA, with Invertase signal peptide and a I.) 2-aa linker (HA) between INV and CKB1, in in yeast expression vector 4 3404 pSAC35:BNP(2x)/HSA BNP tandem repeat (two copies of BNP) pSAC35 201 114 288 378 379 HSA/kex2 H
fused to the N-terminus of mature HSA
I.) 3409 pSAC35:HSA.GFP GFP (a red-shifted form known as EGFP) pSAC35 202 115 289 380 381 HSA/kex2 i with an N-terminal HSA fusion.
6 3422 pSAC35:APsp.HSA.IFN Acid Phosphatase signal peptide followed pSAC35 203 116 290 382 383 Acid a by mature RSA and IFNa.
phoshpatase 7 3423 pSAC35:INVsp.HSA.IF Invertase signal peptid followed by mature pSAC35 204 117 291 384 385 Invertase .0 Na HSA and IFNa.
Na mature HSA and IFNa.
cp t.) o 9 3430 pSAC35:KT.GLP-1(7- Killer Toxin signal peptide followed by pSAC35 206 119 293 Killer toxin c' .6.
36(A8G))x2.HSA(34A) tandem copies of amino acids 7-36 of GLP-'a o 1(A8G)followed by HSA(34A).


w o 3437 pSAC35:INV.Somatosta Invertase signal peptide followed by 207 120 294 Invertase ' Table 2 Fusion Construct Construct Name Description Expressio SEQ SEQ SEQ SEQ SEQ Leader g No. ID n Vector ID ID ID ID ID Sequence a) NO:Y NO: NO:Z NO: NO:B
o tin(S14).HSA(D25- Somatostatin(S14).HSA(D25-E40).HSA
t..) o E40).HSA
o 11 3438 pSAC35:KT.GLP-1(7- GLP-1(7-36(A8G)) is tandemly repeated as pSAC35 208 121 295 388 389 Killer toxin 36(A8G))x2.HSA.GFP a dimer and fused upstream from mature HSA with a C-terminal GFP tag and downstream from the killer toxin signal sequence. The GFP used here is a red-shifted form known as EGFP.
n -12 3446 pSAC.KT.GLP-1(7- Killer toxin signal peptide followed by a pSAC35 209 122 296 390 391 Killer toxin 0 36(A8G)).DAHK(25D- single copy of GLP-1 with a C-terminal "
in 30E).HSA fusion of HSA through a linker of first 6 aa H
CA
(J1 IV
. from the C-terminal of HSA.
H
u.) 13 3447 pSAC.KT.GLP-1(7- Killer toxin signal peptide followed by a pSAC35 210 123 297 392 393 Killer toxin I.) 36(A8G)).DAHK(25D- single copy of GLP-1 with a C-terminal in 38L).HSA fusion of HSA through a linker of first -,1 aa from the C-terminal of HSA.

H
14 3448 pC4:BNP/HSA HSA prepro followed by BNP followed by pC4 211 124 298 394 395 HSA I.) mature HSA.
(Mammalia n) 15 3458 pSAC.KT.GLP-1(7- Killer toxin signal peptide followed by a pSAC35 212 125 299 Killer toxin 36(A8G)).DAHK(25D- single copy of GLP-1 with a C-terminal 27H).HSA fusion of HSA through a linker of first 3 aa 1-d from the C-terminal of HSA.
n 1-i 16 3459 pSAC.KT.GLP-1(7- This construct contains a toxin signal pSAC35 213 126 300 Killer toxin cp 36(A8G)).DAHK(25D- peptide followed by single copy of GLP-1 t..) o 29S).HSA with a C-terminal fusion of HSA through a 4a 'a linker of first 5 aa from the C-terminal of o HSA.

c,.) o 17 3460 pSAC.KT.GLP-1(7- This construct contains a killer toxin signal pSAC35 214 127 301 Killer toxin '''' Table 2 Fusion Construct Construct Name Description Expressio SEQ SEQ SEQ SEQ SEQ Leader g No. ID n Vector ID ID ID ID ID Sequence õ
NO:Y NO: NO:Z NO: NO:B
X
A
36(A8G)).DAHK(25D- peptide followed by a single copy of GLP-28K).HSA 1 with a C-terminal fusion of HSA through a linker of first 4 aa from the C-terminal of HSA.
18 3461 pSAC.KT.GLP-1(7- This construct contains a killer toxin signal pSAC35 215 128 302 Killer toxin 36(A8G)).DAHK(25D- peptide followed by a single copy of GLP-33H).HSA 1 with a C-terminal fusion of HSA through a linker of first 9 aa from the C-terminal of HSA.

19 3462 pSAC.KT.GLP-1(7- This construct contains a killer toxin signal pSAC35 216 129 303 396 397 Killer toxin 36(A8G)).DAHK(25D- peptide followed by a single copy of GLP-37D).HSA 1 with a C-terminal fusion of HSA through a linker of first 13 aa from the C-terminal of HSA.

20 3477 pC4:BNP(2X)/HSA HSA prepro followed by tandem copies of pC4 BNP fused to N-terminal of mature HSA. (Mammalia n) 32A).HSA 1 with a C-terminal fusion of HSA through a linker of first 8 aa from the C-terminal of HSA.
22 3480 pSAC.KT.GLP-1(7- This construct contains a killer toxin signal pSAC35 219 132 306 Killer toxin n 36(A8G)).DAHK(25D- peptide followed by a single copy of GLP-34R).HSA 1 with a C-terminal fusion of HSA through a linker of first 10 aa from the C-terminal of HSA.
23 3481 pSAC.KT.GLP-1(7- This construct contains a killer toxin signal pSAC35 220 133 307 Killer toxin 36(A8G)).DAHK(25D- peptide followed by a single copy of GLP-Table 2 Fusion Construct Construct Name Description Expressio SEQ SEQ SEQ .SEQ SEQ Leader No. ID n Vector ID ID ID ID ID Sequence 2 NO:Y NO: NO:Z NO: NO:B
vi X
A 'a o 35F).HSA 1 with a C-terminal fusion of HSA
through c,.) t..) vD
a linker of first 11 aa from the C-terminal c7, of HSA.
24 3482 pSAC.KT.GLP-1(7- This construct contains a killer toxin signal pSAC35 221 134 308 Killer toxin 36(A8G)).DAHK(25D- peptide followed by a single copy of GLP-39G).HSA 1 with a C-terminal fusion of HSA
through a linker of first 15 aa from the C-terminal of HSA.
n 25 3484 pSAC35:ANP/HSA HSA/kex2 leader followed by atrial pSAC35 222 135 309 400 401 HSA/kex2 natriuretic peptide followed by mature I.) in HSA.
H

IA
UJ 26 3493 pSAC.KT.GLP-1(7- This construct contains a killer toxin signal pSAC35 223 136 310 Killer toxin "
H
IA
36(A8G)).DAHK(25D- peptide followed by a single copy of GLP-I.) 26A).HSA 1 with a C-terminal fusion of HSA
through 0 in a linker of first 2 aa from the C-terminal of HSA.
I
27 3494 pSAC.KT.GLP-1(7- Killer toxin signal peptide followed by a pSAC35 224 137 311 402 403 Killer toxin H
IV
36(A8G)).DAHK(25D- single copy of GLP-1 with a C-terminal 31V).HSA fusion of HSA through a linker of first 7 aa from the C-terminal of FISA.
36K).HSA fusion of HSA through a linker of first 12 1-d n aa from the C-terminal of HSA.
29 3510 pSAC35:INV.PYY3- PYY3-36 is fused to HSA via a 16aa HSA
pSAC35 226 139 313 406 407 Invertase r. , 36.HSA(D25-E40).HSA derived linker.
o o .6.
30 3513 pSAC35:BNP1- HSA/kex2 leader followed by a C-terminal pSAC35 227 140 314 408 409 HSA/kex2 o 29(2x)/HSA truncation version of BNP1-29 without the c,.) c7, terminal dibasic amino acid residues vD

Table 2 Fusion Construct Construct Name Description Expressio SEQ SEQ SEQ SEQ SEQ Leader No.

n Vector ID ID ID ID ID Sequence NO:Y NO: NO:Z NO: NO:B
- o o vi X
A .-....
= o o tandemly repeated twice, fused to N-i..) oo terminal of mature HSA.
31 3514 pSAC35INV:BNP1- Yeast invertase secretory signal peptide pSAC35 228 141 315 410 411 Invertase :
29/HSA followed by human BNP1-29/HSA fusion.
.
32 3515 pSAC35INV:PYY3- Yeast invertase signal peptide followed by pSAC35 229 142 316 Invertase 36/HSA PYY3-36 fused to N-terminus of mature i HSA.
.
33 3516 pEE12.1:BNP/HSA HSA prepro followed by BNP fused to N-pEE12.1 230 143 317 412 413 HSA .
-n terminus of mature HSA.

34 3517 pEE12.1:BNP2x/HSA HSA prepro followed by a tandem repeat pEE12.1 231 144 318 414 415 HSA I.) u-i (2x) of BNP fused to mature human serum H
CA
lil -P. albumin.
I.) H
u.) 35 3518 pSAC35:HSA/kex2.HS HSA/kex2 leader followed by mature HSA
pSAC35 232 145 319 HSA/kex2 I.) A.GLP-2 and GLP-2.

u-i ' 36 3519 pSAC35:HSA/kex2.GL HSA/kex2 leader followed by GLP-2 and 233 146 320 HSA/kex2 0 P-2.HSA mature HSA.
I
H
37 3524 pSAC35INV:BNP1- Invertase signal peptide followed by a C- pSAC35 234 147 321 416 417 Invertase "
26/HSA terminal truncation of BNP (aa 1-26)fused to mature HSA.
38 3525 pSAC35INV:BNP1- Invertase signal peptide followed by a C- pSAC35 235 148 322 418 419 Invertase 27/HSA terminal truncation of BNP (aa 1-27)fused to mature HSA.
_ Iv 39 3526 pSAC35INV:BNP1- invertase signal peptide followed by a C- pSAC35 236 149 323 420 421 Invertase n 1-i 28/HSA terminal truncation of BNP (aa 1-28)fused cp to mature HSA.
o 40 3535 pSAC35:HSA/kex2.HS HSA/kex2 leader followed by mature HSA
pSAC35 237 150 324 HSA/kex2 5;
A.GLP2ana and GLP2 analog ALX0600 O' o 41 3536 pSAC35:HSA/kex2.GL HSA/kex2 leader followed by GLP2 analog pSAC35 238 151 325 HSA/kex2 (...) o P2ana.HSA AXL0600 followed by mature FISA.
o ' Table 2 , Fusion Construct Construct Name Description Expressio SEQ SEQ SEQ SEQ SEQ Leader No. ID n Vector ID ID ID ID ID Sequence , õ
NO:Y NO: NO:Z NO: NO:B
o .o vi X
ik E 'a .o 42 3537 pSAC35:HSA/kex2.1-IS HSA/kex2 leader followed by mature HSA
pSAC35 239 152 326 HSA/kex2 !=,',' A.PACAP27 and PACAP27.
o o 43 3538 pSAC35:HSA/kex2.PA HSA/kex2 leader followed by PACAP27 pSAC35 240 153 327 HSA/kex2 CAP27.HSA followed by mature HSA.
44 3539 pSAC35:HSA/kex2.HS HSA/kex2 leader followed by mature HSA pSAC35 241 154 328 HSA/kex2 A.PACAP38 and PACAP38.
45 3540 pSAC35:HSA/kex2.PA HSA/kex2 leader followed by PACAP38 pSAC35 242 155 329 HSA/kex2 ' CAP38.HSA followed by mature HSA.
n 46 3541 pSAC35:HSA/kex2.HS HSA/kex2 leader followed by mature HSA phoAl 243 156 330 HSA/kex2 =

A.BDNFa followed by mature BDNF isoform a.
I.) in 47 3542 pSAC35:HSA/kex2.BD HSA/kex2 leader followed by mature pSAC35 244 157 331 HSA/kex2 H
CA
N
LA NFa.HSA BDNF isoform a (Genbank AF400438) H
CA
followed by mature HSA.
I.) _ 48 3543 pSAC35:HSA/kex2.HS HSA/kex2 leader followed by mature HSA pSAC35 245 158 332 HSA/kex2 0 in ' A.BDNFb followed by mature BDNF isoform b.

-,1 49 3544 pSAC35:HSA/kex2.BD HSA/kex2 leader followed by mature pSAC35 246 159 333 HSA/kex2 1 H
NFb.HSA BDNF isoform b followed by mature HSA.
"
50 3545 pSAC35:HSA/kex2.HS HSA/kex2 leader followed by mature HSA pSAC35 247 160 334 HSA/kex2 A.BDNFc and mature BDNF isoform c.
51 3550 pSAC35:HSA/kex2SS.B HSA/kex2 leader followed by mature pSAC35 248 161 335 HSA/kex2 DNFc.HSA BDNF isoform c (encoded by splice variant 6) followed by mature HSA.
1-d 52 3551 pSAC35:HSA/kex2.HS HSA/kex2 leader followed by mature HSA pSAC35 249 162 336 HSA/kex2 r) 1-i A.GDNF followed by mature GDNF.
cp 53 3552 pSAC35:HSA/kex2.GD HSAJkex2 leader followed by mature pSAC35 250 163 337 HSA/kex2 t-) o NF.HSA GDNF followed by mature HSA.
o .6.
54 3553 pSAC35:HSA/kex2.1-lS HSA/kex2 leader followed by mature HSA
pSAC35 251 164 338O' HSA/kex2 o 1¨

A.neurturin followed by mature neurturin.
c,.) o o 55 3554 pSAC35:HSA/kex2.neur HSA/kex2 leader followed by mature pSAC35 252 165 339 HSA/kex2 Table 2 Fusion Construct Construct Name Description Expressio SEQ SEQ SEQ SEQ SEQ Leader No. ID n Vector ID ID ID ID ID Sequence 2 NO:Y NO: NO:Z NO: NO:B
o o vi o turin.HSA neurturin followed by mature HSA.
c,.) t..) 56 3555 pSAC35:HSA/kex2.HS HSA/kex2 leader followed by mature HSA pSAC35 253 166 340 HSA/kex2 *
A.NT3 followed by mature NT3.
57 3556 pSAC35:HSA/kex2.NT HSA/kex2 leader followed by mature NT3 pSAC35 254 167 341 HSA/kex2 3.HSA followed by mature HSA.
58 3557 pSAC35:HSA/kex2.HS 1-ISA/kex2 leader followed by mature HSA
pSAC35 255 168 342 HSA/kex2 A.persephin followed by mature persephin.
.
59 3558 pSAC35:HSA/kex2.pers HSA/kex2 leader followed by mature pSAC35 256 169 343 HSA/kex2 n ephin.HSA persephin followed by mature HSA.

60 3559 pSAC35:HSA/kex2.HS HSA/kex2 leader followed by mature HSA pSAC35 257 170 344 HSA/kex2 I.) in A.artemin1 followed by mature artemin isoform 1.
H
t.J1 CA
N
' 61 3561 pSAC35:HSA/kex2SS.a HSA/kex2 leader followed by mature pSAC35 258 171 345 HSA/kex2 H
CA
rteminl.HSA artemin isoform 1 followed by mature I.) HSA.

in ' 62 3562 pSAC35:HSA/kex2.HS HSA/kex2 leader followed by mature HSA pSAC35 259 172 346 HSA/kex2 0 -,1 A.artemin2 followed by mature artemin isoform 2.

- H
63 3563 pSAC35:HSA/kex2.arte HSA/kex2 leader followed by mature pSAC35 260 173 347 HSA/kex2 "
min2.HSA artemin isoform 2 followed by mature HSA.
64 3564 pSAC35:HSA/kex.2.HS HSA/kex2 leader followed by mature HSA
pSAC35 261 174 348 HSA/kex2 A.artemin3 followed by mature artemin3.
65 3565 pSAC35:HSA/kex2.arte HSA/kex2 leader followed by mature pSAC35 262 175 349 HSA/kex2 Iv min3.HSA artemin isoform 3 followed by mature n 1-i HSA.
66 3566 pSAC35:HSA/kex2.HS HSA/kex2 leader followed by mature HSA pSAC35 263 176 350 HSA/kex2 r.) o A.NT5 followed by mature NT5.
.6.
67 3567 pSAC35:HSA/kex2.NT HSA/kex2 leader followed by mature NT5 pSAC35 264 177 351 HSA/kex2 5.HSA followed by mature HSA.
c,.) o 68 3568 pSAC35:HSA/kex2.HS HSA/kex2 leader followed by mature HSA pSAC35 265 178 352 _ HSA/kex2 '''' Table 2 :
Fusion Construct Construct Name Description Expressio SEQ SEQ SEQ SEQ SEQ Leader =

No. ID n Vector ID ID ID ID ID Sequence NO:Y NO: NO:Z NO: NO:B
o o vi X
A O' o A.VIP followed by mature VIP.
c,.) i..) o 69 3569 pSAC35:HSA/kex2.VIP HSA/kex2 leader followed by mature VIP
pSAC35 266 179 353 HSA/kex2 c:
.HSA followed by mature HSA.
70 3570 pSAC35:HSA/kex2.HS HSA/kex2 leader followed by mature HSA pSAC35 267 180 354 HSA/kex2 A.secretin followed by mature secretin.
71 3571 pSAC35:HSA/kex2.secr HSA/kex2 leader followed by mature pSAC35 268 181 355 HSA/kex2 etin.HSA secretin followed by mature HSA.
72 3572 pSAC35:HSA/kex2.1-IS HSA/kex2 leader followed by mature HSA
pSAC35 269 182 356 HSA/kex2 n A.NGF followed by mature NGF.
.
. 0 73 3573 pSAC35:HSA/kex2.NG HSA/kex2 leader followed by mature NGF pSAC35 270 183 357 HSA/kex2 I.) in LA
F.HSA followed by mature HSA.
I.) -4 74 3574 pSAC35:HSA/kex2.HS HSA/kex2 leader followed by mature HSA
pSAC35 271 184 358 HSA/kex2 H
CA
A.NGFB followed by mature NGFbeta.
I.) 75 3575 pSAC35:HSA/kex2.NG HSA/kex2 leader followed by mature pSAC35 272 185 359 HSA/kex2 0 in FB.HSA NGFbeta followed by mature HSA.
' -,1 76 3577 pSAC35:HSA/kex2.HS HSA/kex2 leader followed by mature HSA pSAC35 273 186 360 HSA/kex2 1 H
A.glicentin followed by mature glicentin.
"
77 3578 pSAC35:HSA/kex2.glic HSA/kex2 leader followed by mature pSAC35 274 187 361 HSA/kex2 entin.HSA glicentin followed by mature HSA.
78 3579 pSAC35:HSA/kex2.HS HSA/kex2 leader followed by mature HSA pSAC35 275 188 362 HSA/kex2 A.oxyntomodulin followed by mature oxyntomodulin.
79 3580 pSAC35:HSA/kex2.oxy HSA/kex2 leader followed by mature pSAC35 276 189 363 HSA/kex2 .0 ntomodulin.HSA oxyntomodulin followed by mature HSA.
n 1-i 80 3581 pSAC35:HSA/kex2.HS HSA/kex2 leader followed by mature HSA pSAC35 277 190 364 HSA/kex2 cp A.PHM followed by mature peptide histidine i..) o o methionine.
.6.
O' 81 3582 pSAC35:HSA/kex2.PH HSA/kex2 leader followed by mature PI-IM
pSAC35 278 191 365 HSA/kex2 o M.HSA followed by mature HSA.
c,.) o 82 3583 pSAC35:HSA/kex2.HS HSA/kex2 leader followed by mature HSA pSAC35 279 192 366 HSA/kex2 vD

Table 2 Fusion Construct Construct Name Description Expressio SEQ SEQ SEQ SEQ SEQ Leader .

No. ID n Vector ID ID ID ID ID Sequence õ
NO:Y NO: NO:Z NO: NO:B
o o vi X
A 'a o A.CD4M33 followed by CD4M33.
c,.) t..) o 83 3584 pSAC35:HSA/kex2.CD HSA/kex2 leader followed by CD4M33 pSAC35 280 193 367 HSA/kex2 c:
4M33.HSA followed by mature HSA.
84 3616 pSAC35:BNP26(2x)/HS HSA/kex2 leader followed by BNP1-26 x2 pSAC35 281 194 368 422 423 HSA/kex2 A fused to mature HSA.
85 3617 pSAC35:BNP1- HSA/kex2 leader followed by BNP1-28 pSAC35 282 195 369 424 425 HSA/kex2 28(2x)/HSA 2x fused to mature HSA.
86 3618 pC4:SPC0n.BNP32(2x)/ A consensus signal peptide (SEQ ID
pC4 283 196 370 426 427 consensus n HSA NO:550) followed by BNP 1-32 (x2) fused (Mammalia to mature HSA. n) I.) in 87 3619 pSAC35:BNP27(2x)/HS HSA/kex2 leader followed by BNP 1-27 pSAC35 284 197 371 428 429 HSA/kex2 H
CA
CA
IV
oo A (2x) fused to mature HSA.
H
CA
88 2249 pSAC35:IFNa2-HSA Mature IFNa2 fused upstream of mature pSAC35 430 431 432 433 434 HSA/kex2 I.) HSA and downstream of HSA/kex2 leader in I
also named: sequence.

pSAC23:IFNa2-HSA
I
H
89 2343 pSAC35.INV- Mature Interferon alpha2 fused upstream of pSAC35 435 436 437 438 439 invertase "
IFNA2.HSA mature HSA and downstream of invertase signal peptide.
90 2366 pSAC35.MAF- Mature IFNa2 fused upstream of mature PSAC35 440 441 442 443 444 MFa-1 IFNa2.HSA HSA and downstream of yeast mating factor alpha leader sequence.
1-d 91 2381 pC4:HSA-IFNa2(C17- Amino acids C17 to E181 of IFNa2 pC4 445 446 447 448 449 HSA n 1-i E181) (fragment shown as amino acids Cl to E165 of SEQ ID NO:618) fused cp k.) o downstream of HSA.
=
.6.
92 2382 pC4:IFNa2-HSA IFNa2 fused upstream of mature HSA.
pC4 450 451 452 453 454 Native o IFNa2 k-Z
o leader vp Table 2 :
Fusion Construct Construct Name Description Expressio SEQ SEQ SEQ SEQ SEQ Leader i No. ID n Vector ID ID ID ID ID Sequence NO:Y NO: NO:Z NO: NO:B
o o vi X
A O' o 93 2410 pSAC35INV:1FNa-FISA Mature IFNa2 fused downstream of the pSAC35 455 456 457 458 459 invertase ',.,=) o invertase signal peptide and upstream of o mature HSA.
94 3165 pSAC35:HSA.IFNa HSA fused upstream of IFNa and pSAC35 460 461 462 HSA/kex2 downstream of the HSA/kex2 leader.
also named CID 3165, pSAC35:HSA.INFa 95 1778 pSAC35:IFNbeta.M22- Residues M22-N187 of full-length IFNb pSAC35 463 464 465 466 467 HSA/kex2 n N187:HSA (shown as M1 to N166 of SEQ ID

NO:463) fused upstream of mature HSA
I.) in and downstream of HSA/kex2 leader H
LA
CA
v: sequence.
I.) H
CA
96 1779 pSAC35:HSA:IFNbeta. Residues M22-N187 of full-length IFNb pSAC35 468 469 470 HSA/kex2 I.) M22-N187 (shown as M1 to N166 of SEQ ID

in NO:464) fused downstream of HSA with HSA/kex2 leader sequence.
I
97 2011 pC4:IFNb-HSA Full length IFNb fused upstream of mature pC4 471 472 473 474 475 Native IFNb r) HSA.
leader 98 2013 pC4:HSA-IFNb.M22- Amino acids M22 to N187 of IFNb pC4 476 477 478 HSA
N187 (fragment shown as amino acids M1 to N166 of SEQ ID NO:527) fused downstream of HSA.
1-d 99 2053 pEE12:IFNb-HSA Full length IFNb fused upstream of mature pEE12.1 479 480 481 Native IFNb n 1-i HSA.
leader -----also named cp t..) pEE12.1:IFNbeta-HSA
o o .6.
100 2054 pEE12:HSA-IFNb Mature IFNb fused downstream of HSA.
pEE12.1 482 483 484 HSA -a-, =
c., ,., Table 2 Fusion Construct Construct Name Description Expressio SEQ SEQ SEQ SEQ SEQ Leader o No. ID n Vector ID ID ID ID ID Sequence a ) =
NO:Y NO: NO:Z NO: NO:B
u.
O-X
A =
_ c,..
101 2492 pC4.IFNb(deltaM22).H Mutant full length INFbeta fused upstream pC4 485 486 487 Native IFN(3 SA of mature HSA. First residue of native, leader mature IFNbeta (M22) has been deleted.
102 2580 pC4.1FNb(deltaM22,C3 IFNb fused upstream of mature HSA.
The pC4 488 489 490 Native IFI\11:1 8S).HSA IFNb used in this fusion lacks the first residue of the mature form of IFNb, which corresponds to M22 of SEQ ID NO:1687.
Also amino acid 38 of SEQ ID NO:1687 n has been mutated from Cys to Ser.0 103 2795 pC4:HSA(A14)- The mature form of IFNb is fused to the pC4 491 492 493 Modified Ul H
IFNb.M22-N187 C-terminus of HSA, which contains an HSA (A14) u.) I.) cr, H
0 modified signal peptide, designed to u.) improve processing and homogeneity. _ I.) _ _ 0 104 2796 pC4:HSA(S14)- The mature form of IFNb is fused to the pC4 494 495 496 Modified in IFNb.M22-N187 C-terminus of HSA, which contains a HSA (S14) 0 -,1 I
modified signal peptide, designed to H
IV
_improve processing and homogeneity.
105 2797 pC4:HSA(G14)- The mature form of IFNb is fused to the pC4 497 498 499 Modified IFNb.M22-N187 C-terminus of HSA, which contains an HSA (G14) modified signal peptide.
_ 106 2872 pSAC35:HSA.IFNaA(C This construct contains a hybrid form of pSAC35 500 501 502 503 504 HSA/kex2 1-Q91)/ D(L93-E166) IFNaA and IFNaD fused downstream of 1-d n mature HSA.
107 2873 pSAC35:HSA.IFNaA(C This construct contains a hybrid form of pSAC35 505 506 507 508 509 HSA/kex2 c7, 1-Q91)/ B(L93-E166) IFNaA and IFNaB fused downstream of t..) o o mature HSA.
.6.
'a 108 2874 pSAC35:HSA.IFNaA(C This construct contains a hybrid form of pSAC35 510 511 512 513 514 HSA/kex2 ,O
1-Q91)/ F(L93-E166) IFNaA and IFNaF fused downstream of c,.) o o mature HSA.

Table 2 Fusion Construct Construct Name Description Expressio SEQ SEQ SEQ SEQ SEQ Leader g No. ID n Vector ID ID ID ID ID Sequence NO:Y NO: NO:Z NO: NO:B
X
A

pSAC35:HSA.IFNaA(C This construct contains a hybrid form of pSAC35 515 516 517 518 519 HSA/kex2 1Q-62)/D(Q64-E166) IFNaA and IFNaD fused downstream of mature FISA.

pSAC35:HSA.IFNaA(C This construct contains a hybrid form of pSAC35 520 521 522 523 524 HSA/kex2 1-Q91)/ D(L93-E166); IFNaA and IFNaD fused downstream of R23K,A113V mature HSA.
111 1757 pSAC35:IL2.A21- Mature human IL-2 with a single amino pSAC35 525 526 527 HSAJkex2 T153.145C/S.HSA acid mutation (C to S at position 145) cloned downstream of the HSA/KEX2 leader and upstream of mature HSA

pSAC35:HSA.1L2.A21- Mature human 1L-2 with a single amino pSAC35 528 529 530 HSA/kex2 T153.145C/S acid mutation (C to S at position 145) cloned downstream of HSA with HSA/kex2 leader sequence.

113 1812 pSAC35:1L2.A21- Amino acids A21 to T153 of IL-2 fused pSAC35. 531 532 533 HSA/kex2 0 T153.HSA downstream of the HSA/kex2 leader and upstream of mature HSA.
T153 downstream of HSA with HSA/kex2 leader sequence.
to Serine mutation at amino acid 145, fused leader 1-d upstream of mature HSA.
to Serine mutation at amino acid 145, fused leader c6 upstream of mature HSA.

Table 2 Fusion Construct Construct Name Description Expressio SEQ SEQ SEQ SEQ SEQ Leader No. ID n Vector ID ID ID ID ID Sequence NO:Y NO: NO:Z NO: NO:B
X
A
117 2030 pSAC35.ycolL-2.A21- Amino acids A21 to T153 of IL-2 fused pSAC35 543 544 545 HSA/kex2 T153.HSA upstream of mature HSA and downstream of HSA/kex2 leader sequence. DNA
encoding IL-2 has been codon optimized.
118 2031 pSAC35.HSA.ycolL- Amino acids A21 to T153 of IL-2 fused pSAC35 546 547 548 HSA/kex2 2.A21-T153 downstream of HSA with the HSA/kex2 leader sequence. DNA encoding IL-2 has been codon optimized.
119 3702 pSAC35:HSA/kex2.HS HSA/kex2 leader fused to N-terminus of pSAC35 557 551 563 HSA/kex2 A.salusin-a mature HSA followed by salusin alpha.

120 3703 pSAC35:HSA/kex2.salu HSAJkex2 leader fused to N-terminus of pSAC35 558 552 564 HSA/kex2¨
a, 1µ..) s i n-a.H S A salusin alpha followed by mature FISA.
121 3704 pSAC35:HSA/kex2.HS HSA/kex2 leader fused to N-terminus of pSAC35 559 553 565 HSA/kex2¨

A.salusin-a(29G) mature HSA followed by salusin alpha (29G).

122 3705 pSAC35:HSA/kex2.salu HSA/kex2 leader fused to N-terminus of pSAC35 560 554 566 HSA/kex2 sin-a(29G).HSA salusin-a(29G) followed by mature HSA.
123 3706 pSAC35:HSA/kex2.HS HSA/kex2 leader fused to the N-terminus pSAC35 561 555 567 HSA/kex2 A.salusin-B of mature HSA followed by salusin beta.
124 3707 pSAC35:HSA/kex2.salu HSA/kex2 leader fused to N-terminus of pSAC35 562 556 568 HSA/kex2 sin-B.HSA salusin beta followed by mature HSA.
1-d [0050] 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.
[0051] The fourth column in Table 2, "Description" provides a general description of a given albumin fusion construct, and the fifth column, "Expression Vector"
lists the vector into which a polynucleotide comprising, or alternatively consisting of, a nucleic acid molecule encoding a given albumin fusion protein was cloned. Vectors are known in the art, and are available commercially or described elsewhere. For example, as described in the Examples, 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. In one embodiment, for expression in S. cervisiae, an expression cassette comprising, or alternatively consisting of, a nucleic acid molecule encoding an albumin fusion protein is cloned into pSAC35. In another embodiment, for expression in CHO cells, an expression cassette comprising, or alternatively consisting of, a nucleic acid molecule encoding an albumin fusion protein is cloned into pC4. In a further embodiment, 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. In a still further embodiment, for expression in NSO cells, 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.
[0052] Column 6, "SEQ ID NO:Y," provides the full length amino acid sequence of the albumin fusion protein of the invention. In most instances, SEQ ID NO: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 specific amino acid sequence of the listed signal sequence is shown later in the specification or is well known in the art. Thus, most preferred embodiments of the present invention include the albumin fusion protein produced by a cell (which would lack the leader sequence shown in columns 4 and/or 11 of Table 2). Also most preferred are polypeptides comprising SEQ ID
NO:Y without the specific leader sequence listed in columns 4 and/or 11 of Table 2.
Compositions comprising these two preferred embodiments, including pharmaceutical compositions, are also preferred. Moreover, it is well within the ability of the skilled artisan to replace the signal sequence listed in columns 4 and/or 11 of Table 2 with a different signal sequence, such as those described later in the specification to facilitate secretion of the processed albumin fusion protein.
[0053] The seventh column, "SEQ ID NO:X," provides the parent nucleic acid sequence from which a polynucleotide encoding a Therapeutic protein portion of a given albumin fusion protein may be derived. In one embodiment, 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. In an alternative embodiment, 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.
[0054] The eighth column, "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. Moreover, it is well within the ability of the skilled artisan to use the sequence shown as SEQ ID NO:Z to derive the =õõ.
construct described in the same row. For example, if 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.
[0055] 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. In one embodiment of the invention, 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 ID 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.
[0056] In an alternative embodiment, oligonucleotide primers may be used in overlapping PCR reactions to generate mutations within a template DNA
sequence. PCR
methods are known in the art.
[0057] As shown in Table 3, certain albumin fusion constructs disclosed in this application have been deposited with the ATCCO.

õ.õ
Table 3 Construct ID Construct Name ATCC Deposit No./ Date 2053 I pEE12:IFNb-HSA PTA-3764 Oct. 4, 2001 also named pEE12.1:IFNP-HSA
2054 pEE12:HSA-IFNb PTA-3941 Dec. 19, 2001 2249 pSAC35:IFNa2-HSA PTA-3763 Oct. 4, 2001 also named pSAC23:IFNa2-HSA
2343 pSAC35.INV-IFNA2.HSA PTA-3940 Dec. 19, 2001 2381 pC4:HSA-IFNa2(C17-E181) PTA-3942 Dec. 19, 2001 2382 pC4:IFNa2-HSA PTA-3939 Dec. 19, 2001 2492 pC4.IFNb(deltaM22).HSA PTA-3943 Dec. 19, 2001 3165 pSAC35:HSA.IFNa PTA-4670 Sept. 16, 2002 also named CID 3165, pSAC35:HSA.INFa 3070 pSAC35:KT.GLP-1(7-36(A8G))x2.HSA PTA-4671 Sept. 16, 2002 [0058] It is possible to retrieve a given albumin fusion construct from the deposit by techniques known in the art and described elsewhere herein (see, Example 10).
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.
[0059] In a further embodiment of the invention, 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.
[0060] In preferred embodiments, 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. In further preferred embodiments, 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.
Polypeptide and Polynucleotide Fragments and Variants Fragments [0061] 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.
[0062] 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.
[0063] Even if deletion of one or more amino acids from the N-terminus of a protein results in modification or loss of one or more biological functions of the Therapeutic protein, albumin protein, and/or albumin fusion protein of the invention, other Therapeutic activities and/or functional activities (e.g., biological activities, ability to multimerize, ability to bind a ligand) may still be retained. For example, the ability of polypeptides with N-terminal deletions to induce and/or bind to antibodies which recognize the complete or mature forms of the polypeptides generally will be retained when less than the majority of the residues of the complete polypeptide are removed from the N-terminus. Whether a particular polypeptide lacking N-terminal residues of a complete polypeptide retains such immunologic activities can readily be determined by routine methods described herein and otherwise known in the art. It is not unlikely that a mutein with a large number of deleted N-terminal amino acid residues may retain some biological or immunogenic activities. In fact, peptides composed of as few as six amino acid residues may often evoke an immune response.
[0064] Accordingly, 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). In particular, 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.
[0065] In addition, 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). In preferred embodiments, 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:1), and m is defined as any integer ranging from 2 to 579. Polynucleotides encoding these polypeptides are also encompassed by the invention. In additional embodiments, N-terminal deletions may be described by the general formula m to 609, where 609 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.
[0066] Moreover, 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). In particular, 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.
[0067] Also as mentioned above, even if deletion of one or more amino acids from the N-terminus or C-terminus of a reference polypeptide (e.g., a Therapeutic protein; serum albumin protein; or albumin fusion protein of the invention) results in modification or loss of one or more biological functions of the protein, other functional activities (e.g., biological activities, ability to multimerize, ability to bind a ligand) and/or Therapeutic activities may still be retained. For example 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.
[0068] 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 2). In 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 a reference polypeptide (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 2). Polynucleotides encoding these polypeptides are also encompassed by the invention.
[0069] In addition, 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). In particular, 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. In 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.
[0070] Moreover, the present invention provides polypeptides having one or more residues deleted from the carboxy terminus of an albumin fusion protein of the invention. In 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.
[0071] In addition, 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 are integers as described above. Polynucleotides encoding these polypeptides are also encompassed by the invention.
[0072] 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. In preferred embodiments, the application is directed to proteins comprising polypeptides at least 80%, 85%, 90%, 95%, 96%, 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.
[0073] 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.
[0074] 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.
Variants [0075] "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.
[0076] As used herein, "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. see "therapeutic" column of Table 1), albumin protein, and/or albumin fusion protein, respectively, but retaining at least one functional and/or therapeutic property thereof as described elsewhere herein or otherwise known in the art. Generally, 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.
[0077] 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 protein:X 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 thereof), 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/or albumin fusion proteins.
Fragments of these polypeptides are also provided (e.g., those fragments described herein).
Further 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.
etal., eds., 1989 Current protocol in Molecular Biology, Green publishing associates, Inc., and John Wiley &
Sons Inc., New York, at pages 6.3.1 - 6.3.6 and 2.10.3). Polynucleotides encoding these polypeptides are also encompassed by the invention.
[0078] By 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. In other words, to obtain a polypeptide having an amino acid sequence at least 95% identical to a 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.
[0079] As a practical matter, whether 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 õ
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.
(Comp. App. Biosci.6:237-245 (1990)). In a sequence alignment the query and subject sequences are either both nucleotide sequences or both amino acid sequences.
The result of said global sequence alignment is expressed as percent identity. Preferred parameters used in a FASTDB amino acid alignment are: Matrix=PAM 0, k-tuple=2, Mismatch Penalty=1, Joining Penalty=20, Randomization Group Length=0, Cutoff Score=1, Window Size=sequence length, Gap Penalty=5, Gap Size Penalty=0.05, Window Size=500 or the length of the subject amino acid sequence, whichever is shorter.
[0080] If the subject sequence is shorter than the query sequence due to N- or C-terminal deletions, not because of internal deletions, a manual correction must be made to the results. This is because the FASTDB program does not account for N- and C-terminal truncations of the subject sequence when calculating global percent identity.
For subject sequences truncated at the N- and C-termini, relative to the query sequence, 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.
[0081] For example, a 90 amino acid residue subject sequence is aligned with a 100 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%. In another example, a 90 residue subject sequence is compared with a 100 residue query sequence. This time the 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. In this case the percent identity calculated by FASTDB is not manually corrected. Once again, only 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.
[00821 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, blasbc, tblastn and tblastx (Karlin et al., Proc. Natl. Acad. Sci. USA 87: 2264-2268 (1990) and Altschul, J. Mol.
Evol. 36:
290-300 (1993)) which are tailored for sequence similarity searching.
[00831 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. For a discussion of basic issues in similarity searching of sequence databases, see Altschul et al., (Nature Genetics 6: 119-129 (1994)). 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. Natl. Acad. Sci. USA 89: 10915-10919 (1992)).
For blastn, 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. Four blastn parameters may be adjusted as follows: Q=10 (gap creation penalty); R=10 (gap extension penalty); wink=1 (generates word hits at every winkth position along the query); and gapvv=16 (sets the window width within which gapped alignments are generated). The equivalent Blastp parameter settings were Q=9; R=2; wink=1; and gapw=32. A Bestfit comparison between sequences, available in the GCG package version 10.0, uses DNA parameters GAP=50 (gap creation penalty) and LEN=3 (gap extension penalty) and the equivalent settings in protein comparisons are GAP=8 and LEN=2.
[0084] The polynucleotide variants of the invention may contain alterations in the coding regions, non-coding regions, or both. Especially preferred are polynucleotide variants containing alterations which produce silent substitutions, additions, or deletions, but do not alter the properties or activities of the encoded polypeptide. Nucleotide variants produced by silent substitutions due to the degeneracy of the genetic code are preferred.
Moreover, 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).
[0085] In a preferred embodiment, a polynucleotide of the invention which encodes the albumin portion of an albumin fusion protein is optimized for expression in yeast or mammalian cells. In a further preferred embodiment, a polynucleotide of the invention which encodes the Therapeutic protein portion of an albumin fusion protein is optimized for expression in yeast or mammalian cells. In a still further preferred embodiment, a polynucleotide encoding an albumin fusion protein of the invention is optimized for expression in yeast or mammalian cells.
[0086] In an alternative embodiment, 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. In a further embodiment, 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. In another embodiment, 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.
[0087] In an additional embodiment, 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. In a further embodiment, 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.
In an alternative embodiment, 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.
[0088] 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 II, 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.
[0089] Using known methods of protein engineering and recombinant DNA
technology, variants may be generated to improve or alter the characteristics of the polypeptides of the present invention. For instance, 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. As an example, Ron et al. (J. Biol.
Chem. 268: 2984-2988 (1993)) reported variant KGF proteins having heparin binding activity even after deleting 3, 8, or 27 amino-terminal amino acid residues. Similarly, Interferon gamma exhibited up to ten times higher activity after deleting 8-10 amino acid residues from the carboxy terminus of this protein. (Dobeli et al., J. Biotechnology 7:199-216 (1988).) [0090] Moreover, ample evidence demonstrates that variants often retain a biological activity similar to that of the naturally occurring protein. For example, Gayle and coworkers (J. Biol. Chem. 268:22105-22111(1993)) conducted extensive mutational analysis of human cytokine IL-la. They used random mutagenesis to generate over 3,500 individual IL-la mutants that averaged 2.5 amino acid changes per variant over the entire length of the molecule. Multiple mutations were examined at every possible amino acid position. The investigators found that "[m]ost of the molecule could be altered with little effect on either [binding or biological activity]." In fact, only 23 unique amino acid sequences, out of more than 3,500 nucleotide sequences examined, produced a protein that significantly differed in activity from wild-type.
[0091] Furthermore, even if deleting one or more amino acids from the N-terminus or C-terminus of a polypeptide results in modification or loss of one or more biological functions, other biological activities may still be retained. For example, the ability of a deletion variant to induce and/or to bind antibodies which recognize the secreted form will likely be retained when less than the majority of the residues of the secreted form are removed from the N-terminus or C-terminus. Whether a particular polypeptide lacking N- or C-terminal residues of a protein retains such immunogenic activities can readily be determined by routine methods described herein and otherwise known in the art.
[0092] Thus, the invention further includes polypeptide variants which have a functional activity (e.g., biological activity and/or therapeutic activity).
In one embodiment, 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. In another embodiment, 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.
[0093] In preferred embodiments, the variants of the invention have conservative substitutions. By "conservative substitutions" is intended swaps within groups such as replacement of the aliphatic or hydrophobic amino acids Ala, Val, Leu and Ile;
replacement of the hydroxyl residues Ser and Thr; replacement of the acidic residues Asp and Glu;
replacement of the amide residues Asn and Gln, 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 Gly.
[0094] Guidance concerning how to make phenotypically silent amino acid substitutions is provided, for example, in Bowie et al., "Deciphering the Message in Protein Sequences: Tolerance to Amino Acid Substitutions," Science 247:1306-1310 (1990), wherein the authors indicate that there are two main strategies for studying the tolerance of an amino acid sequence to change.
[0095] 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.
[0096] 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 244:1081-1085 (1989). The resulting mutant molecules can then be tested for biological activity.
[0097] As the authors state, these two strategies have revealed that proteins are surprisingly tolerant of amino acid substitutions. The authors further indicate which amino acid changes are likely to be permissive at certain amino acid positions in the protein. For example, most buried (within the tertiary structure of the protein) amino acid residues require nonpolar side chains, whereas few features of surface side chains are generally conserved.
Moreover, tolerated conservative amino acid substitutions involve replacement of the aliphatic or hydrophobic amino acids Ala, Val, Leu and Ile; replacement of the hydroxyl residues Ser and Thr; replacement of the acidic residues Asp and Glu;
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 Gly. Besides conservative amino acid substitution, 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 substituent 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. Such variant polypeptides are deemed to be within the scope of those skilled in the art from the teachings herein.
[0098] For example, 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 ............................ õ.
immunogenic activity. See Pinckard et al., Clin. Exp. Irnmunol. 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).
[0099] In specific embodiments, 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. In preferred embodiments, the amino acid substitutions are conservative. Nucleic acids encoding these polypeptides are also encompassed by the invention.
[0100] 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. 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. Also, a given polypeptide may contain many types of modifications. 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.

(See, for instance, PROTEINS - STRUCTURE AND MOLECULAR PROPERTIES, 2nd Ed., T. E. Creighton, W. H. Freeman and Company, New York (1993); POST-TRANSLATIONAL COVALENT MODIFICATION OF PROTEINS, B. C. Johnson, Ed., Academic Press, New York, pgs. 1-12 (1983); Seifter et al., Meth. Enzymol.
182:626-646 (1990); Rattan etal., Ann. N.Y. Acad. Sci. 663:48-62 (1992)).
Functional activity [0101] "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.
[0102] "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 25-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).
[0103] In preferred embodiments, 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.
[0104] 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. Additionally, 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.
[0105] For example, in one embodiment where one is assaying for the ability of an albumin fusion protein to bind or compete with a Therapeutic protein for binding to an anti-Therapeutic polypeptide antibody and/or anti-albumin antibody, 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, etc. In one embodiment, antibody binding is detected by detecting a label on the primary antibody. In another embodiment, the primary antibody is detected by detecting binding of a secondary antibody or reagent to the primary antibody. In a further embodiment, 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.
[0106] In a preferred embodiment, where a binding partner (e.g., a receptor or a ligand) of a Therapeutic protein is identified, binding to that binding partner by an albumin fusion protein which comprises that Therapeutic protein as the Therapeutic protein portion of the fusion 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., Microbiol. Rev. 59:94-123 (1995). In another embodiment, 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.
[0107] In an alternative embodiment, where the ability of an albumin fusion protein to multimerize is being evaluated, 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.
[0108] In preferred embodiments, 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. In other preferred embodiments, 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.
[0109] 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. In particular, 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.
[0110] Assays for the ability of the albumin fusion proteins (e.g., comprising at least a fragment or variant of an antibody that binds a Therapeutic protein) 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. 5,571,698; 5,403,484; and 5,223,409), on plasmids (e.g., Cull et al., Proc. Natl.
Acad. Sci. USA 89:1865-1869 (1992)) or on phage (e.g., Scott and Smith, Science 249:386-390 (1990); Devlin, Science 249:404-406 (1990); Cwirla et al., Proc.
Natl. Acad.
Sci. USA 87:6378-6382 (1990); and Felici, J. Mol. Biol. 222:301-310 (1991)), 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.
[0111] 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.

101121 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, irnmunodiffitsion assays, agglutination assays, complement-fixation assays, immunoradiometric assays, fluorescent immunoassays, and protein A immunoassays, to name but a few. Such assays are routine and well known in the art (see, Ausubel et al, eds, 1994, Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York).
Exemplary immunoassays are described briefly below (but are not intended by way of limitation).
[0113] Immunoprecipitation protocols generally comprise lysing a population of cells in a lysis buffer such as RIPA buffer (1% NP-40 or TritoTti X-100, 1% sodium deoxycholate, 0.1% SDS, 0.15 M NaC1, 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 sepharosTe 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 beads in SDS/sample buffer. The ability of 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 sepharosig beads). For further discussion regarding 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.
101141 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-Tweernm20), 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., 32P or 125e diluted in blocking buffer, washing the membrane in wash buffer, and detecting the presence of the antigen. One of skill in the art would be knowledgeable as to the parameters that can be modified to increase the signal detected and to reduce the background noise. For further discussion regarding western blot protocols see, e.g., Ausubel et al, eds, 1994, Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York at 10.8.1.
[0115] 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. In ELISAs 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.
Further, instead of coating the well with the antigen, the albumin fusion protein may be coated to the well. In 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). One of skill in the art would be knowledgeable as to the parameters that can be modified to increase the signal detected as well as other variations of ELISAs known in the art. For further discussion regarding ELISAs see, e.g., Ausubel et al, eds, 1994, Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York at 11.2.1.
[0116] 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. One example of a competitive binding assay is a radioimmunoassay comprising the incubation of labeled antigen (e.g., 3H or 1251) 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. The affinity of the albumin fusion protein for 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. In this case, the protein, antigen or epitope is incubated with an albumin fusion protein conjugated to a labeled compound (e.g., 3H or 1251) 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.
[0117] In a preferred embodiment, 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.
[0118] 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, 10-2 M, 5 X 10-3 M, 10-3 M, 5 X 104 M, 104 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, 10-6M, 5 X le m, 107 M, 5 X 104 M or 104 M. Even more preferred binding affinities include those with a dissociation constant or Kd less than 5 X 10-9 M, 10-9 M, 5 X
10-1 M, 10-10 M, 5 10-11 10-11 m, 5 10-121\45 10-12 m¶, X 10-13 M, 10-13 M, 5 X 10-14 M, 10-14 1µ4, 5 X 10-15 M, or 10.15 M. In preferred embodiments, 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. In addition, assays described herein (see Examples and Table 1) and otherwise known in the art 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. Other methods will be known to the skilled artisan and are within the scope of the invention.

Albumin [0119] As described above, 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.
[0120] 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.
[01211 The terms, human serum albumin (HSA) and human albumin (HA) are used interchangeably herein. The terms, "albumin and "serum albumin" are broader, and encompass human serum albumin (and fragments and variants thereof) as well as albumin from other species (and fragments and variants thereof).
[0122] As used herein, "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. In particular, "albumin" refers to human albumin or fragments thereof (see for example, EP 201 239, EP 322 094 WO 97/24445, W095/23857) 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.
[01231 In preferred embodiments, 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 Val, 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. W095/23857 ).
In even more preferred embodiments, albumin fusion proteins of the invention that contain one or both of above-described sets of point mutations have improved stability/resistance to yeast Yap3p proteolytic cleavage, allowing increased production of recombinant albumin fusion proteins expressed in yeast host cells.
[0124] As used herein, a portion of albumin sufficient to prolong the therapeutic activity 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 of the protein so that the shelf life of the Therapeutic protein portion of the albumin fusion protein is prolonged or extended compared to the shelf-life 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.
[0125] 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. The term "variants" includes insertions, deletions and substitutions, either conservative or non conservative, where such changes do not substantially alter one or more of the oncotic, useful ligand-binding and non-immunogenic properties of albumin, or the active site, or active domain which confers the therapeutic activities of the Therapeutic proteins.
[0126] In particular, 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.
[0127] Generally speaking, 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 NO:1), 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 (195-585 of SEQ ID NO:1) or domains 1 and 3 (amino acids 1-194 of SEQ ID NO:1 and amino acids 388-585 of SEQ ID NO:1). 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 Lys106 to G1u119, G1u292 to Va1315 and G1u492 to A1a511.
[0128] Preferably, 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 [0129] 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.
Antibody structure and background [0130] 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 IcDa) and one "heavy" chain (about 50-70 IcDa). 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 isotype as IgM, IgD, 1gG, IgA, and IgE, respectively. See generally, Fundamental Immunology Chapters 3-5 (Paul, W., ed., 4th ed. Raven Press, N.Y.
(1998)), The variable regions of each light/heavy chain pair form the antibody binding site.
[0131]
Thus, an intact IgG antibody has two binding sites. Except in bifunctional or bispecific antibodies, the two binding sites are the same.
[0132] 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 FR1, CDR1, FR2, CDR2, FR3, CDR3 and FR4. The 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 MoL Biol. 196:901-917 (1987); Chothia et al. Nature 342:878-883 (1989).
[0133] As used herein, "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).
Antibodies that bind Therapeutic Proteins [0134] 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.
[0135] Antibodies that bind a Therapeutic protein (or fragment or variant thereof) may be from any animal origin, including birds and mammals. Preferably, the antibodies are human, murine (e.g., mouse and rat), donkey, sheep, rabbit, goat, guinea pig, camel, horse, or chicken antibodies. Most preferably, the antibodies are human antibodies. As used herein, "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.
[0136] 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., IgG 1 , IgG2, IgG3, IgG4, IgAl and IgA2) or subclass of immunoglobulin molecule. In preferred embodiments, 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.
[0137] Most preferably 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, CH1, CH2, and CH3 domains.
[0138] 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, bispecific, trispecific or of greater multispecificity. 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; Tuft, et al., J. Immunol. 147:60-69 (1991); U.S. Patent Nos.
4,474,893; 4,714,681;
4,925,648; 5,573,920; 5,601,819; Kostelny et al., J. Immunol. 148:1547-1553 (1992).
[0139] Antibodies that bind a Therapeutic protein (or fragment or variant thereof) 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. 148:1547 1553 (1992). In addition, bispecific antibodies may be formed as "diabodies" (Holliger et al. "Diabodies': 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 HIV
infected cells" EMBO J 10:3655-3659 (1991) and Traunecker et al. "Janusin: new molecular design for bispecific reagents" Int J Cancer Suppl 7:51-52 (1992)).
[0140] 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. Preferably, the variants (including derivatives) 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, VHCDR1, VHCDR2, VHCDR3, VL domain, VLCDR1, VLCDR2, or VLCDR3. In specific embodiments, the variants encode substitutions of VHCDR3. In a preferred embodiment, the variants have conservative amino acid substitutions at one or more predicted non-essential amino acid residues.
[0141] 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. In 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 [0142] 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. In specific embodiments, 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. In a specific embodiment, the above-described cross-reactivity is with respect to any single specific antigenic or immunogenic polypeptide, or combination(s) of 2, 3, 4, 5, or more of the specific antigenic and/or immunogenic polypeptides disclosed herein. In preferred embodiments, 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 of that particular antibody itself [0143] Further included in the present invention are antibodies which bind polypeptides encoded by polynucleotides which hybridize to a polynucleotide encoding a Therapeutic protein under stringent hybridization conditions (as described herein).
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, 10-2 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, 10-6M, 5 X 10-7 M, 107 M, 5 X 10-8 M or 10-8 M. Even more preferred 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, 10-10 M, 5 X 10-11 M, 10-111\455 10-12 1\4, 1012 M, X 10-13 M, 10-13 A455 10-14 1\4510-14 m-5 5 X 10-15M, or 10-15 M. In preferred embodiments, 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.
[0144] 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. In preferred embodiments, 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%. In preferred embodiments, 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. In other preferred embodiments, 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 95%, at least 90%, at least 85 %, at least 80%, at least 75%, at least 70%, at least 60%, or at least 50%.
[0145] 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 act as agonists or antagonists of the Therapeutic protein. For example, 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.
For example, 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). In specific embodiments, 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. In preferred embodiments, 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.
[0146] 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. Likewise, included in the invention are neutralizing antibodies which bind the ligand and prevent binding of the ligand to the receptor, as well as antibodies which bind the ligand, thereby preventing receptor activation, but do not prevent the ligand from binding the receptor. Further included in the invention are antibodies which activate the receptor. These antibodies 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. See, e.g., PCT publication WO 96/40281; U.S.
Patent No.
5,811,097; Deng et al., Blood 92(6):1981-1988 (1998); Chen et al., Cancer Res.

58(16):3668-3678 (1998); Harrop et al., J. Immunol. 161(4):1786-1794 (1998);
Zhu et al., Cancer Res. 58(15):3209-3214 (1998); Yoon et al., J. Immunol. 160(7):3170-3179 (1998);
Prat et al., J. Cell. Sci. 111(Pt2):237-247 (1998); Pitard et al., J. Immunol.
Methods 205(2):177-190 (1997); Liautard et al., Cytoldne 9(4):233-241 (1997); Carlson et al., J. Biol.
Chem. 272(17):11295-11301 (1997); Taryman et al., Neuron 14(4):755-762 (1995);
Muller et al., Structure 6(9):1153-1167 (1998); Bartunek et al., Cytokine 8(1):14-20 (1996).
In preferred embodiments, 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.
[0147] 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 in vitro and in vivo diagnostic and therapeutic methods. For example, the antibodies have utility in immunoassays for qualitatively and quantitatively measuring levels of the Therapeutic protein in biological samples. See, e.g., Harlow et al., Antibodies: A Laboratory Manual, (Cold Spring Harbor Laboratory Press, 2nd ed. 1988).
Likewise, 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.
[0148] 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. For example, but not by way of limitation, 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 ligand 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.
Methods of Producing Antibodies that bind Therapeutic Proteins [0149]
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. For example, 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.
Various 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 lysolecithin, 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.
[0150]
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. For example, 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 1-Cell Hybridomas (Elsevier, N.Y., 1981) 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.
[0151] Methods for producing and screening for specific antibodies using hybridoma technology are routine and well known in the art. In a non-limiting example, 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. Once an immune response is detected, e.g., antibodies specific for the antigen are detected in the mouse serum, 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.
[0152] Accordingly, 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.
[0153] Another well known method for producing both polyclonal and monoclonal human B cell lines is transformation using Epstein Barr Virus (EBV). 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.
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.
[0154] In general, the sample containing human B cells is innoculated with EBV, and cultured for 3-4 weeks. A typical source of EBV is the culture supernatant of the B95-8 cell line (ATCC #VR-1492). Physical signs of EBV transformation can generally be seen towards the end of the 3-4 week culture period. By phase-contrast microscopy, transformed cells may appear large, clear, hairy and tend to aggregate in tight clusters of cells.
Initially, 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. Alternatively, 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. Suitable fusion partners for EBV transformed cell lines include mouse myeloma cell lines (e.g., SP2/0, X63-Ag8.653), heteromyeloma cell lines (human x mouse; e.g, SPAM-8, SBC-H20, and CB-F7), and human cell lines (e.g., GM 1500, SKO-007, RPMI 8226, and KR-4). Thus, 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.
[0155] Antibody fragments which recognize specific epitopes may be generated by known techniques. For example, 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 CH1 domain of the heavy chain.
[0156] For example, antibodies that bind to a Therapeutic protein can also be generated using various phage display methods known in the art. In phage display methods, functional antibody domains are displayed on the surface of phage particles which carry the polynucleotide sequences encoding them. In a particular embodiment, such 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 III or gene VIII protein. Examples of 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-(1995); Kettleborough et al., Eur. J. Immunol. 24:952-958 (1994); Persic et al., Gene 187 9-18(1997); Burton et al., Advances in Immunology 57:191-280 (1994); PCT
application No.
PCT/GB91/01134; PCT publications WO 90/02809; WO 91/10737; WO 92/01047; WO
92/18619; WO 93/11236; WO 95/15982; WO 95/20401; and U.S. Patent Nos.
5,698,426;
5,223,409; 5,403,484; 5,580,717; 5,427,908; 5,750,753; 5,821,047; 5,571,698;
5,427,908;

. , 5,516,637; 5,780,225; 5,658,727; 5,733,743 and 5,969,108.
[0157]
As described in the above references, after phage selection, 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. For example, techniques to recombinantly produce Fab, Fab' and F(ab')2 fragments can also be employed using methods known in the art such as those disclosed in PCT publication WO 92/22324; Mullinax et al., BioTechniques 12(6):864-869 (1992); and Sawai et al., AJRI 34:26-34 (1995); and Better et al., Science 240:1041-1043 (1988).
[0158]
Examples of techniques which can be used to produce single-chain Fvs and antibodies include those described in U.S. Patents 4,946,778 and 5,258,498;
Huston et al., Methods in Enzymology 203:46-88 (1991); Shu et al., PNAS 90:7995-7999 (1993);
and Skerra et al., Science 240:1038-1040 (1988). For some uses, including in vivo use of antibodies in humans and in vitro detection assays, it may be preferable to use chimeric, humanized, or human antibodies. 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 at., BioTechniques 4:214 (1986);
Gillies et at., (1989) J. Immunol. Methods 125:191-202; U.S. Patent Nos. 5,807,715; 4,816,567;
and 4,816397.
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.
Often, 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 at., U.S.
Patent No.
5,585,089; Riechmann et al., Nature 332:323 (1988).

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; PadIan, 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).
[0159]
Completely 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.
[0160]
Human antibodies can also be produced using transgenic mice which are incapable of expressing functional endogenous immunoglobulins, but which can express human immunoglobulin genes. For example, the human heavy and light chain immunoglobulin gene complexes may be introduced randomly or by homologous recombination into mouse embryonic stem cells. Alternatively, 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. In particular, 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. Thus, using such a technique, it is possible to produce therapeutically useful IgG, IgA, IgM and IgE antibodies. For an overview of this technology for producing human antibodies, see Lonberg and Huszar, Int. Rev. Immunol.
13:65-93 (1995). For a detailed discussion of this technology for producing human antibodies and human monoclonal antibodies and protocols for producing such antibodies, see, e.g., PCT
publications WO 98/24893; WO 92/01047; WO 96/34096; WO 96/33735; European Patent No. 0 598 877; U.S. Patent Nos. 5,413,923; 5,625,126; 5,633,425; 5,569,825;
5,661,016;
5,545,806; 5,814,318; 5,885,793; 5,916,771; 5,939,598; 6,075,181; and 6,114,598.
In addition, companies such as Abgenix, Inc. (Freemont, CA) and Genpharm (San Jose, CA) can be engaged to provide human antibodies directed against a selected antigen using technology similar to that described above.
[0161] Completely human antibodies which recognize a selected epitope can be generated using a technique referred to as "guided selection." In this approach 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)).
Polynucleotides Encoding Antibodies [0162] 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.
[0163] The polynucleotides may be obtained, and the nucleotide sequence of the polynucleotides determined, by any method known in the art. For example, if the nucleotide sequence of the antibody is known, 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.
[0164] Alternatively, 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' 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 be cloned into replicable cloning vectors using any method well known in the art (See Example 65).
[0165] Once the nucleotide sequence and corresponding amino acid sequence of the antibody is determined, the nucleotide 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. (see, for example, the techniques described in Sambrook et al., 1990, Molecular Cloning, A Laboratory Manual, 2d Ed., Cold Spring Harbor Laboratory, Cold Spring Harbor, NY and Ausubel et al., eds., 1998, Current Protocols in Molecular Biology, John Wiley & Sons, NY), to generate antibodies having a different amino acid sequence, for example to create amino acid substitutions, deletions, and/or insertions.
[0166] In a specific embodiment, 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. Using routine recombinant DNA
techniques, 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. Mol. Biol. 278: 457-479 (1998) for a listing of human framework regions). Preferably, the polynucleotide generated by the combination of the framework regions and CDRs encodes an antibody that specifically binds a polypeptide of the invention. Preferably, as discussed supra, 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. Other alterations to the polynucleotide are encompassed by the present invention and within the skill of the art.
[0167] In addition, techniques developed for the production of "chimeric antibodies"
(Morrison et al., Proc. Natl. Acad. Sci. 81:851-855 (1984); Neuberger et al., Nature 312:604-608 (1984); Takeda et al., Nature 314:452-454 (1985)) by splicing genes from a mouse antibody molecule of appropriate antigen specificity together with genes from a human antibody molecule of appropriate biological activity can be used. As described supra, 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.
[0168] Alternatively, techniques described for the production of single chain antibodies (U.S. Patent No. 4,946,778; Bird, Science 242:423- 42 (1988);
Huston et al., Proc.
Natl. Acad. Sci. USA 85:5879-5883 (1988); and Ward et al., Nature 334:544-54 (1989)) can be adapted to produce single chain antibodies. Single chain antibodies are formed by linking the heavy and light chain fragments of the Fv region via an amino acid bridge, resulting in a single chain polypeptide. Techniques for the assembly of functional Fv fragments in E. coli may also be used (Skerra et al., Science 242:1038- 1041 (1988)).
Recombinant Expression of Antibodies [0169] Recombinant expression of an antibody, or fragment, derivative or analog thereof, (e.g., a heavy or light chain of an antibody or a single chain antibody), requires construction of 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. Methods which are well known to those skilled in the art can be used to construct expression vectors containing antibody coding sequences and appropriate transcriptional and translational control signals.
These methods include, for example, in vitro recombinant DNA techniques, synthetic techniques, and in vivo genetic recombination. The invention, thus, 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.
10170] 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.
Thus, 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. In preferred embodiments for the expression of double-chained antibodies, 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.
[0171] A variety of 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 mammalian viruses (e.g., the adenovirus late promoter; the vaccinia virus 7.5K promoter). Preferably, 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. For example, 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)).
[0172] In bacterial systems, a number of expression vectors may be advantageously selected depending upon the use intended for the antibody molecule being expressed. For example, when a large quantity of such a protein is to be produced, for the generation of pharmaceutical compositions of an antibody molecule, 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. 13:3101-3109 (1985); Van Heeke &
Schuster, J. Biol.
Chem. 24:5503-5509 (1989)); and the like. pGEX vectors may also be used to express foreign polypeptides as fusion proteins with glutathione S-transferase (GST).
In general, such 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.
[0173] In an insect system, Autographa californica nuclear polyhedrosis virus (AcNPV) is used as a vector to express foreign genes. The virus grows in Spodoptera frugiperda 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).
[0174] In mammalian host cells, a number of viral-based expression systems may be utilized. In cases where an adenovirus is used as an expression vector, 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. Acad. Sci. USA 81:355-359 (1984)). 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)).
[0175] In addition, 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. To this end, 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, Hela, 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.
[01761 For long-term, high-yield production of recombinant proteins, stable expression is preferred. For example, cell lines which stably express the antibody molecule may be engineered. Rather than using expression vectors which contain viral origins of replication, 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. Following the introduction of the foreign DNA, 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.

, . , [0177] 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. Also, 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. USA 78:2072 (1981)); neo, which confers resistance to the aminoglycoside G-418 Clinical Pharmacy 12:488-505; Wu and Wu, Biotherapy 3:87-95 (1991); Tolstoshev, Ann. Rev. Phannacol. Toxicol. 32:573-596 (1993);
Mulligan, Science 260:926-932 (1993); and Morgan and Anderson, Ann. Rev.
Biochem.
62:191-217 (1993); May, 1993, TIB TECH 11 (5):155-215 (1993)); and hygro, which confers resistance to hygromycin (Santerre et al., Gene 30:147 (1984)). Methods commonly known in the art of recombinant DNA technology may be routinely applied to select the desired recombinant clone, and such methods are described, for example, in Ausubel et al. (eds.), Current Protocols in Molecular Biology, John Wiley & Sons, NY (1993);
Kriegler, Gene Transfer and Expression, A Laboratory Manual, Stockton Press, NY (1990); and in Chapters 12 and 13, Dracopoli et al. (eds), Current Protocols in Human Genetics, John Wiley & Sons, NY (1994); Colberre-Garapin et al., J. Mol. Biol. 150:1 (1981), [0178] 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)). When a marker in the vector system expressing antibody is amplifiable, increase in the level of inhibitor present in culture of host cell will increase 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., Mol.
Cell. Biol. 3:257 (1983)).
[0179] Vectors which use glutamine synthase (GS) or DITFR 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: W087/04462;
W086/05807; W089/01036; W089/10404; and W091/06657.
Additionally, glutamine synthase expression vectors that may be used according to the present invention are commercially available from suppliers, including, for example Lonza Biologics, 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.
Frog. 11:1 (1995).
[0180] 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.
Alternatively, 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.
[0181] Once an antibody molecule of the invention has been produced by an animal, chemically synthesized, or recombinantly expressed, it 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. In addition, 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.
Modifications ofAntibodies [0182] Antibodies that bind a Therapeutic protein or fragments or variants can be fused to marker sequences, such as a peptide to facilitate purification. In preferred embodiments, 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. As described in Gentz et al., Proc.
Natl. Acad. Sci. USA 86:821-824 (1989), for instance, hexa-histidine provides for convenient purification of the fusion protein. Other 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.
[0183] 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. Examples of 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 1251, 1311, 111In or 99Tc. Other examples of detectable substances have been described elsewhere herein.
[0184] Further, 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, 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., vincristine and vinblastine).
[0185] 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. For example, 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, 13-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 II (See, International Publication No.
WO 97/34911), Fas Ligand (Takahashi et al., mt. Immund, 6:1567-1574 (1994)), VEGI
(See, International Publication No. WO 99/23105), a thrombotic agent or an anti- angiogenic agent, e.g., angiostatin or endostatin; or, biological response modifiers such as, for example, lymphokines, interleukin-1 ("IL-1"), 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.
[0186] Antibodies may also be attached to solid supports, which are particularly useful for immunoassays or purification of the target antigen. Such solid supports include, but are not limited to, glass, cellulose, polyacrylamide, nylon, polystyrene, polyvinyl chloride or polypropylene.
[0187] 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. 1985); Hellstrom et al., "Antibodies For Drug Delivery", in Controlled Drug Delivery (2nd Ed.), Robinson et al. (eds.), pp.
623-53 (Marcel Dekker, Inc. 1987); Thorpe, "Antibody Carriers Of Cytotoxic Agents In Cancer Therapy: A
Review", in Monoclonal Antibodies '84: Biological And Clinical Applications, Pinchera et al. (eds.), pp. 475-506 (1985); "Analysis, Results, And Future Prospective Of The Therapeutic Use Of Radiolabeled Antibody In Cancer Therapy", in Monoclonal Antibodies For Cancer Detection And Therapy, Baldwin et al. (eds.), pp. 303-16 (Academic Press 1985), and Thorpe et al., "The Preparation And Cytotoxic Properties Of Antibody-Toxin Conjugates", Immunol. Rev. 62:119-58 (1982).
[0188] Alternatively, 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.
[0189] An antibody, with or without a therapeutic moiety conjugated to it, administered alone or in combination with cytotoxic factor(s) ancUor cytokine(s) can be used as a therapeutic.
Antibody-albumin fusion [0190] 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.
[0191] In specific embodiments, 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.
In other embodiments, 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. In other embodiments, 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 CDR1. In other embodiments, 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. In other embodiments, 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.
[0192] In specific embodiments, 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.
In other embodiments, 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. In other embodiments, 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 CDR1. In other embodiments, 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. In other embodiments, 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 CDR3.
[0193] In other embodiments, 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.
[0194] In preferred embodiments, 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 (Gly4Ser)3(SEQ ID NO:4).
Immunophenotyping [0195] 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, 96:737-49 (1999)).
[0196] These techniques allow for the screening of particular populations of cells, such as might be found with hematological malignancies (i.e. minimal residual disease (MRD) in acute leukemic patients) and "non-self' cells in transplantations to prevent Graft-versus-Host Disease (GVHD). Alternatively, these techniques allow for the screening of hematopoietic stem and progenitor cells capable of undergoing proliferation and/or differentiation, as might be found in human umbilical cord blood.
Characterizing Antibodies that bind a Therapeutic Protein and Albumin Fusion Proteins Comprising a Fragment or Variant of an Antibody that binds a Therapeutic Protein [0197] 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. In particular, 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.
[0198] 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. 5,571,698;
5,403,484; and 5,223,409), on plasmids (e.g., Cull et al., Proc. Natl. Acad.
Sci. USA
89:1865-1869 (1992)) or on phage (e.g., Scott and Smith, Science 249:386-390 (1990);
Devlin, Science 249:404-406 (1990); Cwirla et al., Proc. Natl. Acad. Sci. USA
87:6378-6382 (1990); and Felici, J. Mol. Biol. 222:301-310 (1991)).
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.
[0199] 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.
[0200] 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, irrununoprecipitation assays, precipitin reactions, gel diffusion precipitin reactions, immunodiffusion assays, agglutination assays, complement-fixation assays, inununoradiometric assays, fluorescent immunoassays, and protein A immunoassays, to name but a few. Such assays are routine and well known in the art (see, e.g., Ausubel et al, eds, 1994, Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York, Exemplary immunoassays are described briefly below (but are not intended by way of limitation).
[0201] 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 NaC1, 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 lysate, 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 or variant of a Therapeutic antibody) to the cell lysate, incubating for about an hour or more at 40 degrees C, washing the beads in lysis buffer and resuspending the beads in SDS/sample buffer. 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). For further discussion regarding 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.
[0202] 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 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., 32P or 125I) diluted in blocking buffer, washing the membrane in wash buffer, and detecting the presence of the antigen. One of skill in the art would be knowledgeable as to the parameters that can be modified to increase the signal detected and to reduce the background noise. For further discussion regarding western blot protocols see, e.g., Ausubel et al, eds, 1994, Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York at 10.8.1.
[0203] 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-specifically bound albumin fusion proteins, and detecting the presence of the antibody or albumin fusion proteins specifically bound to the antigen coating the well. In ELISAs 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. In 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).
One of skill in the art would be knowledgeable as to the parameters that can be modified to increase the signal detected as well as other variations of ELISAs known in the art. For further discussion regarding ELISAs see, e.g., Ausubel et al, eds, 1994, Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York at 11.2.1.
[0204] 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. One example of a competitive binding assay is a radioimmunoassay comprising the incubation of labeled antigen (e.g., 3H or 1251) 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.
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 Scatchard 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. In this case, the protein, antigen or epitope is incubated with an antibody or albumin fusion protein of the invention conjugated to a labeled compound (e.g., 3H or 1251) 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.
[0205] In a preferred embodiment, 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.
Therapeutic Uses [0206] 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.
[0207] In a specific and preferred embodiment, 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.
[0208] 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. Armed with the teachings provided herein, one of ordinary skill in the art will know how to use 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 for diagnostic, monitoring or therapeutic purposes without undue experimentation.
[0209] 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 IL-7), for example, which serve to increase the number or activity of effector cells which interact with the antibodies.
[0210] 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.
[0211] It is preferred to use high affinity and/or potent in vivo inhibiting and/or neutralizing antibodies against Therapeutic proteins, fragments or regions thereof, (or the albumin fusion protein correlate of such an antibody) for both immunoassays directed to and therapy of disorders related to polynucleotides or polypeptides, including fragments thereof, of the present invention. Such antibodies, fragments, or regions, will preferably have an affinity for polynucleotides or polypeptides of the invention, including fragments thereof Preferred binding affinities include dissociation constants or Kd's less than 5 X 10-2 M, 10-2 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, 10-6M, 5 X 10-7 M, 107 M, 5 X 10-8 M or 10-8 M. Even more preferred 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, 10-10 M, 5 X 1011 M, 10-11 M, 5 x 10-12 m, 10-12 M, X 10-13 M, 10-13 m, 5 x 10-14 m, 10-14 m¨, 5 X 1045 M, or 10-m.
Gene Therapy [0212] In a specific embodiment, 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.
In this embodiment of the invention, the nucleic acids produce their encoded protein that mediates a therapeutic effect.
[0213] Any of the methods for gene therapy available in the art can be used according to the present invention. Exemplary methods are described in more detail elsewhere in this application.
Demonstration of Therapeutic or Prophylactic Activity [0214] 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. For example, 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 accordance with the invention, 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.
Therapeutic/Prophylactic Administration and Composition [0215] 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. In a preferred embodiment, 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.
[0216] 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.
[0217] 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. In addition, it may be desirable to introduce the pharmaceutical compounds or compositions of the invention into the central nervous system by any suitable route, including intraventricular and intrathecal injection; 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.
[0218] In a specific embodiment, it may be desirable to administer the pharmaceutical compounds or compositions of the invention locally to the area in need of treatment; this may be achieved by, for example, and not by way of limitation, local infusion during surgery, topical application, e.g., in conjunction with a wound dressing after surgery, by injection, by means of a catheter, by means of a suppository, or by means of an implant, said implant being of a porous, non-porous, or gelatinous material, including membranes, such as sialastic membranes, or fibers. Preferably, when administering a protein, including an antibody, of the invention, care must be taken to use materials to which the protein does not absorb.
[0219] In another embodiment, 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.) [0220] In yet another embodiment, the compound or composition can be delivered in a controlled release system. In one embodiment, 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)). In another embodiment, 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., Arm. Neurol. 25:351 (1989); Howard et al., J.Neurosurg. 71:105 (1989)). In yet another embodiment, 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)).
[0221] Other controlled release systems are discussed in the review by Langer (Science 249:1527-1533 (1990)).
[0222] In a specific embodiment where the compound of the invention is a nucleic acid encoding a protein, 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. 4,980,286), or by direct injection, or by use of microparticle bombardment (e.g., a gene gun; Biolistic, Dupont), or coating with lipids or cell-surface receptors or transfecting agents, or by administering it in linkage to a homeobox- like peptide which is known to enter the nucleus (see e.g., Joliot et al., Proc. Natl. Acad. Sci.
USA 88:1864-1868 (1991)), etc. Alternatively, a nucleic acid can be introduced intracellularly and incorporated within host cell DNA for expression, by homologous recombination.
[0223] The present invention also provides pharmaceutical compositions.
Such compositions comprise a therapeutically effective amount of a compound, and a pharmaceutically acceptable carrier. In a specific embodiment, the term "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. The term "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. These 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.
[0224] In a preferred embodiment, the composition is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous administration to human beings. Typically, compositions for intravenous administration are solutions in sterile isotonic aqueous buffer.
Where necessary, the composition may also include a solubilizing agent and a local anesthetic such as lignocaine to ease pain at the site of the injection. Generally, 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. Where the composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline. Where the composition is administered by injection, an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
[0225] 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.
[0226] 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. In 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.

[0227] For antibodies, the dosage administered to a patient is typically 0.1 mg/kg to 100 mg/kg of the patient's body weight. Preferably, the dosage administered to a patient is between 0.1 mg/kg and 20 mg/kg of the patient's body weight, more preferably 1 mg/kg to 10 mg/kg of the patient's body weight. Generally, 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. Further, 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.
Diagnosis and Imaging [0228] Labeled antibodies and derivatives and analogs thereof that bind a Therapeutic protein (or fragment or variant thereof) (including albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein), 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.
[0229] 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. With respect to cancer, 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.
[0230] 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); Jalkanem 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 are known in the art and 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.
[0231] One facet of the invention is the detection and diagnosis of a disease or disorder associated with aberrant expression of a Therapeutic protein in an animal, preferably a mammal and most preferably a human. In one embodiment, 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 that 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.
[0232] It will be understood in the art that the size of the subject and the imaging system used will determine the quantity of imaging moiety needed to produce diagnostic images. In the case of a radioisotope moiety, for a human subject, 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. 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)).
[0233] Depending on several variables, including the type of label used and the mode of administration, 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.
[0234] In an embodiment, 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.
[0235] 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.
[0236] In a specific embodiment, 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). In another embodiment, the molecule is labeled with a fluorescent compound and is detected in the patient using a fluorescence responsive scanning instrument.
In another embodiment, the molecule is labeled with a positron emitting metal and is detected in the patent using positron emission-tomography. In yet another embodiment, the molecule is labeled with a paramagnetic label and is detected in a patient using magnetic resonance imaging (MRI). Antibodies that specifically detect the albumin fusion protein but not albumin or the therapeutic protein alone are a preferred embodiment. These can be used to detect the albumin fusion protein as described throughout the specification.
Kits [0237] The present invention provides kits that can be used in the above methods. In one embodiment, a kit comprises an antibody, preferably a purified antibody, in one or more containers. In a specific embodiment, 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. Preferably, the kits of the present invention further comprise a control antibody which does not react with the polypeptide of interest. In another specific embodiment, the 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).
[0238] In another specific embodiment of the present invention, the kit is a diagnostic kit for use in screening serum containing antibodies specific against proliferative and/or cancerous polynucleotides and polypeptides. Such a kit may include a control antibody that does not react with the polypeptide of interest. Such a kit may include a substantially isolated polypeptide antigen comprising an epitope which is specifically immunoreactive with at least one anti-polypeptide antigen antibody. Further, such 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). In specific embodiments, 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.
[0239] In a more specific embodiment the detecting means of the above-described kit includes a solid support to which said polypeptide antigen is attached. Such a kit may also include a non-attached reporter-labeled anti-human antibody. In this embodiment, binding of the antibody to the polypeptide antigen can be detected by binding of the said reporter-labeled antibody.
[0240] In an additional embodiment, 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 immunoreactive with polypeptide or polynucleotide antigens, and means for detecting the binding of the polynucleotide or polypeptide antigen to the antibody. In one embodiment, the antibody is attached to a solid support. In a specific embodiment, 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.

[0241] In one diagnostic configuration, test serum is reacted with a solid phase reagent having a surface-bound antigen obtained by the methods of the present invention.
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. Typically, 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).
[0242] 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).
[0243] Thus, 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 Proteins [0244] The present invention relates generally to albumin fusion proteins and methods of treating, preventing, or ameliorating diseases or disorders. As used herein, "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 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) 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.
[0245] In a preferred embodiment, 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.
[0246] 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 in frame to at least one polynucleotide encoding at least one molecule of a Therapeutic protein (or fragment or variant thereof), further comprising, for example, one or more of the following elements: (1) a functional self-replicating vector (including but not limited to, a shuttle vector, an expression vector, an integration vector, and/or a replication system), (2) a region for initiation of transcription (e.g., a promoter region, such as for example, a regulatable or inducible promoter, a constitutive promoter), (3) a region for termination of transcription, (4) a leader sequence, and (5) a selectable marker.
[0247] In one embodiment, 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. In other embodiments, 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. In other embodiments, 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. In preferred embodiments, the serum albumin protein component of the albumin fusion protein is the mature portion of serum albumin.
[0248] In further embodiments, 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. In further embodiments, 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. In preferred embodiments, the Therapeutic protein portion of the albumin fusion protein is the mature portion of the Therapeutic protein.
102491 In further embodiments, 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. In preferred embodiments, 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.
10250] Preferably, the albumin fusion protein comprises HA as the N-terminal portion, and a Therapeutic protein as the C-terminal portion. Alternatively, an albumin fusion protein comprising HA as the C-terminal portion, and a Therapeutic protein as the N-terminal portion may also be used.
[0251] In other embodiments, the albumin fusion protein has a Therapeutic protein fused to both the N-terminus and the C-terminus of albumin. In a preferred embodiment, the Therapeutic proteins fused at the N- and C- termini are the same Therapeutic proteins. In an alternative preferred embodiment, the Therapeutic proteins fused at the N- and C- termini are different Therapeutic proteins. In another preferred embodiment, 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). In another preferred embodiment, 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.
102521 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).
[0253] In one embodiment, one, two, three, or more tandemly oriented Therapeutic protein X polypeptides (or fragments or variants thereof) 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.
[0254]
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.
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.
[0255]
Further, 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. In one embodiment of the invention, albumin fusion proteins may be in monomeric or multimeric forms (i.e., dimers, trimers, tetramers and higher multimers). In a further embodiment of the invention, 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). In a specific embodiment, 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.
[0256] In addition to albumin fusion protein in which the albumin portion is fused N-terminal and/or C-terminal of the Therapeutic protein portion, 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 a-helices, which are stabilized by disulphide bonds. The loops, as determined from the crystal structure of HA (PDB identifiers 1A06, 1BJ5, 1BKE, 1BMO, 1E7E to 1E71 and lUOR) 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.
[0257] Loops in human albumin structure into which peptides or polypeptides may be inserted to generate albumin fusion proteins of the invention include: Va154-Asn61, Thr76-Asp89, A1a92-G1u100, G1n170-A1a176, His 247 - G1u252, Glu 266 - G1u277, Glu His288, A1a362-G1u368, Lys439-Pro447, Va1462-Lys475, Thr478-Pro486, and Lys560-Thr566. In more preferred embodiments, peptides or polypeptides are inserted into the Va154-Asn61, Glnl 70-Alal 76, and/or Lys560-Thr566 loops of mature human albumin (SEQ
ID NO:1).
[0258] 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.
[0259] Such library(s) could be generated on HA or domain fragments of HA
by one of the following methods:
[0260] 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;
[0261] 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 Xn (where X is an amino acid and n is the number of residues;
[0262] N-, C- or N- and C- terminal peptide/protein fusions in addition to (a) and/or (b).
[0263] 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.

[0264] In preferred embodiments, 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. For example, short peptides described in Table 1 and 2 (e.g., Therapeutic Y) can be inserted into the albumin loops.
[0265] Generally, 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 the same or different Therapeutic protein molecules. The structure of bifunctional albumin fusion proteins may be represented as: X-HA-Y or Y-HA-X.
[0266] For example, an anti-BLySTM scFv-HA-IFNa-2b fusion may be prepared to modulate the immune response to IFNa-2b by anti-BLySTM scFv. An alternative is making a bi (or even multi) functional dose of HA-fusions e.g. HA-IFNa-2b fusion mixed with HA-anti-BLySTM scFv fusion or other HA-fusions in various ratio's depending on function, half-life etc.
[0267] 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.
[0268] As an alternative to the fusion of known therapeutic molecules, 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 Xn (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. 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.
[0269] Additionally, 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.
[0270] The linker sequence may be cleavable by a protease or chemically to yield the growth hormone related moiety. Preferably, the protease is one which is produced naturally by the host, for example the S. cerevisiae protease kex2 or equivalent proteases.
[0271] Therefore, as described above, the albumin fusion proteins of the invention may have the following formula RI -L-R2; R2-L-R1; or R1-L-R2-L-R1, wherein R1 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.
[0272] In preferred embodiments, 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.
[0273] 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. When 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. As an example, an albumin fusion protein of the invention may retain greater than about 100% of the therapeutic activity, or greater than about 105%, 110%, 120%, 130%, 150% 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.
[0274] 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 60%, 70%, 80%, or 90% or more of the therapeutic activity of the equivalent unfused Therapeutic protein when subjected to the same conditions.
Expression of Fusion Proteins [0291] 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. Preferably, the polypeptide is secreted from the host cells.
[0292] 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.
[0293] The Saccharomyces cerevisiae invertase signal is a preferred example of a yeast-derived signal sequence.
[0294] Conjugates of the kind prepared by Poznansky et al., (FEBS Lett.
239:18 (1988)), in which separately-prepared polypeptides are joined by chemical cross-linking, are not contemplated.
[0295] The present invention also includes a cell, preferably a yeast cell transformed to express an albumin fusion protein of the invention. In addition to the transformed host cells themselves, 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. coli and Bacillus subtilis), yeasts (for example Saccharomyces cerevisiae, Kluyveromyces lactis and Pichia pastoris, filamentous fungi (for example Aspergillus), plant cells, animal cells and insect cells.
[0296] Preferred yeast strains to be used in the production of albumin fusion proteins are D88, DXY1 and BXP1 O. D88 [leu2-3, leu2-122, can], pral, ubc4} is a derivative of parent strain AH22his+ (also known as DB1; see, e.g., Sleep et al.
Biotechnology 8:42-46 (1990)). The strain contains a leu2 mutation which allows for auxotropic selection of 2 micron-based plasmids that contain the LEU2 gene. D88 also exhibits a derepression of PRB1 in glucose excess. The PRB1 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 PRA1 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. W099/00504).
[0297] DXY1, a derivative of D88, has the following genotype: [leu2-3, leu2-122, can], pral, ubc4, ura3::yap3]. In addition to the mutations isolated in D88, 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)).
[0298] BXP10 has the following genotype: leu2-3, leu2-122, can], pral, ubc4, ura3, yap3::URA3, lys2, hsp150::LYS2, pmt1::URA3. In addition to the mutations isolated in DXY1, this strain also has a knockout of the PMT1 gene and the HSP150 gene.
The PMT1 gene is a member of the evolutionarily conserved family of dolichyl-phosphate-D-mannose protein 0-mannosyltransferases (Pmts). The transmembrane topology of Pmtlp suggests that it is an integral membrane protein of the endoplasmic reticulum with a role in 0-linked glycosylation. This mutation serves to reduce/eliminate 0-linked glycosylation of HSA
fusions (see, e.g., International Publication No. W000/44772).
Studies revealed that the Hsp150 protein is inefficiently separated from rHA by ion exchange chromatography. The mutation in the HSP150 gene removes a potential contaminant that has proven difficult to remove by standard purification techniques. See, e.g., U.S. Patent No. 5,783,423.
[0299] 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.
[0300] 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)J
MoL Biol.
98, 503 or Berent et al. (1985) Biotech. 3, 208. Alternatively, the presence of the protein in the supernatant can be detected using antibodies.
[0301] Useful yeast plasmid vectors include pRS403-406 and pRS413-416 and are generally available from Stratagene Cloning Systems, La Jolla, CA 92037, USA.
Plasmids pRS403, pRS404, pRS405 and pRS406 are Yeast Integrating plasmids (Yips) and incorporate the yeast selectable markers HIS3, 7RP1, LEU2 and URA3. Plasmids pRS413-416 are Yeast Centromere plasmids (Ycps).
[0302] Preferred vectors for making albumin fusion proteins for expression in yeast include pPPC0005, 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 PRB1 S. cerevisiae promoter (PRB1p), a Fusion leader sequence (FL), DNA encoding HA (rHA) and an ADH1 S. cerevisiae terminator sequence. The 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-387 319).
[0303] The plasmids, pPPC0005, pScCHSA, pScNHSA, and pC4:HSA were deposited on April 11, 2001 at the American Type Culture Collection, 10801 University Boulevard, Manassas, Virginia 20110-2209 and given accession numbers ATCC PTA-3278, PTA-3276, PTA-3279, and PTA-3277, respectively. Another vector useful for expressing an albumin fusion protein in yeast the pSAC35 vector which is described in Sleep et al., BioTechnology 8:42 (1990).
[0304] Another yeast promoter that can be used to express the albumin fusion protein is the MET25 promoter. See, for example, Dominik Mumburg, Rolf Muller and Martin Funk. Nucleic Acids Research, 1994, Vol. 22, No. 25, pp. 5767-5768. The Met25 promoter is 383 bases long (bases ¨382 to ¨1) and the genes expressed by this promoter are also known as Met15, Met17, 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:
GCGGCCGCCGGATGCAAGGGTTCGAATCCCTTAGCTCTCATTATTTTTTGCTTTTT
CTCTTGAGGTCACATGATCGCAAAATGGCAAATGGCACGTGAAGCTGTCGATATT
GGGGAACTGTGGTGGTTGGCAAATGACTAATTAAGTTAGTCAAGGCGCCATCCTC
ATGAAAACTGTGTAACATAATAACCGAAGTGTCGAAAAGGTGGCACCTTGTCCA
ATTGAACACGCTCGATGAAAAAAATAAGATATATATAAGGTTAAGTAAAGCGTC
TGTTAGAAAGGAAGTTTTTCCTTTTTCTTGCTCTCTTGTCTTTTCATCTACTATTTC
CTTCGTGTAATACAGGGTCGTCAGATACATAGATACAATTCTATTACCCCCATCC
ATACAATG (SEQ ID NO:5) [0305] 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.
[0306] 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'-exonucleolytic activities, and fill in recessed 3'-ends with their polymerizing activities.
[0307] The combination of these activities therefore generates blunt-ended DNA
segments. The blunt-ended segments are then incubated with a large molar excess of linker molecules in the presence of an enzyme that is able to catalyze the ligation of blunt-ended DNA molecules, such as bacteriophage T4 DNA ligase. Thus, the products of the reaction are DNA segments carrying polymeric linker sequences at their ends. These DNA
segments are then cleaved with the appropriate restriction enzyme and ligated to an expression vector that has been cleaved with an enzyme that produces termini compatible with those of the DNA
segment.
[0308] Synthetic linkers containing a variety of restriction endonuclease sites are commercially available from a number of sources including International Biotechnologies Inc, New Haven, CT, USA.
[0309] A desirable way to modify the DNA in accordance with the invention, if, for example, HA variants are to be prepared, is to use the polymerase chain reaction as disclosed by Saiki et al. (1988) Science 239, 487-491. In this method 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.
[0310] 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, Schizosaccharomyces, Citeromyces, Pachysolen, Debaromyces, Metschunikowia, Rhodosporidium, Leucosporidium, Bonyoascus, Sporidiobolus, Endomycopsis, and the like. Preferred genera are those selected from the group consisting of Saccharomyces, Schizosaccharomyces, Kluyveromyces, Pichia and Torulaspora. Examples of Saccharomyces spp. are S. cerevisiae, S. italicus and S. rouxii.
[0311] 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. polymorpha), 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.
[0312] Preferred exemplary species of Saccharomyces include S.
cerevisiae, S.
italicus, S. diastaticus, and Zygosaccharomyces rouxii. 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. For example, the following preferred 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 hsp150 mutant (ATCC
Accession No. 4021266); Saccharomyces cerevisiae Hansen, teleomorph strain BY4743 pmtl mutant (ATCC Accession No. 4023792); Saccharomyces cerevisiae Hansen, teleomorph (ATCC Accession Nos. 20626; 44773; 44774; and 62995); Saccharomyces diastaticus Andrews et Gilliland ex van der Walt, teleomorph (ATCC Accession No. 62987);
Kluyveromyces lactis (Dombrowski) van der Walt, teleomorph (ATCC Accession No.

76492); Pichia angusta (Teunisson et al.) Kurtzman, teleomorph deposited as Hansenula polymorpha de Morais et Maia, teleomorph (ATCC Accession No. 26012);
Aspergillus niger van Tieghem, anamorph (ATCC Accession No. 9029); Aspergillus niger van Tieghem, anamorph (ATCC Accession No. 16404); Aspergillus nidulans (Eidam) Winter, anamorph (ATCC Accession No. 48756); and Yarrowia lipolytica (Wickerham et al.) van der Walt et von Arx, teleomorph (ATCC Accession No. 201847).
[0313] Suitable promoters for S. cerevisiae include those associated with the PGKI
gene, GAL1 or GAL10 genes, CYCI, PH05, TRPI, ADHI, ADH2, the genes for glyceraldehyde-3-phosphate dehydrogenase, hexokinase, pyruvate decarboxylase, phosphofructokinase, triose phosphate isomerase, phosphoglucose isomerase, glucolcinase, 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-258 067).
[0314] Convenient regulatable promoters for use in Schizosaccharomyces pombe are the thiamine-repressible promoter from the nmt gene as described by Maundrell (1990) 1 Biol. Chem. 265, 10857-10864 and the glucose repressible jbpl gene promoter as described by Hoffman & Winston (1990) Genetics 124, 807-816.
[0315] Methods of transforming Pichia for expression of foreign genes are taught in, for example, Cregg et al. (1993), and various Phillips patents (e.g. US 4 857 467), and Pichia expression kits are commercially available from Invitrogen By, 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 MOX1 and FMD1; 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.
[0316] 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.
[0317] The desired albumin fusion protein 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-1 signal sequence (e.g., amino acids 1-21 of GenBank Accession number AAB51134) MKVSVAALSCLMLVTALGSQA (SEQ ID NO:6) b) the stanniocalcin signal sequence (MLQNSAVLLLLVISASA, SEQ ID NO:7) c) the pre-pro region of the HSA signal sequence (e.g., MKWVTFISLLFLFSSAYSRGVFRR, 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) 0 the yeast mating factor alpha signal sequence (e.g., MRFP S IFTAVLAFAAS S ALAAPVNTTTEDETAQIPAEAVIGY S DLEGDFDV
AVLPFSNSTNNGLLFINTTIASIAAKEEGVSLEKR, SEQ ID NO:12 or MRFPSIFTAVLAFAASSALAAPVNTTTEDETAQIPAEAVIGYSDLEGDFDV
AVLPFSNSTNNGLLFINTTIASIAAKEEGVSLDKR, SEQ ID NO:12) g) K lactis killer toxin leader sequence h) a hybrid signal sequence (e.g., MKWVSFISLLFLFSSAYSRSLEICR, SEQ ID
NO:13) i) an HSA/MFa-1 hybrid signal sequence (also known as HSA/kex2) (e.g., MKWVSFISLLFLFSSAYSRSLDKR, SEQ ID NO:14) j) a K lactis killer/ MFcc-1 fusion leader sequence (e.g., MNIFYIFLFLLSFVQGSLDKR, SEQ ID NO:15) k) the Immunoglobulin Ig signal sequence (e.g., MGWSCIILFLVATATGVHS, SEQ
ID NO:16) 1) the Fibulin B precursor signal sequence (e.g., MERAAPSRRVPLPLLLLGGLALLAAGVDA, SEQ ID NO:17) m) the clusterin precursor signal sequence (e.g., MMKTLLLFVGLLLTWESGQVLG, SEQ ID NO:18) n) the insulin-like growth factor-binding protein 4 signal sequence (e.g., MLPLCLVAALLLAAGPGPSLG, SEQ ID NO:19) o) variants of the pre-pro-region of the HSA signal sequence such as, for example, MKWVSFISLLFLFSSAYSRGVFRR (SEQ ID NO :20), MKWVTFISLLFLFAGVLG (SEQ ID NO:21), MKWVTFISLLFLFSGVLG (SEQ ID NO:22), MKWVTFISLLFLFGGVLG (SEQ ID NO:23), Modified HSA leader HSA #64 MKWVTFISLLFLFAGVSG (SEQ ID NO:24);
Modified HSA leader HSA #66 MKWVTFISLLFLFGGVSG (SEQ ID NO :25);
Modified HSA (A14) leader ¨
MKWVTFISLLFLFAGVSG (SEQ ID NO :26);
Modified HSA (S14) 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
NO :30) q) acid phosphatase (PH05) leader (e.g., MFKSVVYSILAASLANA SEQ ID
NO: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) S. diastaticus glucoamylase Ii secretion leader sequence x) S. carlsbergensis a-galactosidase (MEL1) secretion leader sequence y) Candida glucoarnylase leader sequence z) The hybrid leaders disclosed in EP-A-387 319 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); Or dd) Inulinase ¨ MKLAYSLLLPLAGVSASVINYKR (SEQ ID NO:33).
ee) A modified TA57 propeptide leader variant #1 ¨
MKLKTVRSAVLSSLFASQVLGQPIDDTESQTTSVNLMADDTESAFATQTN
SGGLDVVGLISMAKR (SEQ ID NO:34) ff) A modified TA57 propeptide leader variant #2 ¨
MKLKTVRSAVLSSLFASQVLGQPIDDTESQTTSVNLMADDTESAFATQTN
SGGLDVVGLISMAEEGEPKR (SEQ ID NO:35) gg) A consensus signal peptide ¨
MWWRLWWLLLLLLLLWPMVWA (SEQ ID NO:550) Additional Methods of Recombinant and Synthetic Production of Albumin Fusion Proteins [0318] 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.
[0319] The polynucleotides encoding albumin fusion proteins of the invention may be joined to a vector containing a selectable marker for propagation in a host.
Generally, 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.
[0320] The polynucleotide insert should be operatively linked to an appropriate promoter, such as the phage lambda PL promoter, the E. coil lac, trp, phoA and tac promoters, the SV40 early and late promoters and promoters of retroviral LTRs, to name a few. Other suitable promoters will be known to the skilled artisan. 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.
[0321] As indicated, the expression vectors will preferably include at least one selectable marker. Such 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. coil and other bacteria. Representative examples of appropriate hosts include, but are not limited to, bacterial cells, such as E.
colt, Streptomyces and Salmonella typhimurium cells; fungal cells, such as yeast cells (e.g., Saccharomyces cerevisiae or Pichia pastoris (ATCC Accession No. 201178)); insect cells such as Drosophila S2 and Spodoptera Sf9 cells; animal cells such as CHO, COS, NSO, 293, and Bowes melanoma cells; and plant cells. Appropriate culture mediums and conditions for the above-described host cells are known in the art.
[0322] Among 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, pl(K223-3, pKK233-3, pDR540, pRIT5 available from Pharmacia Biotech, Inc. Among preferred eukaryotic vectors are pWLNEO, pSV2CAT, p0G44, pXT1 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, pYD1, pTEF1/Zeo, pYES2/GS, pPICZ, pGAPZ, pGAPZalph, pPIC9, pPIC3.5, pHIL-D2, pHIL-S1, pPIC3.5K, pPIC9K, and PA0815 (all available from Invitrogen, Carlbad, CA).
Other suitable vectors will be readily apparent to the skilled artisan.
[0323] In one embodiment, 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.
For example, in E. colt, 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. 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.
In a specific embodiment, 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.
103241 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-1 signal sequence (e.g., amino acids 1-21 of GenBank Accession number AAB51134) MKVSVAALSCLMLVTALGSQA (SEQ ID NO:6) b) the stanniocalcin signal sequence (MLQNSAVLLLLVISASA, SEQ ID NO:7) c) the pre-pro region of the HSA signal sequence (e.g., MKWVTFISLLFLFSSAYSRGVFRR, SEQ ID NO:8) d) the pre region of the USA signal sequence (e.g., MKWVTF1SLLFLFSSAYS, SEQ
ID NO:9) or variants thereof, such as, for example, MKWVSFISLLFLFSSAYS, (SEQ
ID NO:10) e) the invertase signal sequence (e.g., MLLQAFLFLLAGFAAICISA, SEQ ID NO:11) f) the yeast mating factor alpha signal sequence (e.g., MRFPSIFTAVLAFAASSALAAPVNITIEDETAQIPAEAVIGYSDLEGDFDVAVL
PFSNSTNNGLLFINTTIASIAAKEEGVSLEKR, SEQ ID NO:12 or MRFPSIFTAVLAFAASSALAAPVNTTTEDETAQIPAEAVIGYSDLEGDFDVAVL
PFSNSTNNGLLFINTTIASIAAKEEGVSLDKR, 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/MFoc-1 hybrid signal sequence (also known as HSA/kex2) (e.g., MKWVSFISLLFLFSSAYSRSLDKR, SEQ ID NO:14) j) a K lactis killer/ MFcc-1 fusion leader sequence (e.g., MNIFYIFLFLLSFVQGSLDKR, SEQ ID NO:15) k) the Immunoglobulin Ig signal sequence (e.g., MGWSCIILFLVATATGVHS, SEQ
ID NO:16) 1) the Fibulin B precursor signal sequence (e.g., MERAAPSRRVPLPLLLLGGLALLAAGVDA, SEQ ID NO:17) m) the clusterin precursor signal sequence (e.g., MMKTLLLFVGLLLTWESGQVLG, SEQ ID NO:18) n) the insulin-like growth factor-binding protein 4 signal sequence (e.g., MLPLCLVAALLLAAGPGPSLG, SEQ ID NO:19) o) variants of the pre-pro-region of the HSA signal sequence such as, for example, MKWVSFISLLFLFSSAYSRGVFRR (SEQ ID NO :20), MKWVTFISLLFLFAGVLG (SEQ ID NO:21), MKWVTFISLLFLFSGVLG (SEQ ID NO:22), MKWVTFISLLFLFGGVLG (SEQ ID NO:23), Modified HSA leader HSA #64 MKWVTFISLLFLFAGVSG (SEQ ID NO:24);
Modified HSA leader HSA #66 MKWVTFISLLFLFGGVSG (SEQ ID NO:25);
Modified HSA (A14) leader ¨
MKWVTFISLLFLFAGVSG (SEQ ID NO:26);
Modified HSA (S14) 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
NO :30) q) acid phosphatase (PH05) leader (e.g., MFKSVVYSILAASLANA SEQ ID NO: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) S. diastaticus glucoarnylase II secretion leader sequence x) S. carlsbergensis ce-galactosidase (MEL1) secretion leader sequence y) Candida glucoarnylase leader sequence z) The hybrid leaders disclosed in EP-A-387 319 an) 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 );or dd) Inulinase ¨ MICLAYSLLLPLAGVSASVINYKR (SEQ ID NO:33).
ee) A modified TA57 propeptide leader variant #1 ¨
MICLKTVRSAVLSSLFASQVLGQPIDDTESQTTSVNLMADDTESAFATQTNSGG
LDVVGLISMAKR (SEQ ID NO:34) ff) A modified TA57 propeptide leader variant #2 ¨
MICLKTVRSAVLSSLFASQVLGQPIDDTESQTTSVNLMADDTESAFATQTNSGG
LDVVGLISMAEEGEPKR (SEQ ID NO:35) gg) A consensus signal peptide ¨
MWWRLWWLLLLLLLLWPMVWA (SEQ ID NO:550) [0325]
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 (Cl-JO) 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: W087/04462;

=
=
W086/05807; W089/01036; W089/10404; and W091/06657.
Additionally, glutamine synthase expression vectors can be obtained from Lonza Biologics, 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), [0326] 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. Furthermore, 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.
[0327] Introduction of the 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 (1986). It is specifically contemplated that the polypeptides of the present invention may in fact be expressed by a host cell lacking a recombinant vector.
In addition to encompassing host cells containing the vector constructs discussed herein, 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.
103281 In addition, techniques known in the art may be used to operably associate heterologous polynucleotides (e.g., polynucleotides encoding an albumin protein, or a fragment or variant thereof) and/or heterologous control regions (e.g., promoter and/or enhancer) with 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. Sci. USA 86:8932-8935 (1989); and Zijlstra et al., Nature 342:435-438 (1989)).
[0329] 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.
[0330] In preferred embodiments 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.
[0331] In specific embodiments 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 columns and their equivalents and comparables.
[03321 In specific embodiments 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.
[0333] In specific embodiments the albumin fusion proteins of the invention are purified using Size Exclusion Chromatography including, but not limited to, sepharose S100, S200, S300, superdex resin columns and their equivalents and comparables.
[0334] In specific embodiments the 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.
[0335] In preferred embodiments albumin fusion proteins of the invention are purified using one or more Chromatography methods listed above. In other preferred embodiments, 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.
[0336] Additionally, albumin fusion proteins of the invention may be purified using the process described in PCT International Publication WO 00/44772.
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.
[0337] 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.
Thus, it is well known in the art that the 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.
[0338] In one embodiment, the yeast Pichia pastoris 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 02.
This reaction is catalyzed by the enzyme alcohol oxidase. 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 02. Consequently, in a growth medium depending on methanol as a main carbon source, the promoter region of one of the two alcohol oxidase genes (A0X1) is highly active. In the presence of methanol, alcohol oxidase produced from the A0X1 gene comprises up to approximately 30% of the total soluble protein in Pichia pastoris. See Ellis, S.B., et al., Mol. Cell. Biol. 5:1111-21(1985); Koutz, P.J, et al., Yeast 5:167-77 (1989); Tschopp, J.F., et al., NucL Acids Res.
15:3859-76 (1987).
Thus, a heterologous coding sequence, such as, for example, a polynucleotide of the present invention, under the transcriptional regulation of all or part of the A0X1 regulatory sequence is expressed at exceptionally high levels in Pichia yeast grown in the presence of methanol.
[0339] In one example, 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 A0X1 promoter linked to the Pichia pastoris alkaline phosphatase (PHO) secretory signal peptide (i.e., leader) located upstream of a multiple cloning site.
[0340] Many other yeast vectors could be used in place of pPIC9K, such as, pYES2, pYD1, pTEF1/Zeo, pYES2/GS, pPICZ, pGAPZ, pGAPZalpha, pPIC9, pPIC3.5, pHIL-D2, pHIL-S1, pPIC3.5K, and PA0815, 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.
[0341] In another embodiment, 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.
[0342] In addition, 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)). For example, a polypeptide corresponding to a fragment of a polypeptide can be synthesized by use of a peptide synthesizer. Furthermore, if desired, 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 acid, 4-aminobutyric acid, Abu, 2-amino butyric 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, phenylglycine, cyclohexylalanine, b-alanine, fluoro-amino acids, designer amino acids such as b-methyl amino acids, Ca-methyl amino acids, Na-methyl amino acids, and amino acid analogs in general. Furthermore, the amino acid can be D
(dextrorotary) or L
(levorotary).
[0343] 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, NaBILI; acetylation, formylation, oxidation, reduction; metabolic synthesis in the presence of tunicamycin; etc.
[0344] Additional post-translational modifications encompassed by the invention include, for example, e.g., N-linked or 0-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 0-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.
[0345] Examples of 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 (121/, 1231, 1251, 131vs, 1 3min, 112-n, iodine .1) carbon (I4C), sulfur (35S), tritium (3H), indium (II 'In, 115mIn), technetium (99Tc,99"7c), thallium (20ITi), gallium ("Ga, "Ga), palladium (1 3Pd), molybdenum (99Mo), xenon (I33Xe), fluorine (I8F), I53Sm, I77Lu, I"Gd, 149Pm, I40La, I75Yb, 1661/0, 90y, 47se, 186Re, 188Re, , 142-r P '"Rn, and 97Rn.
[0346]
In specific embodiments, albumin fusion proteins of the present invention or fragments or variants thereof are attached to macrocyclic chelators that associate with radiometal ions, including but not limited to, 177Lu, 90Y, I66Ho, and 153Sm, to polypeptides. In a preferred embodiment, the radiometal ion associated with the macrocyclic chelators is IIIIn.
In another preferred embodiment, the radiometal ion associated with the macrocyclic chelator is 90Y.
In specific embodiments, the macrocyclic chelator is 1,4,7,10-tetraazacyclododecane-N,N',N",Nm-tetraacetic acid (DOTA). In other specific embodiments, DOTA is attached to an antibody of the invention or fragment thereof via linker molecule.
Examples of 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. 26(8):943-50 (1999).
[0347]
As mentioned, the 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. (See, for instance, PROTEINS - STRUCTURE AND MOLECULAR
PROPERTIES, 2nd Ed., T. E. Creighton, W. H. Freeman and Company, New York (1993);
POST-TRANSLATIONAL COVALENT MODIFICATION OF PROTEINS, B. C. Johnson, Ed., Academic Press, New York, pgs. 1-12 (1983); Seifter et al., Meth.
Enzymol. 182:626-646 (1990); Rattan et al., Ann. N.Y. Acad. Sci. 663:48-62 (1992)).
[0348] 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. In preferred embodiments, 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. As described in Gentz et al., Proc. Natl. Acad. Sci.
USA 86:821-824 (1989), for instance, hexa-histidine provides for convenient purification of the fusion protein. Other peptide tags useful for purification include, but are not limited to, 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.
[0349] Further, 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 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, 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., vincristine and vinblastine).
[0350] 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. For example, 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, B-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 II (See, International Publication No.
WO 97/34911), Fas Ligand (Takahashi et al., mt. Immunot, 6:1567-1574 (1994)), VEGI
(See, International Publication No. WO 99/23105), a thrombotic agent or an anti- angiogenic agent, e.g., angiostatin or endostatin; or, biological response modifiers such as, for example, lymphokines, interleukin-1 ("IL-1"), 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. Techniques for conjugating such therapeutic moiety to proteins (e.g., albumin fusion proteins) are well known in the art.
[0351] 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. Such solid supports include, but are not limited to, glass, cellulose, polyacrylamide, nylon, polystyrene, polyvinyl chloride or polypropylene.
[0352] 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.
[0353] In embodiments where the albumin fusion protein of the invention comprises only the VH domain of an antibody that binds a Therapeutic protein, it may be necessary and/or desirable to coexpress the fusion protein with the VL domain of the same antibody that binds a Therapeutic protein, such that the VH-albumin fusion protein and VL
protein will associate (either covalently or non-covalently) post-translationally.
[0354] In embodiments where the albumin fusion protein of the invention comprises only the VL domain of an antibody that binds a Therapeutic protein, it may be necessary and/or desirable to coexpress the fusion protein with the VH domain of the same antibody that binds a Therapeutic protein, such that the VL-albumin fusion protein and VH protein will associate (either covalently or non-covalently) post-translationally.
[0355] Some Therapeutic antibodies are bispecific antibodies, meaning the antibody that binds a Therapeutic protein is an artificial hybrid antibody having two different heavy/light chain pairs and two different binding sites. In 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. More particularly, 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/light (VHNL) pair of the original antibody that binds a Therapeutic protein.
[0356] Also provided by the invention are chemically modified derivatives of the albumin fusion proteins of the invention which may provide additional advantages such as increased solubility, stability and circulating time of the polypeptide, or decreased immunogenicity (see U.S. Patent No. 4,179,337). 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.
[0357] The polymer may be of any molecular weight, and may be branched or unbranched. For polyethylene glycol, the preferred molecular weight is between about 11cDa 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). For example, 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 IcDa.
[0358] As noted above, the polyethylene glycol may have a branched structure.
Branched polyethylene glycols are described, for example, in U.S. Patent No.
5,643,575;

Morpurgo et aL, AppL Biochem. Biotechnol. 56:59-72 (1996); Vorobjev et al., Nucleosides Nucleotides /8:2745-2750 (1999); and Caliceti et aL, Bioconjug. Chem. /0:638-646 (1999).
[0359] The polyethylene glycol molecules (or other chemical moieties) should be attached to the protein with consideration of effects on functional or antigenic domains of the protein. There are a number of 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), see also Malik et al., Exp. Hematol. 20:1028-1035 (1992), reporting pegylation of GM-CSF using tresyl chloride. For example, 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.
[0360] As suggested above, polyethylene glycol may be attached to proteins via linkage to any of a number of amino acid residues. For example, 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.
[0361] One may specifically desire proteins chemically modified at the N-terminus.
Using 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) may be by purification of the N-terminally pegylated material from a population of pegylated protein molecules. Selective proteins chemically modified at the N-terminus modification may be accomplished by reductive allcylation 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.
[0362] As indicated above, pegylation of the albumin fusion proteins of the invention may be accomplished by any number of means. For example, 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. J. of Hematol. 68:1-18 (1998); U.S. Patent No. 4,002,531; U.S. Patent No. 5,349,052; WO 95/06058; and WO 98/32466.
[03631 One system for attaching polyethylene glycol directly to amino acid residues of proteins without an intervening linker employs tresylated MPEG, which is produced by the modification of monmethoxy polyethylene glycol (MPEG) using tresylchloride (C1S02CH2CF3). Upon reaction of protein with tresylated MPEG, polyethylene glycol is directly attached to amine groups of the protein. Thus, 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. For example, 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 1,1 '-carbonyldiimidazole, MPEG-2,4,5-trichloropenylcarbonate, MPEG-p-nitrophenolcarbonate, and various MPEG-succinate derivatives. A number of additional polyethylene glycol derivatives and reaction chemistries for attaching polyethylene glycol to proteins are described in International Publication No. WO 98/32466.
Pegylated protein products produced using the reaction chemistries set out herein are included within the scope of the invention.
[0365] The number of polyethylene glycol moieties attached to each albumin fusion protein of the invention (i.e., the degree of substitution) may also vary. For example, 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. Similarly, 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).
[0366] The 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 chromatography and lectin chromatography. Most preferably, high performance liquid chromatography ("HPLC") is employed for purification. Well known techniques for refolding protein may be employed to regenerate active conformation when the polypeptide is denatured during isolation and/or purification.
[0367] The presence and quantity of albumin fusion proteins of the invention may be determined using ELISA, a well known immunoassay known in the art. In one 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. In an alternate version of this protocol, 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.
Uses of the Polynucleotides [0368] Each of the polynucleotides identified herein can be used in numerous ways as reagents. The following description should be considered exemplary and utilizes known techniques.
[0369] The polynucleotides of the present invention are useful to produce the albumin fusion proteins of the invention. As described in more detail below, polynucleotides of the invention (encoding albumin fusion proteins) may be used in recombinant DNA
methods useful in genetic engineering to make cells, cell lines, or tissues that express the albumin fusion protein encoded by the polynucleotides encoding albumin fusion proteins of the invention.
[0370]
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).
Uses of the Polyp eptides [0371]
Each of the polypeptides identified herein can be used in numerous ways. The following description should be considered exemplary and utilizes known techniques.
[0372]
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.
29:577-580 (1981)) or cell type(s) (e.g., immunocytochemistry assays).
[0373]
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, etal., J. Cell. Biol. 101:976-985 (1985); Jalkanen, etal., 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 are known in the art and include enzyme labels, such as, glucose oxidase; radioisotopes, such as iodine (131/, 125/, 123,I , 121J) carbon ('4C), sulfur (35S), tritium (3H), indium (1 ismin, 113min, 1 'In), I
In), and technetium (99Tc, .
99mTc) (68Ga, 67Ga) , , thallium (2 1Ti), gallium palladium (103Pd), molybdenum (99Mo), xenon (133Xe), fluorine (18F), 1535m, 177Lu, 159Gd, 149pm, 140La, 175y-b, 166H0, 90y, 47sc, 186Re, I88Re, 142pr, 1057,1 , K/1 97RU; luminescent labels, such as luminol; and fluorescent labels, such as fluorescein and rhodamine, and biotin.
[0374]
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). For X-radiography, 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.
[0375] An albumin fusion protein which has been labeled with an appropriate detectable imaging moiety, such as a radioisotope (for example, 1311,112in, 99mTC, (1311 1251 121%
1211), carbon (14C), sulfur (355), tritium (3H), indium (115mIn, 3mm, 112/n, 'D

I in), and technetium (99Tc, 99mTc), thallium (201Ti), gallium (68Ga5 67Ga)5 palladium (1 3Pd), molybdenum (99Mo), xenon (133Xe), fluorine (18F, 153sm, '77L u, 159Gd, 149pm, 140La, 175yb, 166/40, 90y, 47sc, 186Re, 188Re, 14 105 105-,-. 5 97 Kn Ru), 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 immune system disorder. It will be understood in the art that the size of the subject and the imaging system used will determine the quantity of imaging moiety needed to produce diagnostic images. In the case of a radioisotope moiety, for a human subject, the quantity of radioactivity injected will normally range from about 5 to 20 millicuries of 99mTc. 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. Alternatively, in the case where the albumin fusion protein comprises at least a fragment or variant of a Therapeutic antibody, the labeled albumin fusion protein will then preferentially accumulate at the locations in the body (e.g., organs, cells, extracellular spaces or matrices) where the 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. 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)). The protocols described therein could easily be modified by one of skill in the art for use with the albumin fusion proteins of the invention.
[0376] In one embodiment, 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. In one example, the invention provides a method for delivering a Therapeutic protein into the targeted cell. In another example, 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.
[0377] In another embodiment, 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.
[0378] By "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. "Toxin" also includes a cytostatic or cytocidal agent, a therapeutic agent or a radioactive metal ion, e.g., alpha-emitters such as, for example, 213Bi, or other radioisotopes such as, for example, 1 3Pd, 133Xe, 131j, 68Ge, 57co, 65zn, 85sr, 32p, 35s, 90y, 153sm, 153Gd, 113sn, 90yttrium, 117Tin, 186Rhenium, 166 169¨Y , b 51Cr, 54Mn, 75Se, Holmium, and 188Rhenium;
luminescent labels, such as luminol; and fluorescent labels, such as fluorescein and rhodamine, and biotin. In a specific embodiment, 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 90Y. In another specific embodiment, 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 111In. In a further specific embodiment, 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 1311.
[0379] Techniques known in the art may be applied to lable polypeptides of the invention. Such techniques include, but are not limited to, the use of bifunctional conjugating agents (see e.g., U.S. Patent Nos. 5,756,065; 5,714,631; 5,696,239; 5,652,361;
5,505,931;
5,489,425; 5,435,990; 5,428,139; 5,342,604; 5,274;119; 4,994,560; and 5,808,003).
[0380] The albumin fusion proteins of the present invention are useful for diagnosis, treatment, prevention and/or prognosis of various disorders in mammals, preferably humans.
Such disorders include, but are not limited to, those described herein under the section heading "Biological Activities," below.
[0381] Thus, 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. With respect to cancer, 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.
[0382] Moreover, 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. For example, 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).
[0383] In particular, 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). For example, 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 protein specifically binds, and/or reduce overproduction of the polypeptide to which the Therapeutic antibody used to make the albumin fusion protein specifically binds.
Similarly, administration of 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).
[0384] At the very least, 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. Moreover, the albumin fusion proteins of the present invention can be used to test the biological activities described herein.
Diagnostic Assays [0385] The compounds of the present invention are useful for diagnosis, treatment, prevention and/or prognosis of various disorders in mammals, preferably humans. Such 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," infra.
[0386] For a number of disorders, 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. Thus, the invention provides 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. These diagnostic assays may be performed in vivo or in vitro, such as, for example, on blood samples, biopsy tissue or autopsy tissue.
[0387] The present invention is also useful as a prognostic indicator, whereby patients exhibiting enhanced or depressed gene expression will experience a worse clinical outcome [0388] By "assaying 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). Preferably, 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. As will be appreciated in the art, once a standard polypeptide level or mRNA level is known, it can be used repeatedly as a standard for comparison.
[0389] By "biological sample" is intended any biological sample obtained from an individual, cell line, tissue culture, or other source containing polypeptides of the invention (including portions thereof) or mRNA. As indicated, 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.
[0390] 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, S 1 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).
[0391]
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. Thus, for instance, 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.
[0392]
Assaying polypeptide levels in a biological sample can occur using a variety of techniques. For example, polypeptide expression in tissues can be studied with classical immunohistological methods (Jalkanen et al., J. Cell. Biol. 101:976-985 (1985); Jalkanen, M., et al., J. Cell . Biol. 105:3087-3096 (1987)). Other methods useful for detecting polypeptide gene expression include immunoassays, such as the enzyme linked immunosorbent assay (ELISA) and the radioimmunoassay (RIA). Suitable antibody assay labels are known in the art and include enzyme labels, such as, glucose oxidase, and , ", , radioisotopes, such as iodine (1251 121 1) carbon (14C), sulfur (35S), tritium (3H), indium (1 '21n), and technetium (99mTc), and fluorescent labels, such as fluorescein and rhodamine, and biotin.
[0393]
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).
The isolated cells can be derived from 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.
[0394] For example, 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.
[0395] In a preferred embodiment, 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.
[0396] 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.
Through the use of such a procedure, it is possible to determine not only the presence of the polypeptides that bind to, are bound by, or associate with albumin fusion proteins, but also its distribution in the examined tissue. Using the present invention, those of ordinary skill will readily perceive that any of a wide variety of histological methods (such as staining procedures) can be modified in order to achieve such in situ detection.
[0397] Immunoassays and non-immunoassays that detect polypeptides that bind to, are bound by, or associate with albumin fusion proteins will 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.

[0398] The biological sample may be brought in 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. 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. Optionally the antibody is subsequently labeled.
The amount of bound label on solid support may then be detected by conventional means.
[0399] By "solid phase support or carrier" is intended 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.) 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. Thus, 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. Alternatively, the surface may be flat such as a sheet, test strip, etc.
Preferred supports include polystyrene beads. Those skilled in the art will know many other suitable carriers for binding antibody or antigen, or will be able to ascertain the same by use of routine experimentation.
[0400] The binding activity of a given lot of albumin fusion protein may be determined according to well known methods. Those skilled in the art will be able to determine operative and optimal assay conditions for each determination by employing routine experimentation.
[0401] In addition to assaying polypeptide levels in a biological sample obtained from an individual, polypeptide can also be detected in vivo by imaging. For example, in one embodiment of the invention, albumin fusion proteins of the invention are used to image diseased or neoplastic cells.
[0402] 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. For X-radiography, 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 of a cell line (or bacterial or yeast strain) engineered.
[0403] Additionally, albumin fusion proteins of the invention whose presence can be detected, can be administered. For example, 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.
[0404] A polypeptide-specific antibody or antibody fragment which has been labeled with an appropriate detectable imaging moiety, such as a radioisotope (for example, 1311, 112In, 99mTc), 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. It will be understood in the art that the size of the subject and the imaging system used will determine the quantity of imaging moiety needed to produce diagnostic images. In the case of a radioisotope moiety, for a human subject, the quantity of radioactivity injected will normally range from about 5 to 20 millicuries of 99mTc. 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)).
[0405] One of the ways in which 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., .1 Clin. Pathol. 31:507-520 (1978); Butler, J.E., Meth. Enzymol. 73:482-523 (1981); Maggio, E. (ed.), 1980, Enzyme Immunoassay, CRC
Press, Boca Raton, FL,; Ishikawa, E. et al., (eds.), 1981, Enzyme Immunoassay, Kgaku Shoin, Tokyo). 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.
10406] Albumin fusion proteins may also be radiolabelled and used in any of a variety of other immunoassays. For example, by radioactively labeling the albumin fusion proteins, it is possible to the use the albumin fusion proteins in a radioinununoassay (MA) (see, for example, Weintraub, B., Principles of Radioitnmunoassays, Seventh Training Course on Radioligand Assay Techniques, The Endocrine Society, March, 1986).
The radioactive isotope can be detected by means including, but not limited to, a gamma counter, a scintillation counter, or autoradiography.
[04071 Additionally, 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. For example, see Subramanian, R. and Meares, C.F., "Bifunctional Chelating Agents for Radiometal-labeled monoclonal Antibodies," in Cancer Imaging with Radiolabeled Antibodies (D. M. Goldenberg, Ed.) Kluwer Academic Publications, Boston;
Saji, H., "Targeted delivery of radiolabeled imaging and therapeutic agents:
bifunctional radiopharmaceuticals." Crit. Rev. Ther. Drug Carrier Syst. 16:209-244 (1999);
Srivastava S.C. and Mease R.C., "Progress in research on ligands, nuclides and techniques for labeling monoclonal antibodies." Int. .1. Rad App!. Instrum. B /8:589-603 (1991); and Liu, S. and Edwards, D.S., "Bifunctional chelators for therapeutic lanthanide radiopharmaceuticals."
Bioconjug. Chem. /2:7-34 (2001). Any 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.
[04081 In one embodiment, 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. As non-limiting examples, the chelator may be 2-(p-isothiocyanatobenzy1)-6- methyldiethylenetriaminepentaacetic acid (1B4M-DPTA, also known as MX-DTPA), 2-methyl-6-(rho-nitrobenzy1)-1,4,7- triazaheptane-N,N,M,N",N"-pentaacetic acid (nitro-1B4M-DTPA or nitro-MX-DTPA); 2-(p-isothiocyanatobenzy1)-cyclohexyldiethylenetriaminepentaacetic acid (CHX-DTPA), or N42-amino-3-(rho-nitrophenyl)propyli-trans-cyclohexane-1,2-diamine-N,N,N"-pentaacetic acid (nitro-CHX-A-DTPA).
[0409] In another embodiment, the Albumin fusion protein of the invention are attached to an acyclic terpyridine chelator such as 6,6"-bisP,N,N",N"-tetra(carboxymethyDaminolmethyll-4'-(3-amino-4-methoxypheny1)-2,2':6',2 "-terpyridine (TMT-amine).
[0410] In specific embodiments, the macrocyclic chelator which is attached to the the Albumin fusion protein of the invention is= 1,4,7,10-tetraazacyclododecane-N,N',N",Nm-tetraacetic acid (DOTA). In other specific embodiments, the DOTA is attached to the the Albumin fusion protein of the invention via a linker molecule. Examples of 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 Bioconjug.
Chem. /0(4):553-7, 1999; and Zimmerman et NucL Med. Biol. 26(8):943-50, 1999.
In addition, U.S. Patents 5,652,361 and 5,756,065, which disclose chelating agents that may be conjugated to antibodies, and methods for making and using them, are hereby incorporated by reference in their entireties. Though U.S. Patents 5,652,361 and 5,756,065 focus on conjugating chelating agents to antibodies, one skilled in the art could readily adapt the method disclosed therein in order to conjugate chelating agents to other polypeptides.
[0411] 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-nitrobenzy1-1,4,7,10-tetraazacyclododecane-N,N',N",N'"-tetraacetic acid); S.
V. Deshpande et al., J. NucL Med. 3/:473 (1990); G. Ruser et al., Bioconj. Chem. /:345 (1990);
C. J. Broan et al., J. C. S. Chem. Comm. 23:1739 (1990); and C. J. Anderson et al., NucL Med.
36:850 (1995).
[0412] In one embodiment, 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.
In another embodiment, the chelator is a chelator selected from the group consisting of DOTA, analogues of DOTA, and derivatives of DOTA.
[0413] In one embodiment, suitable chelator molecules that may be attached to the the Albumin fusion protein of the invention include DMA (1-oxa-4,7,10-triazacyclododecanetriacetic acid), NOTA (1,4,7-triazacyclononanetriacetic acid), TETA
(1,4,8,11-tetraazacyclotetradecanetetraacetic acid), and THT (4'-(3-amino-4-methoxy-pheny1)-6,6"-bis(N',N'-dicarboxymethyl-N-methylhydra zino)-2,2': 6',2 " -terpyridine), and analogs and derivatives thereof. See, e.g., Ohmono et al., J. Med. Chem. 35:

(1992); Kung et al., J. Nucl. Med 25: 326-332 (1984); Jurisson et al., Chem.
Rev. 93:1137-1156 (1993); and U.S. Patent No. 5,367,080. Other suitable chelators include chelating agents disclosed in U.S. Patent Nos. 4,647,447; 4,687,659; 4,885,363; EP-A-71564;
W089/00557; and EP-A-232751.
[0414] In another embodiment, suitable macrocyclic carboxylic acid chelators which can be used in the present invention include 1,4,7,10-tetraazacyclododecane-N,N,N",N"-tetraacetic acid (DOTA); 1,4,8,12-tetraa7acyclopentadecane-N,N,N"N"-tetraacetic acid (15N4); 1,4,7-triazacyclononane-N,N',N"-triacetic acid (9N3); 1,5,9-triazacyclododecane-N,N,N"-triacetic acid (12N3); and 6-bromoacetamido-benzy1-1,4,8,11-tetraa7acyclotetradecane- N,N1,N",N"-tetraacetic acid (BAT).
[0415] A
preferred chelator that can be attached to the Albumin Fusion protein of the invention is a-(5 -isothiocyanato- 2-methoxypheny1)-1,4,7,10-tetra a 7acyclo dodecane-1,4,7,10-tetraacetic acid, which is also known as Me0-DOTA-NCS. A salt or ester of a-(5-isothiocyanato- 2-methoxypheny1)- 1,4,7,10-tetra a 7acyclododecane-1,4,7,10-tetraacetic acid may also be used.
[0416]
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. Examples of appropriate metals include Ag, At, Au, Bi, Cu, Ga, Ho, In, Lu, Pb, Pd, Pm, Pr, Rb, Re, Rh, Sc, Sr, Tc, Tl, Y, and Yb. Examples of the radionuclide used for diagnostic purposes are Fe, Gd, 111In, 67Ga, or 68Ga. In another embodiment, the radionuclide used for diagnostic purposes is "In, or 67Ga. Examples of the radionuclide used for therapeutic purposes are 1661/0, 165Dy, 90y, 115m1n, 52Fe, or 72 Ga. In one embodiment, the radionuclide used for diagnostic purposes is 166Ho or 90Y. Examples of the radionuclides used for both therapeutic and diagnostic purposes include I"Sm, I77Lu, I59Gd, 175Yb, or 47Se.
In one embodiment, the radionuclide is 153sm, 177-Lu, I75Yb, or 159Gd.

[0417] Preferred metal radionuclides include 90y, 99mTe, 47se, 67-a, 51Cr, 177mSn, 67Cu, 167TM, 97Ru, 188Re, 177Lu, 199Au, 47Sc, 67Ga, 51Cr, 177mSn, 67Cu, 167Tm, 95Ru, 188-e, 1771,U, 199AU, 203Pb and 141ce, [0418] In a particular embodiment, Albumin fusion proteins of the invention to which chelators are covalently attached may be labeled with a metal ion selected from the group 177Ln, 166H0, 215Bi, and 225Ac.
consisting of 90Y, 111In, [0419] Moreover, y-emitting radionuclides, such as 99mTc, 67Ga, and 169Yb have been approved or under investigation for diagnostic imaging, while 13-emitters, such as 67Cu, niAg, 186Re, and 90Y are useful for the applications in tumor therapy. Also other useful radionuclides include 7-emitters, such as 99mi,c, 67Ga, and Y to and 13-emitters, such as 67Cu, IllAg,1867,e, R8 x 1-Re and 90Y, as well as other radionuclides of interest such as 211At, 212Bi, 177Ln, 86Rb 105Rh, 1-m 53 , S 198AU, 149PM, 85Sr, 142pr, 214pb, 109pd,166,m, H
208T1, and 44Sc.
Albumin fusion proteins of the invention to which chelators are covalently attached may be labeled with the radionuclides described above.
[0420] In another embodiment, 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.
[0421] In another embodiment, 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., 152Eu), Gd, Tb, Dy, Ho, Er, Tm, Yb, and Lu.
[0422] In another embodiment, 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.
[0423] It is also possible to label the albumin fusion proteins with a fluorescent compound. When the fluorescently labeled antibody is exposed to light of the proper wave length, its presence can then be detected due to fluorescence. Among the most commonly used fluorescent labeling compounds are fluorescein isothiocyanate, rhodamine, phycoerythrin, phycocyanin, allophycocyanin, ophthaldehyde and fluorescamine.
[0424] The albumin fusion protein can also be detectably labeled using fluorescence emitting metals such as 152Eu, or others of the lanthanide series. These metals can be attached to the antibody using such metal chelating groups as diethylenetriaminepentacetic acid (DTPA) or ethylenediaminetetraacetic acid (EDTA).
[0425] 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. Examples of particularly useful chemiluminescent labeling compounds are luminol, isoluminol, theromatic acridinium ester, imidazole, acridinium salt and oxalate ester.
[0426] Likewise, 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 [0427] 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.

[0428] 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.
[0429] 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, electroporation, microinjection, gene targeting in embryonic stem cells and recombinant viral and retroviral infection (see, e.g., U.S. Patent No. 4,736,866; 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.
[0430] 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-1) (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.
5,720,936); have a reduced capacity to mediate cellular adhesion (U.S. Patent No. 5,602,307);
possess a bovine growth hormone gene (Clutter et al. (1996) Genetics 143(4):1753-1760); or, are capable of generating a fully human antibody response (McCarthy (1997) The Lancet 349(9049):405).
[0431] While 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) Mol. 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).
[0432] To direct the secretion of the transgene-encoded protein of the invention into the milk of transgenic mammals, it 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 Soutier et al. (1992) FEBS Letts. 297:13). The transgenic mammals of choice would produce large volumes of milk and have long lactating periods, for example goats, cows, camels or sheep.
[0433] 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. 153 ("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, 482,852, and European Patent Application EP 693 554, Pharmaceutical or Therapeutic Compositions [0434] 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.
[0435] 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.
Typically, 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. For instance, pharmaceutical 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.
[0436] For example, 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.
[0437] In instances where aerosol administration is appropriate, the albumin fusion proteins of the invention can be formulated as aerosols using standard procedures. The term "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.
Specifically, 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. See Ganderton & Jones, Drug Delivery to the Respiratory Tract, Ellis Horwood (19 87); Gonda (1990) Critical Reviews in Therapeutic Drug Carrier Systems 6:273-313; and Raeburn et al,. (1992) Pharmacol. Toxicol. Methods 27:143-159.
[0438] 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. In some cases, wherein either component is non human-derived, that component may be humanized by substitution of key amino acids so that specific epitopes appear to the human immune system to be human in nature rather than foreign.
[0439] The formulations 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.

[0440] 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.
[0441] As an example, when 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 RI/kg/week, or 0.9 to 12.0 ILI/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.
[0442] 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. For instance, such instructions or package inserts may address recommended storage conditions, such 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. As described above, formulations of the invention may be in aqueous form and may be stored under less than ideal circumstances without significant loss of therapeutic activity.

[0443] Albumin fusion proteins of the invention can also be included in nutraceuticals. For instance, 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.
[0444] 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.
[0445] 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.
[0446] As a general proposition, 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.
If given continuously, 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.
[0447] 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. The term "parenteral" as used herein refers to modes of administration which include intravenous, intramuscular, intraperitoneal, intrastemal, subcutaneous and intraarticular injection and infusion.
[0448] Albumin fusion proteins and/or polynucleotides of the invention are also suitably administered by sustained-release systems. Examples of 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. The term "parenteral" as used herein refers to modes of administration which include intravenous, intramuscular, intraperitoneal, intrastemal, subcutaneous and intraarticular injection and infusion.
Additional examples of 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).
[0449] 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).
[0450] 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 Therapy 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. (USA) 82:3688-3692 (1985); Hwang et al., Proc. Natl. Acad.
Sci.(USA) 77:4030-4034 (1980); EP 52,322; EP 36,676; EP 88,046; EP 143,949; EP 142,641;
Japanese Pat. Appl. 83-118008; U.S. Pat. Nos. 4,485,045 and 4,544,545; and EP 102,324.
Ordinarily, 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.
[0451] In yet an additional embodiment, 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)).
[0452] Other controlled release systems are discussed in the review by Langer (Science 249:1527-1533 (1990)).
[0453] For parenteral administration, in one embodiment, 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. For example, the formulation preferably does not include oxidizing agents and other compounds that are known to be deleterious to the Therapeutic.
[0454] Generally, 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. Then, if necessary, the product is shaped into the desired formulation.
Preferably the carrier is a parenteral carrier, more preferably a solution that is isotonic with the blood of the recipient. Examples of such 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.
[0455] The carrier suitably contains minor amounts of 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 irnmunoglobulins;
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 sorbitol; counterions such as sodium;
and/or nonionic surfactants such as polysorbates, poloxamers, or PEG.
[0456] The albumin fusion protein is typically formulated in such vehicles at a concentration of about 0.1 mg/ml to 100 mg/ml, preferably 1-10 mg/ml, at a pH
of about 3 to 8. It will be understood that the use of certain of the foregoing excipients, carriers, or stabilizers will result in the formation of polypeptide salts.
[0457] 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.
[0458] 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. As an example of a lyophilized formulation, 10-ml vials are filled with 5 ml of sterile-filtered 1% (w/v) aqueous albumin fusion protein and/or polynucleotide solution, and the resulting mixture is lyophilized. The infusion solution is prepared by reconstituting the lyophilized albumin fusion protein and/or polynucleotide using bacteriostatic Water-for-Injection.
[0459] In a specific and preferred embodiment, the Albumin fusion protein formulations comprises 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. In another specific and preferred embodiment, 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 tonicifier. Sodium octanoate has been chosen due to its reported ability to increase the thermal stability of the protein in solution. Finally, 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.
[0460] 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. Associated with such container(s) 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. In addition, the albumin fusion proteins and/or polynucleotides may be employed in conjunction with other therapeutic compounds.

[0461] The 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.), (Genentech, Inc.), BCG (e.g., THERACYSO), MPL and nonviable preparations of Corynebacterium parvum. In a specific embodiment, albumin fusion proteins and/or polynucleotides of the invention are administered in combination with alum. In another specific embodiment, 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, Monophosphoryl lipid immunomodulator, AdjuVax 100a, QS-21, QS-18, CRL1005, Aluminum salts, MF-59, 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.
Combinations may be administered either concomitantly, e.g., as an admixture, separately but simultaneously or concurrently; or sequentially. This includes presentations in 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.
[0462] The 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. This includes presentations in 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.
[0463] In one embodiment, 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., COUMADINC), dicumarol, 4-hydroxycoumarin, anisindione (e.g., MIRADONTm), acenocoumarol (e.g., nicoumalone, SINTHROMETm), indan-1,3-dione, phenprocoumon (e.g., MARCUMARTm), ethyl biscoumacetate (e.g., TROMEXANTm), and aspirin. In a specific embodiment, 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 embodiment, compositions of the 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 administered in combination with heparin and aspirin.
[0464] In another embodiment, 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., KABIKINASETm), antiresplace (e.g., EMINASETm), tissue plasminogen activator (t-PA, altevase, ACTIVASETm), urokinase (e.g., ABBOKINASETm), sauruplase, (Prourokinase, single chain urokinase), and aminocaproic acid (e.g., AMICARTm). In a specific embodiment, compositions of the invention are administered in combination with tissue plasminogen activator and aspirin.
[0465] In another embodiment, the 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).
[0466] In specific embodiments, 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. In specific embodiments, 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. Other uses for the therapeutics of the invention, alone or in combination with antiplatelet, anticoagulant, and/or thrombolytic drugs, include, but are not limited to, the prevention of occlusions in extracorporeal devices (e.g., intravascular canulas, vascular access shunts in hemodialysis patients, hemodialysis machines, and cardiopulmonary bypass machines).
[0467] In certain embodiments, 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 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/ddI), HIVIDTM
(zalcitabine/ddC), ZERITTm (stavudine/d4T), EPIVIRTM (lamivudine/3TC), and 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 that may be administered in combination with the albumin fusion proteins and/or polynucleotides of the invention, include, but are not limited to, CRIXIVANTM
(indinavir), NORVIRTM (ritonavir), INVIRASETM (saquinavir), and VIRACEPTTm (nelfinavir).
In a specific embodiment, 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.
[0468] Additional NRTIs include LODENOS1NETM (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); PREVEONO (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/Abbott);
D-D4FC (related to 3TC, with activity against AZT/3TC-resistant virus);
GW420867X (Glaxo Wellcome); ZJAGENTM (abacavir/159U89; Glaxo Wellcome Inc.); CS-87 (3'azido-2',3'-dideoxyuridine; WO 99/66936); and S-acy1-2-thioethyl (SATE)-bearing prodrug forms of 13-L-FD4C and 13-L-FddC (WO 98/17281).
[0469] Additional NNRTIs include COACTINONTm (Emivirine/MKC-442, potent NNRTI of the HEPT class; Triangle/Abbott); CAPRAVIR1NETM (AG-1549/S-1153, a next generation NNRTI with activity against viruses containing the K103N mutation;
Agouron);
PNU-142721 (has 20- to 50-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 K1 03N mutation;
DuPont); GW-420867X (has 25-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 Y181C and K103N mutations);
and Propolis (WO 99/49830).
[0470] Additional 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-756,423 (an indinavir analog; Merck); DMP-450 (a cyclic urea compound; Avid &
DuPont);
AG-1776 (a peptidomimetic with in vitro activity against protease inhibitor-resistant viruses;
Agouron); VX-175/GW-433908 (phosphate prodrug of amprenavir; Vertex & Glaxo Welcome); CGP61755 (Ciba); and AGENERASETM (amprenavir; Glaxo Wellcome Inc.).
[0471] 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).
[0472] Additional antiretroviral agents include fusion inhibitors/chemokine receptor antagonists. Fusion inhibitors/chemokine receptor antagonists include CXCR4 antagonists such as AMD 3100 (a bicyclam), SDF-1 and its analogs, and ALX40-4C (a cationic peptide), T22 (an 18 amino acid peptide; Trimeris) and the T22 analogs T134 and T140;

antagonists such as RANTES (9-68), AOP-RANTES, NNY-RANTES, and TAK-779; and CCR5/CXCR4 antagonists such as NSC 651016 (a distamycin analog). Also included are CCR2B, CCR3, and CCR6 antagonists. Chemokine recpetor agonists such as RANTES, SDF-1, MIP-1p, etc., may also inhibit fusion.
[0473] 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 98/50347.
[0474] 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 (IMPDH) inhibitors sucha as VX-497 (Vertex); and mycopholic acids such as CellCept (mycophenolate mofetil; Roche).
[0475] 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.
[0476] Other antiretroviral therapies and adjunct therapies include cytokines and lymphokines such as MIP-la, MIP-113, SDF-1 a, 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 (HIV
Irnmunogen), APL 400-003 (Apollon), recombinant gp120 and fragments, bivalent (B/E) recombinant envelope glycoprotein, rgp120CM235, MN rgp120, SF-2 rgp120, gp120/soluble CD4 complex, Delta JR-FL protein, branched synthetic peptide derived from discontinuous gp120 C3/C4 domain, fusion-competent immunogens, and Gag, Pol, Nef, and Tat vaccines;
gene-based therapies such as genetic suppressor elements (GSEs; WO 98/54366), and intrakines (genetically modified CC chemokines targetted to the ER to block surface expression of newly synthesized CCR5 (Yang et al., PNAS 94:11567-72 (1997);
Chen et al., Nat. Med. 3:1110-16 (1997)); antibodies such as the anti-CXCR4 antibody 12G5, the anti-CCR5 antibodies 2D7, 5C7, PA8, PA9, PA10, PAH, PA12, and PA14, the anti-CD4 antibodies Q4120 and RPA-T4, the anti-CCR3 antibody 7B11, the anti-gp120 antibodies 17b, 48d, 447-52D, 257-D, 268-D and 50.1, anti-Tat antibodies, anti-TNF-a 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 a-naphthoflavone (WO 98/30213); and antioxidants such as y-L-glutamyl-L-cysteine ethyl ester (y-GCE; WO
99/56764).
[0477] In a further embodiment, the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with an 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, amantadine, remantidine, maxamine, or thymalfasin. Specifically, interferon albumin fusion protein can be administered in combination with any of these agents. Moreover, 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. Furthermore, interferon beta albumin fusion protein can also be admistered with any of these agents.
Additionally, any of the IFN hybrids albumin fusion proteins can be administered in combination with any of these agents.
[0478] In a most preferred embodiment, interferon albumin fusion protein is administered in combination with ribavirin. In a further preferred embodiment, interferon alpha albumin fusion protein is administered in combination with ribavirin. In a further preferred embodiment, interferon alpha 2a albumin fusion protein is administered in combination with ribavirin. In a further preferred embodiment, interferon alpha 2b albumin fusion protein is administered in combination with ribavirin. In a further preferred embodiment, interferon beta albumin fusion protein is administered in combination with ribavirin. In a further preferred embodiment, hybrid interferon albumin fusion protein is administered in combination with ribavirin.

[0479] In other embodiments, 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, DAP SONETm, PENTAMIDINETm, ATOVAQUONETm, ISONIAZIDTM, RIFAMPINTm, PYRAZINAMIDETm, ETHAMBUTOLTm, RIFABUTINTm, CLARITHROMYCINTm, AZITHROMYCINTm, GANCICLOVIRTM, FOSCARNETTm, CIDOFOVIRTM, FLUCONAZOLETM, ITRACONAZOLETm, KETOCONAZOLETm, AC YCLOVIRTm, FAMCICOLVIRTm, PYRIMETHAMINETm, LEUCOVOR1NTM, NEUPOGENTM (filgrastim/G-CSF), and LEUK1NETM (sargramostim/GM-CSF). In a specific embodiment, 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. In another specific embodiment, 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. In another specific embodiment, 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. In another specific embodiment, 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. In another specific embodiment, 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. In another specific embodiment, 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 II infection. In another specific embodiment, albumin fusion proteins and/or polynucleotides of the invention are used in any combination with PYRIMETHAMINETm and/or LEUCOVORIIINTM to prophylactically treat or prevent an opportunistic Toxoplasma gondii infection. In another specific embodiment, albumin fusion proteins and/or polynucleotides of the invention are used in any combination with LEUCOVORINTM and/or NEUPOGENTM to prophylactically treat or prevent an opportunistic bacterial infection.
[0480] In a further embodiment, 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.
[0481] In other embodiments, 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).
[0482] In other embodiments, 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-506, 15-deoxyspergualin, and other immunosuppressive agents that act by suppressing the function of responding T cells. Other 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 (prednisolone), FOLEXTM

and MEXATETm (methotrx ate), OXSORALEN-ULTRATm (metho x sal en) and RAPAMUNETm (sirolimus). In a specific embodiment, immunosuppressants may be used to prevent rejection of organ or bone marrow transplantation.
[04831 In an additional embodiment, 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 fusion proteins and/or polynucleotides of the invention include, but not limited to, GAMMARTm, IVEEGAMTm, SANDOGLOBUL[NTM, GAMMAGARD S/DTM, ATGAMTm (antithymocyte glubulin), and GAMIMUNETm. In a specific embodiment, 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).
[04841 In another embodiment, 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. In a specific embodiment, 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).
[04851 In certain embodiments, the albumin fusion proteins and/or polynucleotides of the 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.
betamethasone, budesonide, cortisone, dexamethasone, hydrocortisone, methylprednisolone, prednisolone, prednisone, and triamcinolone), nonsteroidal anti-inflammatory drugs (e.g., diclofenac, diflunisal, etodolac, fenoprofen, floctafenine, flurbiprofen, ibuprofen, indomethacin, ketoprofen, meclofenamate, mefenamic acid, meloxicam, nabumetone, naproxen, oxaprozin, phenylbutazone, piroxicam, sulindac, tenoxicam, tiaprofenic acid, and tolmetin.), as well as antihistamines, aminoarylcarboxylic acid derivatives, arylacetic acid derivatives, arylbutyric acid derivatives, arylcarboxylic acids, arylpropionic acid derivatives, pyrazoles, pyrazolones, salicylic acid derivatives, thiazinecarboxamides, e-acetamidocaproic acid, S-adenosylmethionine, 3-amino-4-hydroxybutyric acid, amixetrine, bendazac, benzydamine, bucolome, difenpiramide, ditazol, emorfazone, guaiazulene, nabumetone, nimesulide, orgotein, oxaceprol, paranyline, perisoxal, pifoxime, proquazone, proxazole, and tenidap.
[0486] In an additional embodiment, the 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-1, Tissue Inhibitor of Metalloproteinase-2, VEGI, Plasminogen Activator Inhibitor-1, Plasminogen Activator Inhibitor-2, and various forms of the lighter "d group" transition metals.
[0487] 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.
[0488] Representative examples of 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.
[0489] Representative examples of 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 (VI) 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.
[0490] A wide variety of other anti-angiogenic factors may also be utilized within the context of the present invention. Representative examples include, but are not limited to, platelet factor 4; protamine sulphate; sulphated chitin derivatives (prepared from queen crab shells), (Murata et al., Cancer Res. 51:22-26, (1991)); 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-propy1-5-(4-pyridiny1)-2(3H)-oxazo lone; Methotrex ate; Mitoxantrone; Heparin;
Interferons; 2 Macroglobulin-serum; ChIMP-3 (Pavloff et al., J. Bio. Chem. 267:17321-17326, (1992));
Chymostatin (Tomkinson et al., Biochem J. 286:475-480, (1992));
Cyclodextrin Tetradecasulfate; Eponemycin; Camptothecin; Fumagillin (Ingber et al., Nature 348:555-557, (1990)); Gold Sodium Thiomalate ("GST"; Matsubara and Ziff, J. Clin. Invest.
79:1440-1446, (1987)); anticollagenase-serum; alpha2-antiplasmin (Holmes et al., J.
Biol. Chem.
262(4):1659-1664, (1987)); Bisantrene (National Cancer Institute); Lobenzarit disodium (N-(2)-carboxypheny1-4- chloroanthronilic acid disodium or "CCA"; (Takeuchi et al., Agents Actions 36:312-316, (1992)); and metalloproteinase inhibitors such as BB94.
[0491]
Additional anti-angiogenic factors that may also be utilized within the context of the present invention include Thalidomide, (Celgene, Warren, NJ);
Angiostatic steroid;
AGM-1470 (H. Brem and J. Folkman J Pediatr. Surg. 28:445-51 (1993)); an integrin alpha v beta 3 antagonist (C. Storgard et al., J Clin. Invest. 103:47-54 (1999));
carboxynaminolmidazole; Carboxyamidotriazole (CAI) (National Cancer Institute, Bethesda, MD); Conbretastatin A-4 (CA4P) (OXiGENE, Boston, MA); Squalamine (Magainin Pharmaceuticals, Plymouth Meeting, PA); TNP-470, (Tap Pharmaceuticals, Deerfield, IL);
ZD-0101 AstraZeneca (London, UK); APRA (C T2584); Benefin, Byrostatin-1 (SC339555);
CGP-41251 (PKC 412); CM101; Dexrazoxane (ICRF187); DMXAA; Endostatin;
Flavopridiol; Genestein; GTE; ImmTher; Iressa (ZD1839); Octreotide (Somatostatin);
Panretin; Penacillamine; Photopoint; PI-88; Prinomastat (AG-3340) Purlytin;
Suradista (FCE26644); Tamoxifen (Nolvadex); Tazarotene; Tetrathiomolybdate; Xeloda (Capecitabine); and 5-Fluorouracil.
[0492]
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. Examples of 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-12-9566 (Bayer, West Haven, CT), BMS-275291 (Bristol Myers Squibb, Princeton, NJ), CGS-27032A (Novartis, East Hanover, NJ), Marimastat (British Biotech, Oxford, UK), and Metastat (Aeterna, St-Foy, Quebec). Examples of 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). Examples of 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. San Francisco, CA), PTK-787/ZK-225846 (Novartis, Basel, Switzerland), SU-101 (Sugen, S. San Francisco, CA), SU-5416 (Sugen/ Pharmacia Upjohn, Bridgewater, NJ), and (Sugen). Other anti-angiogenic agents act to indirectly inhibit angiogenesis.
Examples of 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).
[0493] In particular embodiments, the use of 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.
[0494] In a particular embodiment, the use of compositions of the invention in combination with anti-angiogenic agents is contemplated for the treatment, prevention, and/or amelioration of arthritis. In a more particular embodiment, 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.
[0495] In another embodiment, the polynucleotides encoding a polypeptide of the present invention are administered in combination with an angiogenic protein, or polynucleotides encoding an angiogenic protein. Examples of angiogenic proteins that may be administered with the compositions of the invention include, but are not limited to, acidic and basic fibroblast growth factors, VEGF-1, 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.
104961 In additional embodiments, 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;
dimethyltriazenoimidazolecarboxamide)), folic acid analogs (for example, Methotrexate (amethopterin)), pyrimidine analogs (for example, Fluorouacil (5-fluorouracil;
5-FU), Floxuridine (fluorodeoxyuridine; FudR), and Cytarabine (cytosine arabinoside)), purine analogs and related inhibitors (for example, Mercaptopurine (6-mercaptopurine;
6-MP), Thioguanine (6-thioguanine; TG), and Pentostatin (2'-deoxycoformycin)), vinca alkaloids (for example, Vinblastine (VLB, vinblastine sulfate)) and Vincristine (vincristine sulfate)), epipodophyllotoxins (for example, Etoposide and Teniposide), antibiotics (for example, Dactinomycin (actinomycin D), Daunorubicin (daunomycin; rubidomycin), Doxorubicin, Bleomycin, Plicamycin (mithramycin), and Mitomycin (mitomycin C), enzymes (for example, L-Asparaginase), biological response modifiers (for example, Interferon-alpha and interferon-alpha-2b), platinum coordination compounds (for example, Cisplatin (cis-DDP) and Carboplatin), anthracenedione (Mitoxantrone), substituted ureas (for example, Hydroxyurea), methylhydrazine derivatives (for example, Procarbazine (N-methylhydrazine;
MIH), adrenocorticosteroids (for example, Prednisone), progestins (for example, Hydroxyprogesterone caproate, Medroxyprogesterone, Medroxyprogesterone acetate, and Megestrol acetate), estrogens (for example, Diethylstilbestrol (DES), Diethylstilbestrol diphosphate, Estradiol, and Ethinyl estradiol), antiestrogens (for example, Tamoxifen), androgens (Testosterone proprionate, and Fluoxymesterone), antiandrogens (for example, Flutamide), gonadotropin-releasing horomone analogs (for example, Leuprolide), other hormones and hormone analogs (for example, methyltestosterone, estramustine, estramustine phosphate sodium, chlorotrianisene, and testolactone), and others (for example, dicarbazine, glutamic acid, and mitotane).
[0497] In one embodiment, the 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-32-3555), Leflunomide (also known as AravaTm from Hoechst Marion Roussel), KineretTM (an IL-1 Receptor antagonist also known as Anakinra from Amgen, Inc.) [0498] In a specific embodiment, 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. In one embodiment, the compositions of the invention are administered in combination with anti-CD20 antibodies, human monoclonal anti-CD20 antibodies. In another embodiment, 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. In a specific embodiment, compositions of the invention are administered in combination with Rituximab. In a further embodiment, 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. In a specific embodiment, compositions of the invention are administered in combination with tositumomab. In a further embodiment, 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.
[0499] In another specific embodiment, the compositions of the invention are administered in combination ZevalinTM. In a further embodiment, 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 90y and 111/n.
[0500] In an additional embodiment, 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, IL3, IL4, IL5, IL6, IL7, IL10, IL12, IL13, IL15, anti-CD40, CD4OL, IFN-gamma and TNF-alpha. In another embodiment, 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, IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, IL-13, IL-14, IL-15, IL-16, IL-17, IL-18, IL-19, IL-20, and IL-21.
10501] In one embodiment, 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, CD3OL, CD4OL, 4-1BBL, DcR3, OX4OL, 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, 0X40, and nerve growth factor (NGF), and soluble forms of Fas, CD30, CD27, CD40 and 4-IBB, TR2 (International Publication No. WO 96/34095), DR3 (International Publication No. WO
97/33904), DR4 (International Publication No. WO 98/32856), TR5 (International Publication No. WO 98/30693), TRANK, TR9 (International Publication No. WO
98/56892),TR10 (International Publication No. WO 98/54202), 312C2 (International Publication No. WO 98/06842), and TR12, and soluble forms CD154, CD70, and CD153.
105021 In an additional embodiment, 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 (PIGF), as disclosed in International Publication Number WO
92/06194; Placental Growth Factor-2 (P1GF-2), as disclosed in Hauser et al., Growth Factors, 4:259-268 (1993); Vascular Endothelial Growth Factor (VEGF), as disclosed in International Publication Number WO 90/13649; Vascular Endothelial Growth Factor-A (VEGF-A), as disclosed in European Patent Number EP-506477; Vascular Endothelial Growth Factor-2 (VEGF-2), as disclosed in International Publication Number WO 96/39515;
Vascular Endothelial Growth Factor B (VEGF-3); Vascular Endothelial Growth Factor B-186 (VEGF-B186), as disclosed in International Publication Number WO 96/26736; Vascular Endothelial Growth Factor-D (VEGF-D), as disclosed in International Publication Number WO
98/02543; Vascular Endothelial Growth Factor-D (VEGF-D), as disclosed in International Publication Number WO 98/07832; and Vascular Endothelial Growth Factor-E (VEGF-E), as disclosed in German Patent Number DE19639601.
[0503] In an additional embodiment, 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-1, FGF-2, FGF-3, FGF-4, FGF-5, FGF-6, FGF-7, FGF-8, FGF-9, FGF-10, FGF-11, FGF-12, FGF-13, FGF-14, and FGF-15.
[0504] In an additional embodiment, the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with hematopoietic growth factors. 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, LEUKJNETM, PROKINETm), granulocyte colony stimulating factor (G-CSF) (filgrastim, NEUPOGENTm), macrophage colony stimulating factor (M-CSF, CSF-1) erythropoietin (epoetin alfa, EPOGENTM, PROCRITTm), stem cell factor (SCF, c-kit ligand, steel factor), megakaryocyte colony stimulating factor, PIXY321 (a GMCSF/IL-3 fusion protein), interleukins, especially any one or more of IL-1 through IL-12, interferon-gamma, or thrombopoietin.
[0505] In certain embodiments, 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, labetalol, metoprolol, nadolol, oxprenolol, penbutolol, pindolol, propranolol, sotalol, and timolol.
[0506] In another embodiment, the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with an antiarrhytlunic drug (e.g., adenosine, amidoarone, bretylium, digitalis, digoxin, digitoxin, diliazem, disopyramide, esmolol, flecainide, lidocaine, mexiletine, moricizine, phenytoin, procainarnide, N-acetyl procainarnide, propafenone, propranolol, quinidine, sotalol, tocainide, and verapamil).

[0507] In another embodiment, 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 Nat-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, methyclothiazide, polythiazide, trichormethiazide, chlorthalidone, indapamide, metolazone, and quinethazone), potassium sparing diuretics (e.g., amiloride and triamterene), and mineralcorticoid receptor antagonists (e.g., spironolactone, canrenone, and potassium canrenoate).
[0508] In one embodiment, 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, 1271, radioactive isotopes of iodine such as 1311 and 1231; 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 PROFASITM (chorionic gonadotropin (CG)), PERGONALTM (menotropins), and METRODINTm (urofollitropin (uFSH)); synthetic human gonadotropin releasing hormone preparations such as FACTRELTm and LUTREPULSETm (gonadorelin hydrochloride); synthetic gonadotropin agonists such as LUPRONTM
(leuprolide acetate), SUPPREL1NTM (histrelin acetate), SYNARELTM (nafarelin acetate), and ZOLADEXTM (goserelin acetate); synthetic preparations of thyrotropin-releasing hormone such as RELEFACT TRHTm and THYPINONETm (protirelin); recombinant human TSH
such as THYROGENTm; synthetic preparations of the sodium salts of the natural isomers of thyroid hormones such as L-T4Tm, SYNTHROIDTm and LEVOTHROIDTm (levothyroxine sodium), L-T3Tm, CYTOMELTm and TRIOSTATTm (liothyroine sodium), and THYROLARTm (liotrix);

antithyroid compounds such as 6-n-propylthiouracil (propylthiouracil), 1 -methy1-2-mercaptoimidazole and TAPAZOLETm (methimazole), NEO-MERCAZOLETm (carbimazole);

beta-adrenergic receptor antagonists such as propranolol and esmolol; Ca2+
channel blockers;

dexamethasone and iodinated radiological contrast agents such as TELEPAQUETm (iopanoic acid) and ORAGRAF1NTM (sodium ipodate).
[0509] 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 CLOMIDTm (clomiphene); progestins such as DURALUTINTm (hydroxyprogesterone caproate), MPATM and DEPO-PROVERATm (medroxyprogesterone acetate), PROVERATm and CYCR1NTM (MPA), MEGACETM
(megestrol acetate), NORLUTINTm (norethindrone), and NORLUTATETm and AYGESTINTm (norethindrone acetate); progesterone implants such as NORPLANT SYSTEMTm (subdermal implants of norgestrel); antiprogestins such as RU 486TM (mifepristone);
hormonal contraceptives such as ENOVIDTM (norethynodrel plus mestranol), PROGESTASERTTm (intrauterine device that releases progesterone), LOESTRINTm, BREVICONTM, MODICONTM, GENORATM, NELONATM, NORINYLTM, OVACON-35TM and OVACON-5OTM (ethinyl estradiol/norethindrone), LEVLENTM, NORDETTETm, TRI-LEVLENTm and TRIPHASIL-21Tm (ethinyl estradiol/levonorgestrel) LO/OVRALTM and OVRALTM (ethinyl estradiol/norgestrel), DEMULENTm (ethinyl estradiol/ethynodiol diacetate), NORINYLTM, ORTHO-NOVUMTm, NORETHINTm, GENORATM, and NELOVATM (norethindrone/mestranol), DESOGENTM and ORTHO-CEPTTm (ethinyl estradiol/desogestrel), ORTHO-CYCLENTm and ORTHO-TRICYCLENTm (ethinyl estradiol/norgestimate), MICRONORTM and NOR-QDTM
(norethindrone), and OVRETTETm (norgestrel).
[0510] 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-50Tm (testosterone), TESTEXTm (testosterone propionate), DELATESTRYLTm (testosterone enanthate), DEPO-TESTOSTERONETm (testosterone cypionate), DANOCR1NETM
(danazol), HALOTESTINTm (fluoxymesterone), ORETON METHYLTm, TESTREDTm and VIRILONTM
(methyltestosterone), and OXANDR1NTM (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 (al clometasone dipropionate), CYCLOCORTTm (amcinonide), BECLOVENTTm and VANCERILTM (beclomethasone dipropionate), CELESTONETm (betamethasone), BENISONETM and UTICORTTm (betamethasone benzoate), DIPROSONETM (betamethasone dipropionate), CELESTONE PHOSPHATETm (betamethasone sodium phosphate), CELESTONE SOLUSPANTM (betamethasone sodium phosphate and acetate), BETA-VALTm and VALISONETM (betamethasone valerate), TEMOVATETm (clobetasol propionate), CLODERMTm (clocortolone pivalate), CORTEFTm and HYDROCORTONETm (cortisol (hydrocortisone)), HYDROCORTONE ACETATETm (cortisol (hydrocortisone) acetate), LOCOIDTM (cortisol (hydrocortisone) butyrate), HYDROCORTONE PHOSPHATETm (cortisol (hydrocortisone) sodium phosphate), A-HYDROCORTTm and SOLU CORTEFTm (cortisol (hydrocortisone) sodium succinate), WESTCORTTm (cortisol (hydrocortisone) valerate), CORTISONE ACETATETm (cortisone acetate), DESOWENTTM and TRIDESILONTm (desonide), TOPICORTTm (desoximetasone), DECADRONTM
(dexamethasone), DECADRON LATM (dexamethasone acetate), DECADRON
PHOSPHATETm and HEXADROL PHOSPHATETm (dexamethasone sodium phosphate), FLORONETM and MAXIFLORTM (diflorasone diacetate), FLORINEF ACETATETm (fludrocortisone acetate), AEROBIDTM and NASALIDETTM (flunisolide), FLUONIDTM
and 54LJ\JTM (fluocinolone acetonide), LIDEXTM (fluocinonide), FLUOR-OPTTM and FMLTm (fluorometholone), CORDRANTM (flurandrenolide), HALOGTM (halcinonide), HMS
LIZUIFILMTm (medrysone), MEDROLTM (methylprednisolone), DEPO-MEDROLTm and MEDROL ACETATETm (methylprednisone acetate), A-METHAPREDTm and SOLUMEDROLTm (Tethylprednisolone sodium succinate), ELOCONTTM (mometasone furoate), HALDRONETM (paramethasone acetate), DELTA-CORTEFTm (prednisolone), ECONOPREDTM (prednisolone acetate), HYDELTRASOLTm (prednisolone sodium phosphate), HYDELTRA-T.B.ATm (prednisolone tebutate), DELTASONETm (prednisone), ARISTOCORTTm and KENACORTTm (triamcinolone), KENALOGTM (triamcinolone acetonide), ARISTOCORTTm and KENACORT DIACETATETm (triamcinolone diacetate), and ARISTOSPANTm (triamcinolone hexacetonide); inhibitors of biosynthesis and action of adrenocortical steroids such as CYTADRENTm (aminoglutethimide), NIZORALTM
(ketoconazole), MODRASTANETm (trilostane), and METOPIRONETm (metyrapone);
bovine, porcine or human insulin or mixtures thereof; insulin analogs; recombinant human insulin such as HUMULINTm and NOVOLINTM; oral hypoglycemic agents such as ORAMIDETm and ORINASETM (tolbutamide), DIAB1NESETM (chlorpropamide), TOLAMIDETm and TOLINASETm (tolazamide), DYMELORTm (acetohexamide), glibenclamide, MICRONASETm, DIBETATm and GLYNASETM (glyburide), GLUCOTROLTm (glipizide), and DIAMICRONTm (gliclazide), GLUCOPHAGETM (metformin), ciglitazone, pioglitazone, and alpha-glucosidase inhibitors; bovine or porcine glucagon; somatostatins such as SANDOSTATINTm (octreotide);
and diazoxides such as PROGLYCEMTm (diazoxide).
[0511] In one embodiment, 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., CLIMARA 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 estsradiol (e.g., FEMHRTTm).
[0512] In an additional embodiment, 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 B12, cyancobalamin injection (e.g., REDISOLTM, RUBRAMIN PCTm), hydroxocobalamin, folic acid (e.g., FOLVITETm), leucovorin (folinic acid, 5-CHOH4PteG1u, citrovorum factor) or WELLCOVORIN
(Calcium salt of leucovorin), transferrin or ferritin.
[0513] In certain embodiments, 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, amoxapine, bupropion, citalopram, clomipramine, desipramine, doxepin, fluvoxamine, fluoxetine, imipramine, isocarboxazid, maprotiline, mirtazapine, nefazodone, nortriptyline, paroxetine, phenelzine, protriptyline, sertraline, tranylcypromine, trazodone, trimipramine, and venlafaxine), antianxiety agents (e.g., alprazolam, buspirone, chlordiazepoxide, clorazepate, diazepam, halazepam, lorazepam, oxazepam, and prazepam), and stimulants (e.g., d-amphetamine, methylphenidate, and pemoline).
[0514] In other embodiments, the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with agents used to treat neurological disorders. 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, levetiracetam, oxcarbazepine, tiagabine, topiramate, zonisamide, diazepam, lorazepam, and clonazepam), antiparkinsonian agents (e.g., levodopa/carbidopa, selegiline, amantidine, bromocriptine, pergolide, ropinirole, pramipexole, benztropine; biperiden; ethopropazine;
procyclidine;
trihexyphenidyl, tolcapone), and ALS therapeutics (e.g. riluzole).
[0515] In another embodiment, albumin fusion proteins and/or polynucleotides of the invention are administered in combination with vasodilating agents and/or calcium channel blocking agents. Vasodilating agents that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, Angiotensin Converting Enzyme (ACE) inhibitors (e.g., papaverine, isoxsuprine, benazepril, captopril, cilazapril, enalapril, enalaprilat, fosinopril, lisinopril, moexipril, perindopril, quinapril, ramipril, spirapril, trandolapril, and nylidrin), and nitrates (e.g., isosorbide dinitrate, isosorbide mononitrate, and nitroglycerin). Examples of calcium channel blocking agents that may be administered in combination with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to amlodipine, bepridil, diltiazem, felodipine, flunarizine, isradipine, nicardipine, nifedipine, nimodipine, and verapamil.

[0516] In certain embodiments, 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, H2 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. CYTOTECTivi (misoprostol)); muscarinic cholinergic antagonists; laxatives (e.g., surfactant laxatives, stimulant laxatives, saline and osmotic laxatives); antidiarrheal agents (e.g., LOMOTILTM (diphenoxylate), MOTOFENTM
(diphenoxin), and IMODIUMTm (loperamide hydrochloride)), synthetic analogs of somatostatin such as SANDOSTATINTm (octreotide), antiemetic agents (e.g., ZOFRANTm (ondansetron), KYTRILTM (granisetron hydrochloride), tropisetron, dolasetron, metoclopramide, chlorpromazine, perphenazine, prochlorperazine, promethazine, thiethylperazine, triflupromazine, domperidone, haloperidol, droperidol, trimethobenzamide, dexamethasone, methylprednisolone, dronabinol, and nabilone); D2 antagonists (e.g., metoclopramide, trimethobenzamide and chlorpromazine); bile salts;
chenodeoxycholic acid;
ursodeoxycholic acid; and pancreatic enzyme preparations such as pancreatin and pancrelipase.
[0517] In additional embodiments, the 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.
[0518] 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. Optionally associated with such container(s) 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.
Gene Therapy [0519] 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. Additionally, 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.
[0520] 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. The development of specialized cell lines (termed "packaging cells") which produce only replication-defective retroviruses has increased the utility of retroviruses for gene therapy, and defective retroviruses are characterized for use in gene transfer for gene therapy purposes (for a review see Miller, A.D. (1990) Blood 76:27 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, F.M. et al., (eds.) Greene Publishing Associates, (1989), Sections 9.10-9.14 and other standard laboratory manuals.
[0521] Another viral gene delivery system useful in the present invention uses adenovirus-derived vectors. The genome of an adenovirus can be manipulated such that it encodes and expresses a gene product of interest but is inactivated in terms of its ability to replicate in a normal lytic viral life cycle. See, for example, Berkner et al., BioTechniques 6:616 (1988); Rosenfeld et al., Science 252:431-434 (1991); and Rosenfeld et al., Cell 68:143-155 (1992). Suitable adenoviral vectors derived from the adenovirus strain Ad type 5 dl 324 or other strains of adenovirus (e.g., Ad2, Ad3, Ad7 etc.) are known to those skilled in the art. Recombinant adenoviruses can be advantageous in certain circumstances in that they are not capable of infecting nondividing cells and can be used to infect a wide variety of cell types, including epithelial cells (Rosenfeld et al., (1992) cited supra).
Furthermore, the virus particle is relatively stable and amenable to purification and concentration, and as above, can be modified so as to affect the spectrum of infectivity. Additionally, 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). Moreover, 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 etal., J. Virol. 57:267 (1986)).
[0522] In another embodiment, 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. In a representative embodiment, 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 etal. (1992) No Shinkei Geka 20:547-5 5 1; PCT publication W091/06309;
Japanese patent application 1047381; and European patent publication EP-A-43075).
[0523]
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. For instance, 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. In other embodiments, initial delivery of the recombinant gene is more limited with introduction into the animal being quite localized. For example, the gene delivery vehicle can be introduced by catheter (see U.S.
Patent 5,328,470) or by Stereotactic injection (e.g. Chen et al. (1994) PNAS
91: 3 054-3 05 7). 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. Where 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.
Additional Gene Therapy Methods [0524]
Also encompassed by the invention are gene therapy methods for treating or preventing disorders, diseases and conditions. 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, W090/11092.
[05251 Thus, for example, 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 the engineered cells then being provided to a patient to be treated with the fusion protein of the present invention. 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. Immunology 153: 4604-4615 (1994); Kaido, T., et al., Int. J.
Cancer 60: 221-229 (1995); Ogura, H., et al., Cancer Research 50: 5102-5106 (1990);
Santodonato, L., et al., Human Gene Therapy 7:1-10 (1996); Santodonato, L., et al., Gene Therapy 4:1246-1255 (1997); and Zhang, J.-F. et al., Cancer Gene Therapy 3: 31-38 (1996)), In one embodiment, 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.
[05261 As discussed in more detail below, 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.
105271 In one embodiment, polynucleotides encoding the albumin fusion proteins of the present invention is delivered as a naked polynucleotide.
The term "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. However, 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.

[0528] 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, p0G44, pXT1 and pSG available from Stratagene; pSVK3, pBPV, pMSG and pSVL
available from Pharmacia; and pEF1N5, pcDNA3.1, and pRc/CMV2 available from Invitrogen. Other suitable vectors will be readily apparent to the skilled artisan.
[0529] Any strong promoter known to those skilled in the art can be used for driving the expression of the polynucleotide sequence. 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 shock promoters;
the albumin promoter; the ApoAI promoter; human globin promoters; viral thymidine kinase promoters, such as the Herpes Simplex thymidine kinase promoter; retroviral LTRs; the b-actin promoter; and human growth hormone promoters. The promoter also may be the native promoter for the gene corresponding to the Therapeutic protein portion of the albumin fusion proteins of the invention.
[0530] 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.
[0531] 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. It is similarly 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.
[0532] For the naked nucleic acid sequence injection, 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.
[0533] The preferred route of administration is by the parenteral route of injection into the interstitial space of tissues. However, other 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. In addition, naked DNA constructs can be delivered to arteries during angioplasty by the catheter used in the procedure.
[0534] 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.
[0535] 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.
[0536] In certain embodiments, 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. However, 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);
mRNA
(Malone et al., Proc. Natl. Acad. Sci. USA (1989) 86:6077-6081;

and purified transcription factors (Debs et al., J. Biol. Chem.
(1990) 265:10189-10192), in functional form.
[0537) Cationic liposomes are readily available. For example, N[1-2,3-dioleyloxy)propyfl-N,N,N-triethylarnmonium (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 Acad. Sci. USA (1987) 84:7413-7416).
Other commercially available liposomes include transfectace (DDAB/DOPE) and DOTAP/DOPE (Boehringer).
[0538]
Other cationic liposomes can be prepared from readily available materials using techniques well known in the art. See, e.g. PCT Publication No. WO

for a description of the synthesis of DOTAP (1,2-bis(oleoyloxy)-3-(trimethylammonio)propane) liposomes. Preparation of DOTMA
liposomes is explained in the literature, see, e.g., P. Feigner et al., Proc. Natl.
Acad. Sci. USA
84:7413-7417.
Similar methods can be used to prepare liposomes from other cationic lipid materials.
[0539]
Similarly, 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. These materials can also be mixed with the DOTMA and DOTAP starting materials in appropriate ratios. Methods for making liposomes using these materials are well known in the art.
[0540]
For example, commercially dioleoylphosphatidyl choline (DOPC), dioleoylphosphatidyl glycerol (DOPG), and dioleoylphosphatidyl ethanolamine (DOPE) can be used in various combinations to make conventional liposomes, with or without the addition of cholesterol. Thus, for example, DOPG/DOPC vesicles can be prepared by drying 50 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 15 degrees celcius.
Alternatively, 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.
[0541] The liposomes can comprise multilamellar vesicles (MLVs), small unilamellar vesicles (SUVs), or large unilamellar vesicles (LUVs), with SUVs being preferred. 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.
For example, 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. When using 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/NaC1, 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 Ca2+-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. and Strittmatter, P., Proc. Natl. Acad. Sci. USA 76:145 (1979)); and reverse-phase evaporation (REV) (Fraley et al., J. Biol. Chem. 255:10431 (1980); Szoka, F. and Papahadjopoulos, D., Proc. Natl. Acad.
Sci. USA 75:145 (1978); Schaefer-Ridder etal., Science 215:166 (1982)).
[0542] Generally, the ratio of DNA to liposomes will be from about 10:1 to about 1:10. Preferably, the ration will be from about 5:1 to about 1:5. More preferably, the ration will be about 3:1 to about 1:3. Still more preferably, the ratio will be about 1:1.
[0543] U.S. Patent No. 5,676,954 reports on the injection of genetic material, complexed with cationic liposomes carriers, into mice.
U.S. Patent Nos. 4,897,355, 4,946,787, 5,049,386, 5,459,127, 5,589,466, 5,693,622, 5,580,859, 5,703,055, and international publication no. WO 94/9469 provide cationic lipids for use in transfecting DNA into cells and mammals. U.S. Patent Nos. 5,589,466, 5,693,622, 5,580,859, 5,703,055, and international publication no. WO 94/9469 provide methods for delivering DNA-cationic lipid complexes to mammals.
[0544] In certain embodiments, cells are engineered, ex vivo or in vivo, using a retroviral particle containing RNA which comprises a sequence encoding an albumin fusion protein of the present invention. Retroviruses from which the retroviral plasmid vectors may be derived include, but are not limited to, Moloney Murine Leukemia Virus, spleen necrosis virus, Row sarcoma Virus, Harvey Sarcoma Virus, avian leukosis virus, gibbon ape leukemia virus, human immunodeficiency virus, Myeloproliferative Sarcoma Virus, and mammary tumor virus.
[0545]
The retroviral plasmid vector is employed to transduce packaging cell lines to form producer cell lines. Examples of packaging cells which may be transfected include, but are not limited to, the PE501, PA317, R-2, R-AM, PA12, T19-14X, VT-19-17-H2, RCRE, RCRIP, GP+E-86, GP+envAm12, and DAN cell lines as described in Miller, Human Gene Therapy 1:5-14 (1990), The vector may transduce the packaging cells through any means known in the art. Such means include, but are not limited to, electroporation, the use of liposomes, and CaPO4 precipitation. In one alternative, the retroviral plasmid vector may be encapsulated into a liposome, or coupled to a lipid, and then administered to a host.
[0546]
The producer cell line generates infectious retroviral vector particles which include polynucleotide encoding an albumin fusion protein of the present invention. Such retroviral vector particles then may be employed, to transduce eukaryotic cells, either in vitro or in vivo. The transduced eulcaryotic cells will express a fusion protein of the present invention.
[0547] In certain other embodiments, cells are engineered, ex vivo or in vivo, with polynucleotide contained in an adenovirus vector. Adenovirus can be manipulated such that it encodes and expresses fusion protein of the present invention, and at the same time is inactivated in terms of its ability to replicate in a normal lytic viral life cycle. Adenovirus expression is achieved without integration of the viral DNA into the host cell chromosome, thereby alleviating concerns about insertional mutagenesis. Furthermore, adenoviruses have been used as live enteric vaccines for many years with an excellent safety profile (Schwartz et al. Am. Rev. Respir. Dis.109:233-238 (1974)). Finally, adenovirus mediated gene transfer has been demonstrated in a number of instances including transfer of alpha-1 -antitrypsin and CFTR to the lungs of cotton rats (Rosenfeld, M. A. et al. (1991) Science 252:431-434;

Rosenfeld et al., (1992) Cell 68:143-155). Furthermore, extensive studies to attempt to establish adenovirus as a causative agent in human cancer were uniformly negative (Green, M. et al. (1979) Proc. Natl. Acad. Sci. USA 76:6606).
[0548] Suitable adenoviral vectors useful in the present invention are described, for example, in Kozarsky and Wilson, Curr. Opin. Genet. Devel. 3:499-503 (1993);
Rosenfeld et al., Cell 68:143-155 (1992); Engelhardt et al., Human Genet. Ther. 4:759-769 (1993); Yang et al., Nature Genet. 7:362-369 (1994); Wilson et al., Nature 365:691-692 (1993); and U.S.
Patent No. 5,652,224.
For example, the adenovirus vector Ad2 is useful and can be grown in human 293 cells. These cells contain the El region of adenovirus and constitutively express Ela and Elb, which complement the defective adenoviruses by providing the products of the genes deleted from the vector. In addition to Ad2, other varieties of adenovirus (e.g., Ad3, Ad5, and Ad7) are also useful in the present invention.
[0549] Preferably, the adenoviruses used in the present invention are replication deficient. Replication deficient adenoviruses require the aid of a helper virus and/or packaging cell line to form infectious particles. The resulting virus is capable of infecting cells and can express a polynucleotide of interest which is operably linked to a promoter, but cannot replicate in most cells. Replication deficient adenoviruses may be deleted in one or more of all or a portion of the following genes: Ela, El b, E3, E4, E2a, or Li through L5.
[0550] In certain other embodiments, the cells are engineered, ex vivo or in vivo, using an adeno-associated virus (AAV). AAVs are naturally occurring defective viruses that require helper viruses to produce infectious particles (Muzyczka, N., Curr.
Topics in Microbiol. Immunol. 158:97 (1992)). It is also one of the few viruses that may integrate its DNA into non-dividing cells. Vectors containing as little as 300 base pairs of AAV can be packaged and can integrate, but space for exogenous DNA is limited to about 4.5 kb.
Methods for producing and using such AAVs are known in the art. See, for example, U.S.
Patent Nos. 5,139,941, 5,173,414, 5,354,678, 5,436,146, 5,474,935, 5,478,745, and 5,589,377.
105511 For example, an appropriate AAV vector for use in the present invention will include all the sequences necessary for DNA replication, encapsidation, and host-cell integration. The polynucleotide construct is inserted into the AAV vector using standard cloning methods, such as those found in Sambrook et al., Molecular Cloning: A
Laboratory Manual, Cold Spring Harbor Press (1989). The recombinant AAV vector is then transfected into packaging cells which are infected with a helper virus, using any standard technique, including lipofection, electroporation, calcium phosphate precipitation, etc.
Appropriate helper viruses include adenoviruses, cytomegaloviruses, vaccinia viruses, or herpes viruses.
Once the packaging cells are transfected and infected, they will produce infectious AAV viral particles which contain the polynucleotide construct. These viral particles are then used to transduce eukaryotic cells, either ex vivo or in vivo. The transduced cells will contain the polynucleotide construct integrated into its genome, and will express a fusion protein of the invention.
[0552] Another method of gene therapy involves operably associating heterologous control regions and endogenous polynucleotide sequences (e.g. encoding a polypeptide of the present invention) via homologous recombination (see, e.g., U.S. Patent No.
5,641,670, issued June 24, 1997; International Publication No. WO 96/29411, published September 26, 1996; International Publication No. WO 94/12650, published August 4, 1994;
Koller et al., Proc. Natl. Acad. Sci. USA 86:8932-8935 (1989); and Zijlstra et al., Nature 342:435-438 (1989), which are herein encorporated by reference. This method involves the activation of a gene which is present in the target cells, but which is not normally expressed in the cells, or is expressed at a lower level than desired.
[0553] Polynucleotide constructs are made, using standard techniques known in the art, which contain the promoter with targeting sequences flanking the promoter. Suitable promoters are described herein. The targeting sequence is sufficiently complementary to an endogenous sequence to permit homologous recombination of the promoter-targeting sequence with the endogenous sequence. The targeting sequence will be sufficiently near the 5' end of the desired endogenous polynucleotide sequence so the promoter will be operably linked to the endogenous sequence upon homologous recombination.
[0554] The promoter and the targeting sequences can be amplified using PCR.
Preferably, the amplified promoter contains distinct restriction enzyme sites on the 5' and 3' ends. Preferably, the 3' end of the first targeting sequence contains the same restriction enzyme site as the 5' end of the amplified promoter and the 5' end of the second targeting sequence contains the same restriction site as the 3' end of the amplified promoter. The amplified promoter and targeting sequences are digested and ligated together.
[0555] The promoter-targeting sequence construct is delivered to the cells, either as naked polynucleotide, or in conjunction with transfection-facilitating agents, such as liposomes, viral sequences, viral particles, whole viruses, lipofection, precipitating agents, etc., described in more detail above. The P promoter-targeting sequence can be delivered by any method, included direct needle injection, intravenous injection, topical administration, catheter infusion, particle accelerators, etc. The methods are described in more detail below.
[0556] The promoter-targeting sequence construct is taken up by cells.
Homologous recombination between the construct and the endogenous sequence takes place, such that an endogenous sequence is placed under the control of the promoter. The promoter then drives the expression of the endogenous sequence.
[0557] The polynucleotide encoding an albumin fusion protein of the present invention may contain a secretory signal sequence that facilitates secretion of the protein.
Typically, the signal sequence is positioned in the coding region of the polynucleotide to be expressed towards or at the 5' end of the coding region. The signal sequence may be homologous or heterologous to the polynucleotide of interest and may be homologous or heterologous to the cells to be transfected. Additionally, the signal sequence may be chemically synthesized using methods known in the art.
[0558] Any mode of administration of any of the above-described polynucleotides constructs can be used so long as the mode results in the expression of one or more molecules in an amount sufficient to provide a therapeutic effect. This includes direct needle injection, systemic injection, catheter infusion, biolistic injectors, particle accelerators (i.e., "gene guns"), gelfoam sponge depots, other commercially available depot materials, osmotic pumps (e.g., Alza minipumps), oral or suppositorial solid (tablet or pill) pharmaceutical formulations, and decanting or topical applications during surgery. For example, direct injection of naked calcium phosphate-precipitated plasmid into rat liver and rat spleen or a protein-coated plasmid into the portal vein has resulted in gene expression of the foreign gene in the rat livers (Kaneda et al., Science 243:375 (1989)).
[0559] A preferred method of local administration is by direct injection.
Preferably, an albumin fusion protein of the present invention complexed with a delivery vehicle is administered by direct injection into or locally within the area of arteries.
Administration of a composition locally within the area of arteries refers to injecting the composition centimeters and preferably, millimeters within arteries.
[0560] Another method of local administration is to contact a polynucleotide construct of the present invention in or around a surgical wound. For example, a patient can undergo surgery and the polynucleotide construct can be coated on the surface of tissue inside the wound or the construct can be injected into areas of tissue inside the wound.

[05611 Therapeutic compositions useful in systemic administration, include fusion proteins of the present invention complexed to a targeted delivery vehicle of the present invention. Suitable delivery vehicles for use with systemic administration comprise liposomes comprising ligands for targeting the vehicle to a particular site. In specific embodiments, suitable delivery vehicles for use with systemic administration comprise liposomes comprising albumin fusion proteins of the invention for targeting the vehicle to a particular site.
[0562]
Preferred methods of systemic administration, include intravenous injection, aerosol, oral and percutaneous (topical) delivery. Intravenous injections can be performed using methods standard in the art. Aerosol delivery can also be performed using methods standard in the art (see, for example, Stribling et al., Proc. Natl. Acad.
Sci. USA
189:11277-11281, 1992).
Oral delivery can be performed by complexing a polynucleotide construct of the present invention to a carrier capable of withstanding degradation by digestive enzymes in the gut of an animal. Examples of such carriers, include plastic capsules or tablets, such as those known in the art. Topical delivery can be performed by mixing a polynucleotide construct of the present invention with a lipophilic reagent (e.g., DMSO) that is capable of passing into the skin.
[0563]
Determining an effective amount of substance to be delivered can depend upon a number of factors including, for example, the chemical structure and biological activity of the substance, the age and weight of the animal, the precise condition requiring treatment and its severity, and the route of administration. The frequency of treatments depends upon a number of factors, such as the amount of polynucleotide constructs administered per dose, as well as the health and history of the subject. The precise amount, number of doses, and timing of doses will be determined by the attending physician or veterinarian.
[0564]
Albumin fusion proteins of the present invention can be administered to any animal, preferably to mammals and birds. Preferred mammals include humans, dogs, cats, mice, rats, rabbits sheep, cattle, horses and pigs, with humans being particularly preferred.
Biological Activities [0565]
Albumin fusion proteins and/or polynucleotides encoding albumin fusion proteins of the present invention, can be used in assays to test for one or more biological activities. If an albumin fusion protein and/or polynucleotide exhibits an activity in a particular assay, it is likely that the Therapeutic protein corresponding to the fusion portein may be involved in the diseases associated with the biological activity. Thus, the fusion protein could be used to treat the associated disease.
[0566] In preferred embodiments, 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 portion corresponding to a Therapeutic protein disclosed in the "Therapeutic Protein X"
column of Table 1 (in the same row as the disease or disorder to be treated is listed in the "Preferred Indication Y" column of Table 1) in an amount effective to treat, prevent or ameliorate the disease or disorder.
[0567] In a further preferred embodiment, the present invention encompasses a method of treating a disease or disorder listed for a particular Therapeutic protein 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 portion corresponding to the Therapeutic protein for which the indications in the Examples are related in an amount effective to treat, prevent or ameliorate the disease or disorder.
[0568] Specifically contemplated by the present invention are albumin fusion proteins produced by a cell when encoded by the 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 explicitly listed in columns 4 and/or 11 of Table 2. The specific amino acid sequence of the listed signal sequence is shown in the specification or is well known in the art.
Thus, most preferred embodiments of the present invention include the albumin fusion protein produced by a cell (which would lack the leader sequence shown in columns 4 and/or 11 of Table 2).
Also most preferred are polypeptides comprising SEQ ID NO:Y without the specific leader sequence listed in columns 4 and/or 11 of Table 2. Compositions comprising these two preferred embodiments, including pharmaceutical compositions, are also preferred. These albumin fusion proteins are specifically contemplated to treat, prevent, or ameliorate a disease or disorder listed for a particular Therapeutic protein in the "Preferred Indication:Y" column of Table 1.
[0569] In preferred embodiments, fusion proteins of the present invention may be used in the diagnosis, prognosis, prevention and/or treatment of diseases and/or disorders relating to diseases and disorders of the endocrine system (see, for example, "Endocrine Disorders" section below), the nervous system (see, for example, "Neurological Disorders"
section below), the immune system (see, for example, "Immune Activity" section below), respiratory system (see, for example, "Respiratory Disorders" section below), cardiovascular system (see, for example, "Cardiovascular Disorders" section below), reproductive system (see, for example, "Reproductive System Disorders" section below) digestive system (see, for example, "Gastrointestinal Disorders" section below), diseases and/or disorders relating to cell proliferation (see, for example, "Hyperproliferative Disorders" section below), and/or diseases or disorders relating to the blood (see, for example, "Blood-Related Disorders"
section below).
[0570] In certain embodiments, an albumin fusion protein of the present invention may be used to diagnose and/or prognose diseases and/or disorders associated with the tissue(s) in which the gene corresponding to the Therapeutic protein portion of the fusion protein of the invention is expressed.
[0571] Thus, fusion proteins of the invention and polynucleotides encoding albumin fusion proteins of the invention are useful in the diagnosis, detection and/or treatment of diseases and/or disorders associated with activities that include, but are not limited to, prohormone activation, neurotransmitter activity, cellular signaling, cellular proliferation, cellular differentiation, and cell migration.
[0572] More generally, fusion proteins of the invention and polynucleotides encoding albumin fusion proteins of the invention may be useful for the diagnosis, prognosis, prevention and/or treatment of diseases and/or disorders associated with the following systems.
Immune Activity [0573] Albumin fusion proteins of the invention and polynucleotides encoding albumin fusion proteins of the invention may be useful in treating, preventing, diagnosing and/or prognosing diseases, disorders, and/or conditions of the immune system, by, for example, activating or inhibiting the proliferation, differentiation, or mobilization (chemotaxis) of immune cells. Immune cells develop through a process called hematopoiesis, producing myeloid (platelets, red blood cells, neutrophils, and macrophages) and lymphoid (B
and T lymphocytes) cells from pluripotent stem cells. The etiology of these immune diseases, disorders, and/or conditions may be genetic, somatic, such as cancer and some autoimmune diseases, acquired (e.g., by chemotherapy or toxins), or infectious. Moreover, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention can be used as a marker or detector of a particular immune system disease or disorder.
[0574] In another embodiment, a fusion protein of the invention and/or polynucleotide encoding an albumin fusion protein of the invention, may be used to treat diseases and disorders of the immune system and/or to inhibit or enhance an immune response generated by cells associated with the tissue(s) in which the polypeptide of the invention is expressed.
[0575]
Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in treating, preventing, diagnosing, and/or prognosing immunodeficiencies, including both congenital and acquired immunodeficiencies. Examples of B cell immunodeficiencies in which immunoglobulin levels B cell function and/or B cell numbers are decreased include: X-linked agammaglobulinemia (Bruton's disease), X-linked infantile agammaglobulinemia, X-linked immunodeficiency with hyper IgM, non X-linked immunodeficiency with hyper IgM, X-linked lymphoproliferative syndrome (XLP), agammaglobulinemia including congenital and acquired agammaglobulinemia, adult onset agammaglobulinemia, late-onset agammaglobulinemia, dysgammaglobulinemia, hypogammaglobulinemia, unspecified hypogammaglobulinemia, recessive agammaglobulinemia (Swiss type), Selective IgM
deficiency, selective IgA deficiency, selective IgG subclass deficiencies, IgG
subclass deficiency (with or without IgA deficiency), Ig deficiency with increased IgM, IgG and IgA
deficiency with increased IgM, antibody deficiency with normal or elevated Igs, Ig heavy chain deletions, kappa chain deficiency, B cell lymphoproliferative disorder (BLPD), common variable immunodeficiency (CVID), common variable immunodeficiency (CVI) (acquired), and transient hypogammaglobulinemia of infancy.
[0576] In specific embodiments, ataxia-telangiectasia or conditions associated with ataxia-telangiectasia are treated, prevented, diagnosed, and/or prognosing using the, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention.
[0577]
Examples of congenital immunodeficiencies in which T cell and/or B cell function and/or number is decreased include, but are not limited to: DiGeorge anomaly, severe combined immunodeficiencies (SCID) (including, but not limited to, X-linked SCID, autosomal recessive SOD, adenosine deaminase deficiency, purine nucleoside phosphorylase (PNP) deficiency, Class II MHC deficiency (Bare lymphocyte syndrome), Wiskott-Aldrich syndrome, and ataxia telangiectasia), thymic hypoplasia, third and fourth pharyngeal pouch syndrome, 22q11.2 deletion, chronic mucocutaneous candidiasis, natural killer cell deficiency (NK), idiopathic CD4+ T-lymphocytopenia, immunodeficiency with predominant T
cell defect (unspecified), and unspecified immunodeficiency of cell mediated immunity.
[0578] In specific embodiments, DiGeorge anomaly or conditions associated with DiGeorge anomaly are treated, prevented, diagnosed, and/or prognosed using fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention.
[0579]
Other immunodeficiencies that may be treated, prevented, diagnosed, and/or prognosed using fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, include, but are not limited to, chronic granulomatous disease, Chediak-Higashi syndrome, myeloperoxidase deficiency, leukocyte glucose-6-phosphate dehydrogenase deficiency, X-linked lymphoproliferative syndrome (XLP), leukocyte adhesion deficiency, complement component deficiencies (including Cl, C2, C3, C4, C5, C6, C7, C8 and/or C9 deficiencies), reticular dysgenesis, thymic alymphoplasia-aplasia, immunodeficiency with thymoma, severe congenital leukopenia, dysplasia with immunodeficiency, neonatal neutropenia, short limbed dwarfism, and Nezelof syndrome-combined immunodeficiency with Igs.
[0580] In a preferred embodiment, the immunodeficiencies and/or conditions associated with the immunodeficiencies recited above are treated, prevented, diagnosed and/or prognosed using fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention.
[0581] In a preferred embodiment fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention could be used as an agent to boost immunoresponsiveness among immunodeficient individuals. In specific embodiments, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention could be used as an agent to boost immunoresponsiveness among B cell and/or T cell immunodeficient individuals.
[0582] The albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in treating, preventing, diagnosing and/or prognosing autoimmune disorders. Many autoimmune disorders result from inappropriate recognition of self as foreign material by immune cells. This inappropriate recognition results in an immune response leading to the destruction of the host tissue.

Therefore, the administration of fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention that can inhibit an immune response, particularly the proliferation, differentiation, or chemotaxis of T-cells, may be an effective therapy in preventing autoimmune disorders.
[0583] Autoimmune diseases or disorders that may be treated, prevented, diagnosed and/or prognosed by fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to, one or more of the following: systemic lupus erythematosus, rheumatoid arthritis, ankylosing spondylitis, multiple sclerosis, autoimmune thyroiditis, Hashimoto's thyroiditis, autoimmune hemolytic anemia, hemolytic anemia, thrombocytopenia, autoimmune thrombocytopenia purpura, autoimmune neonatal thrombocytopenia, idiopathic thrombocytopenia purpura, purpura (e.g., Henloch-Scoenlein purpura), autoimmunocytopenia, Goodpasture's syndrome, Pemphigus vulgaris, myasthenia gravis, Grave's disease (hyperthyroidism), and insulin-resistant diabetes mellitus.
[0584] Additional disorders that are likely to have an autoimmune component that may be treated, prevented, and/or diagnosed with the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to, type II collagen-induced arthritis, antiphospholipid syndrome, dermatitis, allergic encephalomyelitis, myocarditis, relapsing polychondritis, rheumatic heart disease, neuritis, uveitis ophthalmia, polyendocrinopathies, Reiter's Disease, Stiff-Man Syndrome, autoimmune pulmonary inflammation, autism, Guillain-Barre Syndrome, insulin dependent diabetes mellitus, and autoimmune inflammatory eye disorders.
[0585] Additional disorders that are likely to have an autoimmune component that may be treated, prevented, diagnosed and/or prognosed with the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to, scleroderma with anti-collagen antibodies (often characterized, e.g., by nucleolar and other nuclear antibodies), mixed connective tissue disease (often characterized, e.g., by antibodies to extractable nuclear antigens (e.g., ribonucleoprotein)), polymyositis (often characterized, e.g., by nonhistone ANA), pernicious anemia (often characterized, e.g., by antiparietal cell, microsomes, and intrinsic factor antibodies), idiopathic Addison's disease (often characterized, e.g., by humoral and cell-mediated adrenal cytotoxicity, infertility (often characterized, e.g., by antispermatozoal antibodies), glomerulonephritis (often characterized, e.g., by glomerular basement membrane antibodies or immune complexes), bullous pemphigoid (often characterized, e.g., by IgG
and complement in basement membrane), Sjogren's syndrome (often characterized, e.g., by multiple tissue antibodies, and/or a specific nonhistone ANA (SS-B)), diabetes mellitus (often characterized, e.g., by cell-mediated and humoral islet cell antibodies), and adrenergic drug resistance (including adrenergic drug resistance with asthma or cystic fibrosis) (often characterized, e.g., by beta-adrenergic receptor antibodies).
[0586] Additional disorders that may have an autoimmune component that may be treated, prevented, diagnosed and/or prognosed with the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to, chronic active hepatitis (often characterized, e.g., by smooth muscle antibodies), primary biliary cirrhosis (often characterized, e.g., by mitochondria antibodies), other endocrine gland failure (often characterized, e.g., by specific tissue antibodies in some cases), vitiligo (often characterized, e.g., by melanocyte antibodies), vasculitis (often characterized, e.g., by Ig and complement in vessel walls and/or low serum complement), post-MI (often characterized, e.g., by myocardial antibodies), cardiotomy syndrome (often characterized, e.g., by myocardial antibodies), urticaria (often characterized, e.g., by IgG and IgM antibodies to IgE), atopic dermatitis (often characterized, e.g., by IgG
and IgM antibodies to IgE), asthma (often characterized, e.g., by IgG and IgM antibodies to IgE), and many other inflammatory, granulomatous, degenerative, and atrophic disorders.
[0587] In a preferred embodiment, the autoimmune diseases and disorders and/or conditions associated with the diseases and disorders recited above are treated, prevented, diagnosed and/or prognosed using for example, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention. In a specific preferred embodiment, rheumatoid arthritis is treated, prevented, and/or diagnosed using fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention.
[0588] In another specific preferred embodiment, systemic lupus erythematosus is treated, prevented, and/or diagnosed using fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention. In another specific preferred embodiment, idiopathic thrombocytopenia purpura is treated, prevented, and/or diagnosed using fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention.
[0589] In another specific preferred embodiment IgA nephropathy is treated, prevented, and/or diagnosed using fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention.
[0590] In a preferred embodiment, the autoimmune diseases and disorders and/or conditions associated with the diseases and disorders recited above are treated, prevented, diagnosed and/or prognosed using fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention.
[0591] In preferred embodiments, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a immunosuppressive agent(s).
[0592] Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in treating, preventing, prognosing, and/or diagnosing diseases, disorders, and/or conditions of hematopoietic cells. Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention could be used to increase differentiation and proliferation of hematopoietic cells, including the pluripotent stem cells, in an effort to treat or prevent those diseases, disorders, and/or conditions associated with a decrease in certain (or many) types hematopoietic cells, including but not limited to, leukopenia, neutropenia, anemia, and thrombocytopenia. Alternatively, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention could be used to increase differentiation and proliferation of hematopoietic cells, including the pluripotent stem cells, in an effort to treat or prevent those diseases, disorders, and/or conditions associated with an increase in certain (or many) types of hematopoietic cells, including but not limited to, histiocytosis.
[0593] Allergic reactions and conditions, such as asthma (particularly allergic asthma) or other respiratory problems, may also be treated, prevented, diagnosed and/or prognosed using fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention. Moreover, these molecules can be used to treat, prevent, prognose, and/or diagnose anaphylaxis, hypersensitivity to an antigenic molecule, or blood group incompatibility.
[0594] Additionally, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, may be used to treat, prevent, diagnose and/or prognose IgE-mediated allergic reactions. Such allergic reactions include, but are not limited to, asthma, rhinitis, and eczema. In specific embodiments, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to modulate IgE concentrations in vitro or in vivo.
[0595] Moreover, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention have uses in the diagnosis, prognosis, prevention, and/or treatment of inflammatory conditions. For example, since fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may inhibit the activation, proliferation and/or differentiation of cells involved in an inflammatory response, these molecules can be used to prevent and/or treat chronic and acute inflammatory conditions. Such inflammatory conditions include, but are not limited to, for example, inflammation associated with infection (e.g., septic shock, sepsis, or systemic inflammatory response syndrome), ischemia-reperfusion injury, endotoxin lethality, complement-mediated hyperacute rejection, nephritis, cytokine or chemokine induced lung injury, inflammatory bowel disease, Crohn's disease, over production of cytokines (e.g., TNF or IL-1.), respiratory disorders (e.g., asthma and allergy); gastrointestinal disorders (e.g., inflammatory bowel disease); cancers (e.g., gastric, ovarian, lung, bladder, liver, and breast);
CNS disorders (e.g., multiple sclerosis; ischemic brain injury and/or stroke, traumatic brain injury, neurodegenerative disorders (e.g., Parkinson's disease and Alzheimer's disease); AIDS-related dementia; and prion disease); cardiovascular disorders (e.g., atherosclerosis, myocarditis, cardiovascular disease, and cardiopulmonary bypass complications); as well as many additional diseases, conditions, and disorders that are characterized by inflammation (e.g., hepatitis, rheumatoid arthritis, gout, trauma, pancreatitis, sarcoidosis, dermatitis, renal ischemia-reperfusion injury, Grave's disease, systemic lupus erythematosus, diabetes mellitus, and allogenic transplant rejection).
[0596] Because inflammation is a fundamental defense mechanism, inflammatory disorders can effect virtually any tissue of the body. Accordingly, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, have uses in the treatment of tissue-specific inflammatory disorders, including, but not limited to, adrenalitis, alveolitis, angiocholecystitis, appendicitis, balanitis, blepharitis, bronchitis, bursitis, carditis, cellulitis, cervicitis, cholecystitis, chorditis, cochlitis, colitis, conjunctivitis, cystitis, dermatitis, diverticulitis, encephalitis, endocarditis, esophagitis, eustachitis, fibrositis, folliculitis, gastritis, gastroenteritis, gingivitis, glossitis, hepatosplenitis, keratitis, labyrinthitis, laryngitis, lymphangitis, mastitis, media otitis, meningitis, metritis, mucitis, myocarditis, myosititis, myringitis, nephritis, neuritis, orchitis, osteochondritis, otitis, pericarditis, peritendonitis, peritonitis, pharyngitis, phlebitis, poliomyelitis, prostatitis, pulpitis, retinitis, rhinitis, salpingitis, scleritis, sclerochoroiditis, scrotitis, sinusitis, spondylitis, steatitis, stomatitis, synovitis, syringitis, tendonitis, tonsillitis, urethritis, and vaginitis.
[0597] In specific embodiments, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, are useful to diagnose, prognose, prevent, and/or treat organ transplant rejections and graft-versus-host disease.
Organ rejection occurs by host immune cell destruction of the transplanted tissue through an immune response. Similarly, an immune response is also involved in GVHD, but, in this case, the foreign transplanted immune cells destroy the host tissues.
Polypeptides, antibodies, or polynucleotides of the invention, and/or agonists or antagonists thereof, that inhibit an immune response, particularly the activation, proliferation, differentiation, or chemotaxis of T-cells, may be an effective therapy in preventing organ rejection or GVHD. In specific embodiments, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, that inhibit an immune response, particularly the activation, proliferation, differentiation, or chemotaxis of T-cells, may be an effective therapy in preventing experimental allergic and hyperacute xenograft rejection.
[0598] In other embodiments, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, are useful to diagnose, prognose, prevent, and/or treat immune complex diseases, including, but not limited to, serum sickness, post streptococcal glomerulonephritis, polyarteritis nodosa, and immune complex-induced vasculitis.
[0599] Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention can be used to treat, detect, and/or prevent infectious agents. For example, by increasing the immune response, particularly increasing the proliferation activation and/or differentiation of B and/or T cells, infectious diseases may be treated, detected, and/or prevented. The immune response may be increased by either enhancing an existing immune response, or by initiating a new immune response.

Alternatively, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may also directly inhibit the infectious agent (refer to section of application listing infectious agents, etc), without necessarily eliciting an immune response.
[0600] In another embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a vaccine adjuvant that enhances immune ,responsiveness to an antigen. In a specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an adjuvant to enhance tumor-specific immune responses.
[0601] In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an adjuvant to enhance anti-viral immune responses. Anti-viral immune responses that may be enhanced using the compositions of the invention as an adjuvant, include virus and virus associated diseases or symptoms described herein or otherwise known in the art. In specific embodiments, the compositions of the invention are used as an adjuvant to enhance an immune response to a virus, disease, or symptom selected from the group consisting of:
AIDS, meningitis, Dengue, EBV, and hepatitis (e.g., hepatitis B). In another specific embodiment, the compositions of the invention are used as an adjuvant to enhance an immune response to a virus, disease, or symptom selected from the group consisting of:
HIV/AIDS, respiratory syncytial virus, Dengue, rotavirus, Japanese B
encephalitis, influenza A and B, parainfluenza, measles, cytomegalovirus, rabies, Junin, Chilcungunya, Rift Valley Fever, herpes simplex, and yellow fever.
[0602] In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an adjuvant to enhance anti-bacterial or anti-fungal immune responses. Anti-bacterial or anti-fungal immune responses that may be enhanced using the compositions of the invention as an adjuvant, include bacteria or fungus and bacteria or fungus associated diseases or symptoms described herein or otherwise known in the art. In specific embodiments, the compositions of the invention are used as an adjuvant to enhance an immune response to a bacteria or fungus, disease, or symptom selected from the group consisting of: tetanus, Diphtheria, botulism, and meningitis type B.
[0603] In another specific embodiment, the compositions of the invention are used as an adjuvant to enhance an immune response to a bacteria or fungus, disease, or symptom selected from the group consisting of: Vibrio cholerae, Mycobacterium leprae, Salmonella typhi, Salmonella paratyphi, Meisseria meningitidis, Streptococcus pneumoniae, Group B
streptococcus, Shigella spp., Enterotoxigenic Escherichia coli, Enterohemorrhagic E. coli, and Borrelia burgdorferi.
[0604] In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an adjuvant to enhance anti-parasitic immune responses. Anti-parasitic immune responses that may be enhanced using the compositions of the invention as an adjuvant, include parasite and parasite associated diseases or symptoms described herein or otherwise known in the art. In specific embodiments, the compositions of the invention are used as an adjuvant to enhance an immune response to a parasite. In another specific embodiment, the compositions of the invention are used as an adjuvant to enhance an immune response to Plasmodium (malaria) or Leishmania.
[0605] In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may also be employed to treat infectious diseases including silicosis, sarcoidosis, and idiopathic pulmonary fibrosis; for example, by preventing the recruitment and activation of mononuclear phagocytes.
[0606] In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an antigen for the generation of antibodies to inhibit or enhance immune mediated responses against polypeptides of the invention.
[0607] In one embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are administered to an animal (e.g., mouse, rat, rabbit, hamster, guinea pig, pigs, micro-pig, chicken, camel, goat, horse, cow, sheep, dog, cat, non-human primate, and human, most preferably human) to boost the immune system to produce increased quantities of one or more antibodies (e.g., IgG, IgA, IgM, and IgE), to induce higher affinity antibody production and immunoglobulin class switching (e.g., IgG, IgA, IgM, and IgE), and/or to increase an immune response.
[0608] In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a stimulator of B cell responsiveness to pathogens.
[0609] In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an activator of T cells.
[0610] In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an agent that elevates the immune status of an individual prior to their receipt of immunosuppressive therapies.
[0611] In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an agent to induce higher affinity antibodies.
[0612] In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an agent to increase serum immunoglobulin concentrations.
[0613] In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an agent to accelerate recovery of immunocompromised individuals.
[0614] In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an agent to boost immunoresponsiveness among aged populations and/or neonates.
[0615] In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an immune system enhancer prior to, during, or after bone marrow transplant and/or other transplants (e.g., allogeneic or xenogeneic organ transplantation).
With respect to transplantation, compositions of the invention may be administered prior to, concomitant with, and/or after transplantation. In a specific embodiment, compositions of the invention are administered after transplantation, prior to the beginning of recovery of T-cell populations. In another specific embodiment, compositions of the invention are first administered after transplantation after the beginning of recovery of T cell populations, but prior to full recovery of B cell populations.
[0616] In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an agent to boost immunoresponsiveness among individuals having an acquired loss of B
cell function.
Conditions resulting in an acquired loss of B cell function that may be ameliorated or treated by administering the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, include, but are not limited to, HIV
Infection, AIDS, bone marrow transplant, and B cell chronic lymphocytic leukemia (CLL).
[0617] In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an agent to boost immunoresponsiveness among individuals having a temporary immune deficiency.

Conditions resulting in a temporary immune deficiency that may be ameliorated or treated by administering the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, include, but are not limited to, recovery from viral infections (e.g., influenza), conditions associated with malnutrition, recovery from infectious mononucleosis, or conditions associated with stress, recovery from measles, recovery from blood transfusion, and recovery from surgery.
[0618] In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a regulator of antigen presentation by monocytes, dendritic cells, and/or B-cells. In one embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention enhance antigen presentation or antagonize antigen presentation in vitro or in vivo. Moreover, in related embodiments, this enhancement or antagonism of antigen presentation may be useful as an anti-tumor treatment or to modulate the immune system.
[0619] In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an agent to direct an individual's immune system towards development of a humoral response (i.e.
TH2) as opposed to a TH1 cellular response.
[0620] In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a means to induce tumor proliferation and thus make it more susceptible to anti-neoplastic agents. For example, multiple myeloma is a slowly dividing disease and is thus refractory to virtually all anti-neoplastic regimens. If these cells were forced to proliferate more rapidly their susceptibility profile would likely change.
[0621] In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a stimulator of B cell production in pathologies such as AIDS, chronic lymphocyte disorder and/or Common Variable Immunodificiency.
[0622] In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a therapy for generation and/or regeneration of lymphoid tissues following surgery, trauma or genetic defect. In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used in the pretreatment of bone marrow samples prior to transplant.
[0623] In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a gene-based therapy for genetically inherited disorders resulting in immuno-incompetence/immunodeficiency such as observed among SCID patients.
[0624] In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a means of activating monocytes/macrophages to defend against parasitic diseases that effect monocytes such as Leishmania.
[0625] In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a means of regulating secreted cytokines that are elicited by polypeptides of the invention.
[0626] In another embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used in one or more of the applications decribed herein, as they may apply to veterinary medicine.
[0627] In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a means of blocking various aspects of immune responses to foreign agents or self Examples of diseases or conditions in which blocking of certain aspects of immune responses may be desired include autoimmune disorders such as lupus, and arthritis, as well as immunoresponsiveness to skin allergies, inflammation, bowel disease, injury and diseases/disorders associated with pathogens.
[0628] In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a therapy for preventing the B cell proliferation and Ig secretion associated with autoimmune diseases such as idiopathic thrombocytopenic purpura, systemic lupus erythematosus and multiple sclerosis.
[0629] In another specific embodiment, polypeptides, antibodies, polynucleotides and/or agonists or antagonists of the present fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention invention are used as a inhibitor of B and/or T cell migration in endothelial cells. This activity disrupts tissue architecture or cognate responses and is useful, for example in disrupting immune responses, and blocking sepsis.

[0630] In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a therapy for chronic hypergammaglobulinemia evident in such diseases as monoclonal gammopathy of undetermined significance (MGUS), Waldenstrom's disease, related idiopathic monoclonal gammopathies, and plasmacytomas.
[0631] In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be employed for instance to inhibit polypeptide chemotaxis and activation of macrophages and their precursors, and of neutrophils, basophils, B lymphocytes and some T-cell subsets, e.g., activated and CD8 cytotoxic T cells and natural killer cells, in certain autoimmune and chronic inflammatory and infective diseases. Examples of autoimmune diseases are described herein and include multiple sclerosis, and insulin-dependent diabetes.
[0632] The albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may also be employed to treat idiopathic hyper-eosinophilic syndrome by, for example, preventing eosinophil production and migration.
[0633] In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to enhance or inhibit complement mediated cell lysis.
[0634] In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to enhance or inhibit antibody dependent cellular cytotoxicity.
[0635] In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may also be employed for treating atherosclerosis, for example, by preventing monocyte infiltration in the artery wall.
[0636] In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be employed to treat adult respiratory distress syndrome (ARDS).
[0637] In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful for stimulating wound and tissue repair, stimulating angiogenesis, and/or stimulating the repair of vascular or lymphatic diseases or disorders. Additionally, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to stimulate the regeneration of mucosal surfaces.
[0638] In a specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to diagnose, prognose, treat, and/or prevent a disorder characterized by primary or acquired immunodeficiency, deficient serum immunoglobulin production, recurrent infections, and/or immune system dysfunction.
Moreover, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to treat or prevent infections of the joints, bones, skin, and/or parotid glands, blood-borne infections (e.g., sepsis, meningitis, septic arthritis, and/or osteomyelitis), autoimmune diseases (e.g., those disclosed herein), inflammatory disorders, and malignancies, and/or any disease or disorder or condition associated with these infections, diseases, disorders and/or malignancies) including, but not limited to, CVID, other primary immune deficiencies, HIV
disease, CLL, recurrent bronchitis, sinusitis, otitis media, conjunctivitis, pneumonia, hepatitis, meningitis, herpes zoster (e.g., severe herpes zoster), and/or pneumocystis carnii. Other diseases and disorders that may be prevented, diagnosed, prognosed, and/or treated with fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to, HIV infection, HTLV-BLV
infection, lymphopenia, phagocyte bactericidal dysfunction anemia, thrombocytopenia, and hemoglobinuria.
[0639] In another embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to treat, and/or diagnose an individual having common variable immunodeficiency disease ("CVID"; also known as "acquired agammaglobulinemia" and "acquired hypogammaglobulinemia") or a subset of this disease.
[0640] In a specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to diagnose, prognose, prevent, and/or treat cancers or neoplasms including immune cell or immune tissue-related cancers or neoplasms. Examples of cancers or neoplasms that may be prevented, diagnosed, or treated by fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to, acute myelogenous leukemia, chronic myelogenous leukemia, Hodgkin's disease, non-Hodgkin's lymphoma, acute lymphocytic anemia (ALL) Chronic lymphocyte leukemia, plasmacytomas, multiple myeloma, Burkitt's lymphoma, EBV-transformed diseases, and/or diseases and disorders described in the section entitled "Hyperproliferative Disorders"
elsewhere herein.
[0641] In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a therapy for decreasing cellular proliferation of Large B-cell Lymphomas.
[0642] In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a means of decreasing the involvement of B cells and Ig associated with Chronic Myelogenous Leukemia.
[0643] In specific embodiments, the compositions of the invention are used as an agent to boost immunoresponsiveness among B cell immunodeficient individuals, such as, for example, an individual who has undergone a partial or complete splenectomy.
Blood-Related Disorders [0644] The albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to modulate hemostatic (the stopping of bleeding) or thrombolytic (clot dissolving) activity. For example, by increasing hemostatic or thrombolytic activity, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention could be used to treat or prevent blood coagulation diseases, disorders, and/or conditions (e.g., afibrinogenemia, factor deficiencies, hemophilia), blood platelet diseases, disorders, and/or conditions (e.g., thrombocytopenia), or wounds resulting from trauma, surgery, or other causes. Alternatively, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention that can decrease hemostatic or thrombolytic activity could be used to inhibit or dissolve clotting.
These molecules could be important in the treatment or prevention of heart attacks (infarction), strokes, or scarring.
[0645] In specific embodiments, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to prevent, diagnose, prognose, and/or treat thrombosis, arterial thrombosis, venous thrombosis, thromboembolism, pulmonary embolism, atherosclerosis, myocardial infarction, transient ischemic attack, unstable angina. In specific embodiments, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used 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. Other uses for the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, include, but are not limited to, the prevention of occlusions in extrcorporeal devices (e.g., intravascular canulas, vascular access shunts in hemodialysis patients, hemodialysis machines, and cardiopulmonary bypass machines).
[0646] In another embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, may be used to prevent, diagnose, prognose, and/or treat diseases and disorders of the blood and/or blood forming organs associated with the tissue(s) in which the polypeptide of the invention is expressed.
[0647] The fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to modulate hematopoietic activity (the formation of blood cells). For example, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to increase the quantity of all or subsets of blood cells, such as, for example, erythrocytes, lymphocytes (B or T cells), myeloid cells (e.g., basophils, eosinophils, neutrophils, mast cells, macrophages) and platelets. The ability to decrease the quantity of blood cells or subsets of blood cells may be useful in the prevention, detection, diagnosis and/or treatment of anemias and leukopenias described below. Alternatively, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to decrease the quantity of all or subsets of blood cells, such as, for example, erythrocytes, lymphocytes (B or T cells), myeloid cells (e.g., basophils, eosinophils, neutrophils, mast cells, macrophages) and platelets.. The ability to decrease the quantity of blood cells or subsets of blood cells may be useful in the prevention, detection, diagnosis and/or treatment of leukocytoses, such as, for example eosinophilia.
[0648] The fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to prevent, treat, or diagnose blood dyscrasia.
[0649] Anemias are conditions in which the number of red blood cells or amount of hemoglobin (the protein that carries oxygen) in them is below normal. Anemia may be caused by excessive bleeding, decreased red blood cell production, or increased red blood cell destruction (hemolysis). The albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in treating, preventing, and/or diagnosing anemias. Anemias that may be treated prevented or diagnosed by the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include iron deficiency anemia, hypochromic anemia, microcytic anemia, chlorosis, hereditary siderob;astic anemia, idiopathic acquired sideroblastic anemia, red cell aplasia, megaloblastic anemia (e.g., pernicious anemia, (vitamin B12 deficiency) and folic acid deficiency anemia), aplastic anemia, hemolytic anemias (e.g., autoimmune helolytic anemia, microangiopathic hemolytic anemia, and paroxysmal nocturnal hemoglobinuria). The albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in treating, preventing, and/or diagnosing anemias associated with diseases including but not limited to, anemias associated with systemic lupus erythematosus, cancers, lymphomas, chronic renal disease, and enlarged spleens. The albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in treating, preventing, and/or diagnosing anemias arising from drug treatments such as anemias associated with methyldopa, dapsone, and/or sulfadrugs.
Additionally, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in treating, preventing, and/or diagnosing anemias associated with abnormal red blood cell architecture including but not limited to, hereditary spherocytosis, hereditary elliptocytosis, glucose-6-phosphate dehydrogenase deficiency, and sickle cell anemia.
[0650] The albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in treating, preventing, and/or diagnosing hemoglobin abnormalities, (e.g., those associated with sickle cell anemia, hemoglobin C disease, hemoglobin S-C disease, and hemoglobin E disease).
Additionally, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating thalassemias, including, but not limited to, major and minor forms of alpha-thalassemia and beta-thalassemia.
[0651] In another embodiment, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating bleeding disorders including, but not limited to, thrombocytopenia (e.g., idiopathic thrombocytopenic purpura, and thrombotic thrombocytopenic purpura), Von Willebrand's disease, hereditary platelet disorders (e.g., storage pool disease such as Chediak-Higashi and Hermansky-Pudlak syndromes, thromboxane A2 dysfunction, thromboasthenia, and Bernard-Soulier syndrome), hemolytic-uremic syndrome, hemophelias such as hemophelia A or Factor VII deficiency and Christmas disease or Factor IX deficiency, Hereditary Hemorhhagic Telangiectsia, also known as Rendu-Osler-Weber syndrome, allergic purpura (Henoch Schonlein purpura) and disseminated intravascular coagulation.
[0652] The effect of the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention on the clotting time of blood may be monitored using any of the clotting tests known in the art including, but not limited to, whole blood partial thromboplastin time (PTT), the activated partial thromboplastin time (aPTT), the activated clotting time (ACT), the recalcified activated clotting time, or the Lee-White Clotting time.
[0653] Several diseases and a variety of drugs can cause platelet dysfunction. Thus, in a specific embodiment, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating acquired platelet dysfunction such as platelet dysfunction accompanying kidney failure, leukemia, multiple myeloma, cirrhosis of the liver, and systemic lupus erythematosus as well as platelet dysfunction associated with drug treatments, including treatment with aspirin, ticlopidine, nonsteroidal anti-inflammatory drugs (used for arthritis, pain, and sprains), and penicillin in high doses.
[0654] In another embodiment, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating diseases and disorders characterized by or associated with increased or decreased numbers of white blood cells.
Leukopenia occurs when the number of white blood cells decreases below normal. Leukopenias include, but are not limited to, neutropenia and lymphocytopenia. An increase in the number of white blood cells compared to normal is known as leukocytosis. The body generates increased numbers of white blood cells during infection. Thus, leukocytosis may simply be a normal physiological parameter that reflects infection. Alternatively, leukocytosis may be an indicator of injury or other disease such as cancer. Leokocytoses, include but are not limited to, eosinophilia, and accumulations of macrophages. In specific embodiments, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating leukopenia. In other specific embodiments, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating leukocytosis.
[0655] Leukopenia may be a generalized decreased in all types of white blood cells, or may be a specific depletion of particular types of white blood cells. Thus, in specific embodiments, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating decreases in neutrophil numbers, known as neutropenia.
Neutropenias that may be diagnosed, prognosed, prevented, and/or treated by the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to, infantile genetic agranulocytosis, familial neutropenia, cyclic neutropenia, neutropenias resulting from or associated with dietary deficiencies (e.g., vitamin B 12 deficiency or folic acid deficiency), neutropenias resulting from or associated with drug treatments (e.g., antibiotic regimens such as penicillin treatment, sulfonamide treatment, anticoagulant treatment, anticonvulsant drugs, anti-thyroid drugs, and cancer chemotherapy), and neutropenias resulting from increased neutrophil destruction that may occur in association with some bacterial or viral infections, allergic disorders, autoimmune diseases, conditions in which an individual has an enlarged spleen (e.g., Felty syndrome, malaria and sarcoidosis), and some drug treatment regimens.
[0656] The albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating lymphocytopenias (decreased numbers of B and/or T
lymphocytes), including, but not limited to, lymphocytopenias resulting from or associated with stress, drug treatments (e.g., drug treatment with corticosteroids, cancer chemotherapies, and/or radiation therapies), AIDS infection and/or other diseases such as, for example, cancer, rheumatoid arthritis, systemic lupus erythematosus, chronic infections, some viral infections and/or hereditary disorders (e.g., DiGeorge syndrome, Wiskott-Aldrich Syndome, severe combined immunodeficiency, ataxia telangiectsia).
[0657] The albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating diseases and disorders associated with macrophage numbers and/or macrophage function including, but not limited to, Gaucher's disease, Niemann-Pick disease, Letterer-Siwe disease and Hand-Schuller-Christian disease.
[0658] In another embodiment, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating diseases and disorders associated with eosinophil numbers and/or eosinophil function including, but not limited to, idiopathic hypereosinophilic syndrome, eosinophilia-myalgia syndrome, and Hand-Schuller-Christian disease.
[0659] In yet another embodiment, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating leukemias and lymphomas including, but not limited to, acute lymphocytic (lymphpblastic) leukemia (ALL), acute myeloid (myelocytic, myelogenous, myeloblastic, or myelomonocytic) leukemia, chronic lymphocytic leukemia (e.g., B cell leukemias, T cell leukemias, Sezary syndrome, and Hairy cell leukenia), chronic myelocytic (myeloid, myelogenous, or granulocytic) leukemia, Hodgkin's lymphoma, non-hodgkin's lymphoma, Burkitt's lymphoma, and mycosis fungoides.
[0660] In other embodiments, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating diseases and disorders of plasma cells including, but not limited to, plasma cell dyscrasias, monoclonal gammaopathies, monoclonal gammopathies of undetermined significance, multiple myeloma, macroglobulinemia, Waldenstrom's macroglobulinemia, cryoglobulinemia, and Raynaud's phenomenon.
[0661] In other embodiments, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in treating, preventing, and/or diagnosing myeloproliferative disorders, including but not limited to, polycythemia vera, relative polycythemia, secondary polycythemia, myelofibrosis, acute myelofibrosis, agnogenic myelod metaplasia, thrombocythemia, (including both primary and seconday thrombocythemia) and chronic myelocytic leukemia.
[0662] In other embodiments, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful as a treatment prior to surgery, to increase blood cell production.
[0663] In other embodiments, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful as an agent to enhance the migration, phagocytosis, superoxide production, antibody dependent cellular cytotoxicity of neutrophils, eosionophils and macrophages.
[0664] In other embodiments, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful as an agent to increase the number of stem cells in circulation prior to stem cells pheresis. In another specific embodiment, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful as an agent to increase the number of stem cells in circulation prior to platelet pheresis.
[0665] In other embodiments, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful as an agent to increase cytokine production.
[0666] In other embodiments, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in preventing, diagnosing, and/or treating primary hematopoietic disorders.
Hyperproliferative Disorders [0667] In certain embodiments, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention can be used to treat or detect hyperproliferative disorders, including neoplasms. Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may inhibit the proliferation of the disorder through direct or indirect interactions. Alternatively, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may proliferate other cells which can inhibit the hyperproliferative disorder.
[0668] For example, by increasing an immune response, particularly increasing antigenic qualities of the hyperproliferative disorder or by proliferating, differentiating, or mobilizing T-cells, hyperproliferative disorders can be treated. This immune response may be increased by either enhancing an existing immune response, or by initiating a new immune response. Alternatively, decreasing an immune response may also be a method of treating hyperproliferative disorders, such as a chemotherapeutic agent.
[0669] Examples of hyperproliferative disorders that can be treated or detected by fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to neoplasms located in the: colon, abdomen, bone, breast, digestive system, liver, pancreas, peritoneum, endocrine glands (adrenal, parathyroid, pituitary, testicles, ovary, thymus, thyroid), eye, head and neck, nervous (central and peripheral), lymphatic system, pelvis, skin, soft tissue, spleen, thorax, and urogenital tract.

[0670] Similarly, other hyperproliferative disorders can also be treated or detected by fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention. Examples of such hyperproliferative disorders include, but are not limited to:
Acute Childhood Lymphoblastic Leukemia, Acute Lymphoblastic Leukemia, Acute Lymphocytic Leukemia, Acute Myeloid Leukemia, Adrenocortical Carcinoma, Adult (Primary) Hepatocellular Cancer, Adult (Primary) Liver Cancer, Adult Acute Lymphocytic Leukemia, Adult Acute Myeloid Leukemia, Adult Hodgkin's Disease, Adult Hodgkin's Lymphoma, Adult Lymphocytic Leukemia, Adult Non-Hodgkin's Lymphoma, Adult Primary Liver Cancer, Adult Soft Tissue Sarcoma, AIDS-Related Lymphoma, AIDS-Related Malignancies, Anal Cancer, Astrocytoma, Bile Duct Cancer, Bladder Cancer, Bone Cancer, Brain Stem Glioma, Brain Tumors, Breast Cancer, Cancer of the Renal Pelvis and Ureter, Central Nervous System (Primary) Lymphoma, Central Nervous System Lymphoma, Cerebellar Astrocytoma, Cerebral Astrocytoma, Cervical Cancer, Childhood (Primary) Hepatocellular Cancer, Childhood (Primary) Liver Cancer, Childhood Acute Lymphoblastic Leukemia, Childhood Acute Myeloid Leukemia, Childhood Brain Stem Glioma, Childhood Cerebellar Astrocytoma, Childhood Cerebral Astrocytoma, Childhood Extracranial Germ Cell Tumors, Childhood Hodgkin's Disease, Childhood Hodgkin's Lymphoma, Childhood Hypothalamic and Visual Pathway Glioma, Childhood Lymphoblastic Leukemia, Childhood Medulloblastoma, Childhood Non-Hodgkin's Lymphoma, Childhood Pineal and Supratentorial Primitive Neuroectodermal Tumors, Childhood Primary Liver Cancer, Childhood Rhabdomyosarcoma, Childhood Soft Tissue Sarcoma, Childhood Visual Pathway and Hypothalamic Glioma, Chronic Lymphocytic Leukemia, Chronic Myelogenous Leukemia, Colon Cancer, Cutaneous T-Cell Lymphoma, Endocrine Pancreas Islet Cell Carcinoma, Endometrial Cancer, Ependymoma, Epithelial Cancer, Esophageal Cancer, Ewing's Sarcoma and Related Tumors, Exocrine Pancreatic Cancer, Extracranial Germ Cell Tumor, Extragonadal Germ Cell Tumor, Extrahepatic Bile Duct Cancer, Eye Cancer, Female Breast Cancer, Gaucher's Disease, Gallbladder Cancer, Gastric Cancer, Gastrointestinal Carcinoid Tumor, Gastrointestinal Tumors, Germ Cell Tumors, Gestational Trophoblastic Tumor, Hairy Cell Leukemia, Head and Neck Cancer, Hepatocellular Cancer, Hodgkin's Disease, Hodgkin's Lymphoma, Hypergammaglobulinemia, Hypopharyngeal Cancer, Intestinal Cancers, Intraocular Melanoma, Islet Cell Carcinoma, Islet Cell Pancreatic Cancer, Kaposi's Sarcoma, Kidney Cancer, Laryngeal Cancer, Lip and Oral Cavity Cancer, Liver Cancer, Lung Cancer, Lymphoproliferative Disorders, Macroglobulinemia, Male Breast Cancer, Malignant Mesothelioma, Malignant Thymoma, Medulloblastoma, Melanoma, Mesothelioma, Metastatic Occult Primary Squamous Neck Cancer, Metastatic Primary Squamous Neck Cancer, Metastatic Squamous Neck Cancer, Multiple Myeloma, Multiple Myeloma/Plasma Cell Neoplasm, Myelodysplastic Syndrome, Myelogenous Leukemia, Myeloid Leukemia, Myeloproliferative Disorders, Nasal Cavity and Paranasal Sinus Cancer, Nasopharyngeal Cancer, Neuroblastoma, Non-Hodgkin's Lymphoma During Pregnancy, Nonmelanoma Skin Cancer, Non-Small Cell Lung Cancer, Occult Primary Metastatic Squamous Neck Cancer, Oropharyngeal Cancer, Osteo-/Malignant Fibrous Sarcoma, Osteosarcoma/Malignant Fibrous Histiocytoma, Osteosarcoma/Malignant Fibrous Histiocytoma of Bone, Ovarian Epithelial Cancer, Ovarian Germ Cell Tumor, Ovarian Low Malignant Potential Tumor, Pancreatic Cancer, Paraproteinemias, Purpura, Parathyroid Cancer, Penile Cancer, Pheochromocytoma, Pituitary Tumor, Plasma Cell Neoplasm/Multiple Myeloma, Primary Central Nervous System Lymphoma, Primary Liver Cancer, Prostate Cancer, Rectal Cancer, Renal Cell Cancer, Renal Pelvis and Ureter Cancer, Retinoblastoma, Rhabdomyosarcoma, Salivary Gland Cancer, Sarcoidosis Sarcomas, Sezary Syndrome, Skin Cancer, Small Cell Lung Cancer, Small Intestine Cancer, Soft Tissue Sarcoma, Squamous Neck Cancer, Stomach Cancer, Supratentorial Primitive Neuroectodermal and Pineal Tumors, T-Cell Lymphoma, Testicular Cancer, Thymoma, Thyroid Cancer, Transitional Cell Cancer of the Renal Pelvis and Ureter, Transitional Renal Pelvis and Ureter Cancer, Trophoblastic Tumors, Ureter and Renal Pelvis Cell Cancer, Urethral Cancer, Uterine Cancer, Uterine Sarcoma, Vaginal Cancer, Visual Pathway and Hypothalamic Glioma, Vulvar Cancer, Waldenstrom's Macroglobulinemia, Wilms' Tumor, and any other hyperproliferative disease, besides neoplasia, located in an organ system listed above.
[0671] In another preferred embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to diagnose, prognose, prevent, and/or treat premalignant conditions and to prevent progression to a neoplastic or malignant state, including but not limited to those disorders described above. Such uses are indicated in conditions known or suspected of preceding progression to neoplasia or cancer, in particular, where non-neoplastic cell growth consisting of hyperplasia, metaplasia, or most particularly, dysplasia has occurred (for review of such abnormal growth conditions, see Robbins and Angell, 1976, Basic Pathology, 2d Ed., W. B.
Saunders Co., Philadelphia, pp. 68-79.) [0672] Hyperplasia is a form of controlled cell proliferation, involving an increase in cell number in a tissue or organ, without significant alteration in structure or function.
Hyperplastic disorders which can be diagnosed, prognosed, prevented, and/or treated with fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to, angiofollicular mediastinal lymph node hyperplasia, angiolymphoid hyperplasia with eosinophilia, atypical melanocytic hyperplasia, basal cell hyperplasia, benign giant lymph node hyperplasia, cementum hyperplasia, congenital adrenal hyperplasia, congenital sebaceous hyperplasia, cystic hyperplasia, cystic hyperplasia of the breast, denture hyperplasia, ductal hyperplasia, endometrial hyperplasia, fibromuscular hyperplasia, focal epithelial hyperplasia, gingival hyperplasia, inflammatory fibrous hyperplasia, inflammatory papillary hyperplasia, intravascular papillary endothelial hyperplasia, nodular hyperplasia of prostate, nodular regenerative hyperplasia, pseudoepitheliomatous hyperplasia, senile sebaceous hyperplasia, and verrucous hyperplasia.
[0673]
Metaplasia is a form of controlled cell growth in which one type of adult or fully differentiated cell substitutes for another type of adult cell.
Metaplastic disorders which can be diagnosed, prognosed, prevented, and/or treated with fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to, agnogenic myeloid metaplasia, apocrine metaplasia, atypical metaplasia, autoparenchymatous metaplasia, connective tissue metaplasia, epithelial metaplasia, intestinal metaplasia, metaplastic anemia, metaplastic ossification, metaplastic polyps, myeloid metaplasia, primary myeloid metaplasia, secondary myeloid metaplasia, squamous metaplasia, squamous metaplasia of amnion, and symptomatic myeloid metaplasia.
[0674]
Dysplasia is frequently a forerunner of cancer, and is found mainly in the epithelia; it is the most disorderly form of non-neoplastic cell growth, involving a loss in individual cell uniformity and in the architectural orientation of cells.
Dysplastic cells often have abnormally large, deeply stained nuclei, and exhibit pleomorphism.
Dysplasia characteristically occurs where there exists chronic irritation or inflammation. Dysplastic disorders which can be diagnosed, prognosed, prevented, and/or treated with fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to, anhidrotic ectodermal dysplasia, anterofacial dysplasia, asphyxiating thoracic dysplasia, atriodigital dysplasia, bronchopulmonary dysplasia, cerebral dysplasia, cervical dysplasia, chondroectodermal dysplasia, cleidocranial dysplasia, congenital ectodermal dysplasia, craniodiaphysial dysplasia, craniocarpotarsal dysplasia, craniometaphysial dysplasia, dentin dysplasia, diaphysial dysplasia, ectodermal dysplasia, enamel dysplasia, encephalo-ophthalmic dysplasia, dysplasia epiphysialis hemimelia, dysplasia epiphysialis multiplex, dysplasia epiphysialis punctata, epithelial dysplasia, faciodigitogenital dysplasia, familial fibrous dysplasia of jaws, familial white folded dysplasia, fibromuscular dysplasia, fibrous dysplasia of bone, florid osseous dysplasia, hereditary renal-retinal dysplasia, hidrotic ectodermal dysplasia, hypohidrotic ectodermal dysplasia, lymphopenic thymic dysplasia, mammary dysplasia, mandibulofacial dysplasia, metaphysial dysplasia, Mondini dysplasia, monostotic fibrous dysplasia, mucoepithelial dysplasia, multiple epiphysial dysplasia, oculoauriculovertebral dysplasia, oculodentodigital dysplasia, oculovertebral dysplasia, odontogenic dysplasia, ophthalmomandibulomelic dysplasia, periapical cemental dysplasia, polyostotic fibrous dysplasia, pseudoachondroplastic spondyloepiphysial dysplasia, retinal dysplasia, septo-optic dysplasia, spondyloepiphysial dysplasia, and ventriculoradial dysplasia.
[0675] Additional pre-neoplastic disorders which can be diagnosed, prognosed, prevented, and/or treated with fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to, benign dysproliferative disorders (e.g., benign tumors, fibrocystic conditions, tissue hypertrophy, intestinal polyps, colon polyps, and esophageal dysplasia), leukoplakia, keratoses, Bowen's disease, Farmer's Skin, solar cheilitis, and solar keratosis.
[0676] In another embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, may be used to diagnose and/or prognose disorders associated with the tissue(s) in which the polypeptide of the invention is expressed.
[0677] In another embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention conjugated to a toxin or a radioactive isotope, as described herein, may be used to treat cancers and neoplasms, including, but not limited to, those described herein. In a further preferred embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention conjugated to a toxin or a radioactive isotope, as described herein, may be used to treat acute myelogenous leukemia.
[0678] Additionally, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may affect apoptosis, and therefore, would be useful in treating a number of diseases associated with increased cell survival or the inhibition of apoptosis. For example, diseases associated with increased cell survival or the inhibition of apoptosis that could be diagnosed, prognosed, prevented, and/or treated by polynucleotides, polypeptides, and/or agonists or antagonists of the invention, include cancers (such as follicular lymphomas, carcinomas with p53 mutations, and hormone-dependent tumors, including, but not limited to colon cancer, cardiac tumors, pancreatic cancer, melanoma, retinoblastoma, glioblastoma, lung cancer, intestinal cancer, testicular cancer, stomach cancer, neuroblastoma, myxoma, myoma, lymphoma, endothelioma, osteoblastoma, osteoclastoma, osteosarcoma, chondrosarcoma, adenoma, breast cancer, prostate cancer, Kaposi's sarcoma and ovarian cancer); autoimmune disorders such as, multiple sclerosis, Sjogren's syndrome, Hashimoto's thyroiditis, biliary cirrhosis, Behcet's disease, Crohn's disease, polymyositis, systemic lupus erythematosus and immune-related glomerulonephritis and rheumatoid arthritis) and viral infections (such as herpes viruses, pox viruses and adenoviruses), inflammation, graft v. host disease, acute graft rejection, and chronic graft rejection.
[0679] In preferred embodiments, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to inhibit growth, progression, and/or metastasis of cancers, in particular those listed above.
[0680]
Additional diseases or conditions associated with increased cell survival that could be diagnosed, prognosed, prevented, and/or treated by fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, include, but are not limited to, progression, and/or metastases of malignancies and related disorders such as leukemia (including acute leukemias (e.g., acute lymphocytic leukemia, acute myelocytic leukemia (including myeloblastic, promyelocytic, myelomonocytic, monocytic, and erytlu-oleukemia)) and chronic leukemias (e.g., chronic myelocytic (granulocytic) leukemia and chronic lymphocytic leukemia)), polycythemia vera, lymphomas (e.g., Hodgkin's disease and non-Hodgkin's disease), multiple myeloma, Waldenstrom's macroglobulinemia, heavy chain disease, and solid tumors including, but not limited to, sarcomas and carcinomas such as fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angio sarcoma, endothelio sarcoma, lymphangio sarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon carcinoma, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilm's tumor, cervical cancer, testicular tumor, lung carcinoma, small cell lung carcinoma, bladder carcinoma, epithelial carcinoma, glioma, astrocytoma, medulloblastoma, craniopharyngioma, ependymoma, pinealoma, emangioblastoma, acoustic neuroma, oligodendroglioma, menangioma, melanoma, neuroblastoma, and retinoblastoma.
[0681] Diseases associated with increased apoptosis that could be diagnosed, prognosed, prevented, and/or treated by fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, include AIDS;
neurodegenerative disorders (such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, retinitis pigmentosa, cerebellar degeneration and brain tumor or prior associated disease); autoimmune disorders (such as, multiple sclerosis, Sjogren's syndrome, Hashimoto's thyroiditis, biliary cirrhosis, Behcet's disease, Crohn's disease, polymyositis, systemic lupus erythematosus and immune-related glomerulonephritis and rheumatoid arthritis) myelodysplastic syndromes (such as aplastic anemia), graft v. host disease, ischemic injury (such as that caused by myocardial infarction, stroke and reperfusion injury), liver injury (e.g., hepatitis related liver injury, ischemia/reperfusion injury, cholestosis (bile duct injury) and liver cancer); toxin-induced liver disease (such as that caused by alcohol), septic shock, cachexia and anorexia.
[0682] Hyperproliferative diseases and/or disorders that could be diagnosed, prognosed, prevented, and/or treated by fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, include, but are not limited to, neoplasms located in the liver, abdomen, bone, breast, digestive system, pancreas, peritoneum, endocrine glands (adrenal, parathyroid, pituitary, testicles, ovary, thymus, thyroid), eye, head and neck, nervous system (central and peripheral), lymphatic system, pelvis, skin, soft tissue, spleen, thorax, and urogenital tract.
[0683] Similarly, other hyperproliferative disorders can also be diagnosed, prognosed, prevented, and/or treated by fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention. Examples of such hyperproliferative disorders include, but are not limited to: hypergarnmaglobulinemia, lymphoproliferative disorders, paraproteinemias, purpura, sarcoidosis, Sezary Syndrome, Waldenstron's macroglobulinemia, Gaucher's Disease, histiocytosis, and any other hyperproliferative disease, besides neoplasia, located in an organ system listed above.

[0684] Another preferred embodiment utilizes polynucleotides encoding albumin fusion proteins of the invention to inhibit aberrant cellular division, by gene therapy using the present invention, and/or protein fusions or fragments thereof.
[0685] Thus, the present invention provides a method for treating cell proliferative disorders by inserting into an abnormally proliferating cell a polynucleotide encoding an albumin fusion protein of the present invention, wherein said polynucleotide represses said expression.
[0686] Another embodiment of the present invention provides a method of treating cell-proliferative disorders in individuals comprising administration of one or more active gene copies of the present invention to an abnormally proliferating cell or cells. In a preferred embodiment, polynucleotides of the present invention is a DNA construct comprising a recombinant expression vector effective in expressing a DNA sequence encoding said polynucleotides. In another preferred embodiment of the present invention, the DNA
construct encoding the fusion protein of the present invention is inserted into cells to be treated utilizing a retrovirus, or more preferably an adenoviral vector (See G
J. Nabel, et. al., PNAS 1999 96: 324-326, which is hereby incorporated by reference). In a most preferred embodiment, the viral vector is defective and will not transform non-proliferating cells, only proliferating cells. Moreover, in a preferred embodiment, the polynucleotides of the present invention inserted into proliferating cells either alone, or in combination with or fused to other polynucleotides, can then be modulated via an external stimulus (i.e.
magnetic, specific small molecule, chemical, or drug administration, etc.), which acts upon the promoter upstream of said polynucleotides to induce expression of the encoded protein product. As such the beneficial therapeutic affect of the present invention may be expressly modulated (i.e. to increase, decrease, or inhibit expression of the present invention) based upon said external stimulus.
[0687] Polynucleotides of the present invention may be useful in repressing expression of oncogenic genes or antigens. By "repressing expression of the oncogenic genes " is intended the suppression of the transcription of the gene, the degradation of the gene transcript (pre-message RNA), the inhibition of splicing, the destruction of the messenger RNA, the prevention of the post-translational modifications of the protein, the destruction of the protein, or the inhibition of the normal function of the protein.
[0688] For local administration to abnormally proliferating cells, polynucleotides of the present invention may be administered by any method known to those of skill in the art DEMANDES OU BREVETS VOLUMINEUX
LA PRESENTE PARTIE DE CETTE DEMANDE OU CE BREVETS
COMPREND PLUS D'UN TOME.

NOTE. Pour les tomes additionels, veillez contacter le Bureau Canadien des Brevets.
JUMBO APPLICATIONS / PATENTS
THIS SECTION OF THE APPLICATION / PATENT CONTAINS MORE
THAN ONE VOLUME.

NOTE For additional volumes please contact the Canadian Patent Office.

Claims (2)

1. Use of a therapeutically effective amount of an albumin fusion protein for treatment of memory loss in a patient, said albumin fusion protein comprising the amino acid sequence selected from the group of:
a) amino acids 30 to 650 of SEQ ID NO:209;
b) amino acids 30 to 650 of SEQ ID NO:210;
c) amino acids 30 to 647 of SEQ ID NO:212;
d) amino acids 30 to 649 of SEQ ID NO:213;
e) amino acids 30 to 648 of SEQ ID NO:214;
f) amino acids 30 to 653 of SEQ ID NO:215;
g) amino acids 30 to 657 of SEQ ID NO:216;
h) amino acids 30 to 652 of SEQ ID NO:218;
i) amino acids 30 to 654 of SEQ ID NO:219;
j) amino acids 30 to 655 of SEQ ID NO:220;
k) amino acids 30 to 659 of SEQ ID NO:221;
l) amino acids 30 to 646 of SEQ ID NO:223;
m) amino acids 30 to 651 of SEQ ID NO:224; and n) amino acids 30 to 656 of SEQ ID NO:225.
2. Use of an albumin fusion protein for the preparation of a medicament for treatment of memory loss in a patient, said albumin fusion protein comprising the amino acid sequence selected from the group of:
a) amino acids 30 to 650 of SEQ ID NO:209;
b) amino acids 30 to 650 of SEQ ID NO:210;
c) amino acids 30 to 647 of SEQ ID NO:212;
d) amino acids 30 to 649 of SEQ ID NO:213;
e) amino acids 30 to 648 of SEQ ID NO:214;
f) amino acids 30 to 653 of SEQ ID NO:215;
g) amino acids 30 to 657 of SEQ ID NO:216;

h) amino acids 30 to 652 of SEQ ID NO:218;
i) amino acids 30 to 654 of SEQ ID NO:219;
j) amino acids 30 to 655 of SEQ ID NO:220;
k) amino acids 30 to 659 of SEQ ID NO:221;
l) amino acids 30 to 646 of SEQ ID NO:223;
m) amino acids 30 to 651 of SEQ ID NO:224; and n) amino acids 30 to 656 of SEQ ID NO:225.
CA2513213A 2003-01-22 2004-01-20 Albumin fusion proteins Expired - Fee Related CA2513213C (en)

Applications Claiming Priority (15)

Application Number Priority Date Filing Date Title
US44130503P 2003-01-22 2003-01-22
US60/441,305 2003-01-22
US45320103P 2003-03-11 2003-03-11
US60/453,201 2003-03-11
US46722203P 2003-05-02 2003-05-02
US60/467,222 2003-05-02
US47281603P 2003-05-23 2003-05-23
US60/472,816 2003-05-23
US47626703P 2003-06-06 2003-06-06
US60/476,267 2003-06-06
US50517203P 2003-09-24 2003-09-24
US60/505,172 2003-09-24
US50674603P 2003-09-30 2003-09-30
US60/506,746 2003-09-30
PCT/US2004/001369 WO2005003296A2 (en) 2003-01-22 2004-01-20 Albumin fusion proteins

Publications (2)

Publication Number Publication Date
CA2513213A1 CA2513213A1 (en) 2005-01-13
CA2513213C true CA2513213C (en) 2013-07-30

Family

ID=33569028

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2513213A Expired - Fee Related CA2513213C (en) 2003-01-22 2004-01-20 Albumin fusion proteins

Country Status (4)

Country Link
US (2) US7521424B2 (en)
EP (1) EP1594530A4 (en)
CA (1) CA2513213C (en)
WO (1) WO2005003296A2 (en)

Families Citing this family (82)

* 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
CA2405912A1 (en) 2000-04-12 2001-10-18 Human Genome Sciences, Inc. Albumin fusion proteins
AU2002364587A1 (en) 2001-12-21 2003-07-30 Human Genome Sciences, Inc. Albumin fusion proteins
KR101271635B1 (en) * 2001-12-21 2013-06-12 휴먼 게놈 사이언시즈, 인코포레이티드 Albumin fusion proteins
ES2545090T3 (en) 2001-12-21 2015-09-08 Human Genome Sciences, Inc. Albumin and GCSF fusion proteins
US20080194481A1 (en) * 2001-12-21 2008-08-14 Human Genome Sciences, Inc. Albumin Fusion Proteins
US20040248262A1 (en) 2003-01-22 2004-12-09 Koeberl Dwight D. Constructs for expressing lysomal polypeptides
US7077176B2 (en) * 2003-04-28 2006-07-18 Medical Instill Technologies, Inc. Container with valve assembly for filling and dispensing substances, and apparatus and method for filling
AU2005211725B2 (en) * 2004-02-09 2010-07-15 Human Genome Sciences, Inc. Albumin fusion proteins
US20060051859A1 (en) * 2004-09-09 2006-03-09 Yan Fu Long acting human interferon analogs
NZ555464A (en) 2004-12-02 2010-03-26 Domantis Ltd Bispecific domain antibodies targeting serum albumin and glp-1 or pyy
AU2006280312A1 (en) * 2005-08-12 2007-02-22 Human Genome Sciences, Inc. Albumin fusion proteins
PT1965823T (en) * 2005-11-04 2016-08-18 Glaxosmithkline Llc Methods for administering hypoglycemic agents
EP1816201A1 (en) 2006-02-06 2007-08-08 CSL Behring GmbH Modified coagulation factor VIIa with extended half-life
WO2007100535A2 (en) * 2006-02-22 2007-09-07 Merck & Co., Inc. Oxyntomodulin derivatives
MX2008013119A (en) 2006-04-11 2008-10-21 Novartis Ag Hcv/hiv inhibitors an their uses.
EP2013113B1 (en) * 2006-04-24 2018-03-21 Medical Instill Technologies, Inc. Needle penetrable and resealable lyophilization device and related method
WO2007146038A2 (en) 2006-06-07 2007-12-21 Human Genome Sciences, Inc. Albumin fusion proteins
WO2007144685A1 (en) 2006-06-13 2007-12-21 Commissariat A L'energie Atomique Cd4 mimic peptides and their uses
FR2904627B1 (en) * 2006-08-04 2008-11-07 Pasteur Institut NOVEL ACTIVE, PURIFIED AND ISOLATED PEPTIDES, CD4 RECEPTOR DERIVATIVES (MINI-CD4) AND THEIR PREPARATION PROCESS
WO2008021412A2 (en) * 2006-08-14 2008-02-21 Biorexis Pharmaceutical Corporation Interferon beta and transferrin fusion proteins
JO2945B1 (en) * 2006-09-13 2016-03-15 سميث كلاين بيتشام كوربوريشن Methods For Administering Long-lasting Hypoglycemic Agents.
WO2008094538A2 (en) 2007-01-30 2008-08-07 Epivax, Inc. Regulatory t cell epitopes, compositions and uses thereof
EA018471B1 (en) 2008-03-31 2013-08-30 Глаксо Груп Лимитед Drug fusions and conjugates
AU2009324037B2 (en) 2008-12-05 2015-07-30 Glaxo Group Limited Methods for selecting protease resistant polypeptides
EP2373681B1 (en) 2008-12-10 2017-01-18 Glaxosmithkline LLC Pharmaceutical compositions of albiglutide
SG172940A1 (en) 2009-01-16 2011-08-29 Teva Pharma New stable formulations of recombinant human albumin-human granulocyte colony stimulating factor fusion protein
JP5936112B2 (en) 2009-02-11 2016-06-15 アルブミディクス アクティーゼルスカブ Albumin variants and complexes
BRPI1013588A2 (en) 2009-03-27 2016-04-19 Glaxo Group Ltd composition
US8512690B2 (en) 2009-04-10 2013-08-20 Novartis Ag Derivatised proline containing peptide compounds as protease inhibitors
US20110182850A1 (en) 2009-04-10 2011-07-28 Trixi Brandl Organic compounds and their uses
WO2010126968A1 (en) * 2009-04-28 2010-11-04 Optmed, Inc. Compositions and methods for treating or preventing urolithiasis and conditions associated therewith
US8451450B2 (en) * 2009-09-14 2013-05-28 Bio-Rad Laboratories, Inc. Near real time optical phase conjugation
EA201290123A1 (en) 2009-09-30 2012-10-30 Глаксо Груп Лимитед PRODUCTS OF MERGERS AND CONJUGATES OF MEDICINES WITH INCREASED PERIOD OF SEMI-EXTRACT
BR112012010110A2 (en) 2009-10-30 2019-09-24 Boehringer Ingelheim Int hcv combination therapy dosage regimens comprising bi201335, interferon alfa and ribavirin
GB2488077A (en) 2009-10-30 2012-08-15 Novozymes Biopharma Dk As Albumin variants
EP2509616A4 (en) 2009-12-08 2013-05-29 Teva Pharma Bche albumin fusions for the treatment of cocaine abuse
CN106977608A (en) 2010-04-09 2017-07-25 阿尔布麦狄克斯公司 Albumin derivant and variant
EP2566502A4 (en) 2010-05-04 2013-10-09 Glaxosmithkline Llc Methods for treating or preventing cardiovascular disorders and providing cardiovascular protection
JP2013529627A (en) 2010-06-24 2013-07-22 パンメド リミテッド Treatment of hepatitis C virus-related disease using hydroxychloroquine or a combination of hydroxychloroquine and an antiviral agent
US20130143954A1 (en) * 2010-06-24 2013-06-06 Genomictree, Inc. Recombinant vector for suppressing proliferation of human papilloma virus cells including adenylate cyclase activating polypeptide 1 (pituitary) gene and pharmaceutical composition for treating human papilloma virus
US9011833B2 (en) 2010-10-08 2015-04-21 Novartis Ag Vitamin E formulations of sulfamide NS3 inhibitors
US9499605B2 (en) 2011-03-03 2016-11-22 Zymeworks Inc. Multivalent heteromultimer scaffold design and constructs
WO2012136790A1 (en) 2011-04-07 2012-10-11 Glaxo Group Limited Compositions comprising fusion proteins or conjugates with an improved half -life
WO2012136792A2 (en) 2011-04-07 2012-10-11 Glaxo Group Limited Cck compositions
CN110272484A (en) 2011-05-05 2019-09-24 阿尔布梅迪克斯医疗有限公司 Albumin variants
EP3434687B1 (en) * 2011-06-10 2021-03-10 Hanmi Science Co., Ltd. Novel oxyntomodulin derivatives and pharmaceutical composition for treating obesity comprising the same
RU2733544C2 (en) 2011-06-17 2020-10-05 Ханми Сайенс Ко., Лтд. Conjugate containing oxintomodulin and immunoglobulin fragment, and its application
WO2013075066A2 (en) 2011-11-18 2013-05-23 Eleven Biotherapeutics, Inc. Proteins with improved half-life and other properties
US9962430B2 (en) 2012-02-15 2018-05-08 Chirhoclin, Inc. Methods for treating pain associated with chronic pancreatitis
WO2013137869A1 (en) 2012-03-14 2013-09-19 Boehringer Ingelheim International Gmbh Combination therapy for treating hcv infection in an hcv-hiv coinfected patient population
CN104736559B (en) 2012-03-16 2022-04-08 阿尔布梅迪克斯医疗有限公司 Albumin variants
WO2013143581A1 (en) 2012-03-28 2013-10-03 Boehringer Ingelheim International Gmbh Combination therapy for treating hcv infection in specific patient subgenotype sub-population
AU2013261214A1 (en) 2012-05-16 2014-11-20 Glaxo Group Limited Polypeptide loaded POCA nanoparticles for oral administration
US11406718B2 (en) 2012-05-29 2022-08-09 Chirhoclin, Inc. Methods of detecting pancreobiliary ductal leaks
IN2015DN01115A (en) 2012-07-13 2015-06-26 Zymeworks Inc
KR101968344B1 (en) 2012-07-25 2019-04-12 한미약품 주식회사 A composition for treating hyperlipidemia comprising oxyntomodulin analog
ES2748158T3 (en) 2012-11-06 2020-03-13 Hanmi Pharm Ind Co Ltd Liquid formulation of protein conjugate comprising oxyntomodulin and an immunoglobulin fragment
KR101993393B1 (en) 2012-11-06 2019-10-01 한미약품 주식회사 A composition for treating diabetes or diabesity comprising oxyntomodulin analog
EP2917233A1 (en) 2012-11-08 2015-09-16 Novozymes Biopharma DK A/S Albumin variants
US20140205566A1 (en) 2012-11-30 2014-07-24 Novartis Ag Cyclic nucleuoside derivatives and uses thereof
EP3318124A3 (en) 2013-02-16 2018-05-30 Albumedix A/S Pharmacokinetic animal model
KR102111078B1 (en) 2013-07-04 2020-05-18 노파르티스 아게 O-mannosyltransferase deficient filamentous fungal cells and methods of use thereof
EP3035950A4 (en) * 2013-08-14 2017-05-03 The Arizona Board Of Regents On Behalf Of The University Of Arizona Glycosylated pacap/vip analogues with enhanced cns penetration for treatment of neurodegenerative diseases
WO2015173633A2 (en) 2014-05-02 2015-11-19 Cerenis Therapeutics Holding Sa Hdl therapy markers
CN108064266A (en) 2014-07-21 2018-05-22 格利科斯芬兰公司 The preparation of the glycoprotein with mammal sample N- glycan in filamentous fungi
TWI802396B (en) 2014-09-16 2023-05-11 南韓商韓美藥品股份有限公司 Use of a long acting glp-1/glucagon receptor dual agonist for the treatment of non-alcoholic fatty liver disease
CN105753990B (en) * 2014-12-19 2018-07-03 兰州大学 A kind of fusion protein of vasoactive intestinal peptide and its preparation method and application
KR102418477B1 (en) 2014-12-30 2022-07-08 한미약품 주식회사 Gluagon Derivatives
EP3337816B1 (en) 2015-08-20 2024-02-14 Albumedix Ltd Albumin variants and conjugates
EP3526242A1 (en) 2016-10-12 2019-08-21 University of Copenhagen Peptide dual agonists of gipr and glp2r
JP2020500306A (en) 2016-11-04 2020-01-09 オーフス ウニベルシテット Identification and treatment of tumors characterized by neonatal Fc receptor overexpression
WO2018089702A1 (en) * 2016-11-10 2018-05-17 Keros Therapeutics, Inc. Gdnf fusion polypeptides and methods of use thereof
US10767164B2 (en) 2017-03-30 2020-09-08 The Research Foundation For The State University Of New York Microenvironments for self-assembly of islet organoids from stem cells differentiation
CN108727486A (en) * 2017-04-24 2018-11-02 舒泰神(北京)生物制药股份有限公司 Long-acting nerve growth factor, preparation method and combinations thereof
CN110183536B (en) * 2018-02-22 2021-01-15 中国科学院遗传与发育生物学研究所 Composite stent material and application thereof
EP3553079A1 (en) 2018-04-12 2019-10-16 Bayer Aktiengesellschaft C-type natriuretic peptide engrafted antibodies
EP3553082A1 (en) 2018-04-12 2019-10-16 Bayer Aktiengesellschaft Brain natriuretic peptide engrafted antibodies
EP3553081A1 (en) 2018-04-12 2019-10-16 Bayer Aktiengesellschaft Atrial natriuretic peptide engrafted antibodies
WO2020102437A1 (en) * 2018-11-13 2020-05-22 Tocagen Inc. Recombinant vectors comprising genes for binding domains and secretable peptides
US11744878B2 (en) 2020-08-19 2023-09-05 Chirhoclin, Inc. Methods for treatment of COVID-19 syndrome
WO2024068848A1 (en) 2022-09-28 2024-04-04 Zealand Pharma A/S Methods for treating obesity

Family Cites Families (106)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5625041A (en) 1990-09-12 1997-04-29 Delta Biotechnology Limited Purification of proteins
DE2449885C3 (en) 1974-10-21 1980-04-30 Biotest-Serum-Institut Gmbh, 6000 Frankfurt Process for the production of chemically modified, long-life hemoglobin preparations as well as the modified hemoglobin preparation produced by this process
US4440859A (en) 1977-05-27 1984-04-03 The Regents Of The University Of California Method for producing recombinant bacterial plasmids containing the coding sequences of higher organisms
US4264731A (en) 1977-05-27 1981-04-28 The Regents Of The University Of California DNA Joining method
US4363877B1 (en) 1977-09-23 1998-05-26 Univ California Recombinant dna transfer vectors
US4283489A (en) 1977-09-23 1981-08-11 The Regents Of The University Of California Purification of nucleotide sequences suitable for expression in bacteria
US4407948A (en) 1977-09-23 1983-10-04 The Regents Of The University Of California Purification of nucleotide sequences suitable for expression in bacteria
US4366246A (en) 1977-11-08 1982-12-28 Genentech, Inc. Method for microbial polypeptide expression
US4652525A (en) 1978-04-19 1987-03-24 The Regents Of The University Of California Recombinant bacterial plasmids containing the coding sequences of insulin genes
US4447538A (en) 1978-04-19 1984-05-08 Regents Of The University Of California Microorganism containing gene for human chorionic somatomammotropin
US4342832A (en) 1979-07-05 1982-08-03 Genentech, Inc. Method of constructing a replicable cloning vehicle having quasi-synthetic genes
IL63916A0 (en) 1980-09-25 1981-12-31 Genentech Inc Microbial production of human fibroblast interferon
NZ199722A (en) 1981-02-25 1985-12-13 Genentech Inc Dna transfer vector for expression of exogenous polypeptide in yeast;transformed yeast strain
JPS57149228A (en) 1981-03-11 1982-09-14 Ajinomoto Co Inc Novel erythropoietin and its preparation
US4792602A (en) 1981-06-19 1988-12-20 Cornell Research Foundation, Inc. Adaptors, and synthesis and cloning of proinsulin genes
US4450103A (en) 1982-03-01 1984-05-22 Cetus Corporation Process for recovering human IFN-β from a transformed microorganism
US4775622A (en) 1982-03-08 1988-10-04 Genentech, Inc. Expression, processing and secretion of heterologous protein by yeast
US4778879A (en) 1982-04-20 1988-10-18 Sloan-Kettering Institute For Cancer Research Highly purified human interleukin 2 and method
US4925919A (en) 1984-04-25 1990-05-15 Roland Mertelsmann Purified interleukin 2
US4499188A (en) 1982-05-05 1985-02-12 Cetus Corporation Bacterial production of heterologous polypeptides under the control of a repressible promoter-operator
US4462940A (en) 1982-09-23 1984-07-31 Cetus Corporation Process for the recovery of human β-interferon-like polypeptides
US4840934A (en) 1983-01-25 1989-06-20 Eleanor Roosevelt Institute For Cancer Research, Inc. Therapeutic method using T cell growth factor
US4914026A (en) 1983-04-07 1990-04-03 Chiron Corporation Alpha factor leader sequence directed secretion of insulin
US5015575A (en) 1983-04-07 1991-05-14 Chiron Corporation Hybrid DNA synthesis of insulin
US5010003A (en) 1983-04-25 1991-04-23 Genentech, Inc. Use of yeast homologous signals to secrete heterologous proteins
CA1196863A (en) 1983-06-08 1985-11-19 Mattheus F.A. Goosen Slow release injectable insulin composition
NZ210501A (en) 1983-12-13 1991-08-27 Kirin Amgen Inc Erythropoietin produced by procaryotic or eucaryotic expression of an exogenous dna sequence
US4908433A (en) 1984-04-25 1990-03-13 Sloan-Kettering Institute For Cancer Research Uses of interleukin-2
US4908434A (en) 1984-04-25 1990-03-13 Sloan-Kettering Institute For Cancer Research Process for preparing purified interleukin-2
DK58285D0 (en) 1984-05-30 1985-02-08 Novo Industri As PEPTIDES AND MANUFACTURING AND USING THEREOF
US4959314A (en) 1984-11-09 1990-09-25 Cetus Corporation Cysteine-depleted muteins of biologically active proteins
US4970300A (en) 1985-02-01 1990-11-13 New York University Modified factor VIII
GB8504099D0 (en) 1985-02-18 1985-03-20 Wellcome Found Physiologically active substances
FR2579224B1 (en) 1985-03-25 1987-05-22 Genetica PROCESS FOR THE MICROBIOLOGICAL PREPARATION OF HUMAN SERUM-ALBUMIN
US4751180A (en) 1985-03-28 1988-06-14 Chiron Corporation Expression using fused genes providing for protein product
US5102872A (en) 1985-09-20 1992-04-07 Cetus Corporation Controlled-release formulations of interleukin-2
US5641663A (en) 1985-11-06 1997-06-24 Cangene Corporation Expression system for the secretion of bioactive human granulocyte macrophage colony stimulating factor (GM-CSF) and other heterologous proteins from steptomyces
FR2594846B1 (en) 1986-02-21 1989-10-20 Genetica PROCESS FOR THE PREPARATION OF MATURE HUMAN ALBUMIN SERUM
DK179286D0 (en) 1986-04-18 1986-04-18 Nordisk Gentofte INSULIN PREPARATION
US4765980A (en) 1986-04-28 1988-08-23 International Minerals & Chemical Corp. Stabilized porcine growth hormone
US5028422A (en) 1986-05-27 1991-07-02 Schering Corporation Treatment of basal cell carcinoma intralesionally with recombinant human alpha interferon
IT1196484B (en) 1986-07-11 1988-11-16 Sclavo Spa YEAST EXPRESSION AND SECRETION VECTOR, USEFUL FOR THE PREPARATION OF HETEROLOGICAL PROTEINS
US4801575A (en) 1986-07-30 1989-01-31 The Regents Of The University Of California Chimeric peptides for neuropeptide delivery through the blood-brain barrier
US5002764A (en) 1986-08-12 1991-03-26 Schering Corporation Treatment of actinic keratoses with alpha2 interferon
US4929442A (en) 1986-09-26 1990-05-29 Exovir, Inc. Compositions suitable for human topical application including a growth factor and/or related materials
US5508031A (en) 1986-11-21 1996-04-16 Cetus Oncology Corporation Method for treating biological damage using a free-radial scavenger and interleukin-2
US4835260A (en) 1987-03-20 1989-05-30 Genetics Institute, Inc. Erythropoietin composition
DE3888381T2 (en) 1987-04-09 1994-07-28 Delta Biotechnology Ltd Yeast vector.
US5336603A (en) 1987-10-02 1994-08-09 Genentech, Inc. CD4 adheson variants
GB8725529D0 (en) 1987-10-30 1987-12-02 Delta Biotechnology Ltd Polypeptides
JP2791418B2 (en) 1987-12-02 1998-08-27 株式会社ミドリ十字 Method for producing heterologous protein, recombinant DNA, transformant
JPH01240191A (en) 1988-02-16 1989-09-25 Green Cross Corp:The Novel signal peptide functioning by yeast and secretory manifestation of foreign protein using same
US5066489A (en) 1988-03-28 1991-11-19 Cetus Corporation Combination therapy of IL-2 and DTIC for the treatment of melanoma
US4999339A (en) 1988-03-28 1991-03-12 Cetus Corporation Combination therapy of IL-2 and DTIC for the treatment of melanoma
US5061488A (en) 1988-04-15 1991-10-29 The United States Of America As Represented Department Of Health & Human Services Flavone-8-acetic acid and interleukin-2 for cancer therapy
US5096707A (en) 1988-04-15 1992-03-17 The United States Of America As Represented By The Department Of Health And Human Services Flavone-8-acetic acid and interleukin-2 in a method of treating certain cancers
US5096885A (en) 1988-04-15 1992-03-17 Genentech, Inc. Human growth hormone formulation
US5126129A (en) 1988-05-23 1992-06-30 The Government Of The United States Of America As Represented By The Secretary Of The Department Of Health & Human Services Cancer therapy using interleukin-2 and flavone compounds
KR100195632B1 (en) 1988-07-23 1999-06-15 스티븐 조지 가랜드 New secretory leader sequences
US5260202A (en) 1988-09-07 1993-11-09 Delta Biotechnology Limited Fermentation method
US5256410A (en) 1988-12-01 1993-10-26 Schering Corporation Treatment of squamous cell carcinoma intralesionally with recombinant human alpha interferon
DK105489D0 (en) 1989-03-03 1989-03-03 Novo Nordisk As POLYPEPTIDE
US5116964A (en) 1989-02-23 1992-05-26 Genentech, Inc. Hybrid immunoglobulins
US5705363A (en) 1989-03-02 1998-01-06 The Women's Research Institute Recombinant production of human interferon τ polypeptides and nucleic acids
ES2034790T3 (en) 1989-04-11 1993-04-01 Boehringer Ingelheim International Gmbh USE OF AT LEAST ONE CITROQUINE FOR THE PREPARATION OF A MEDICINAL PRODUCT FOR THE SYSTEMATIC TREATMENT OF PRENEOPLASTIC INJURIES.
US5766883A (en) * 1989-04-29 1998-06-16 Delta Biotechnology Limited Polypeptides
EP0429586B1 (en) 1989-06-09 1995-08-02 Gropep Pty. Ltd. Growth hormone fusion proteins
DE3924746A1 (en) 1989-07-26 1991-01-31 Behringwerke Ag ERTHROPOIETIN (EPO) PEPTIDES AND ANTIBODIES THEREFOR
FR2650598B1 (en) * 1989-08-03 1994-06-03 Rhone Poulenc Sante DERIVATIVES OF ALBUMIN WITH THERAPEUTIC FUNCTION
US5073627A (en) 1989-08-22 1991-12-17 Immunex Corporation Fusion proteins comprising GM-CSF and IL-3
FR2653020B1 (en) 1989-10-17 1993-03-26 Roussel Uclaf USE OF A POLYPEPTIDE HAVING THE ACTIVITY OF HUMAN INTERLEUKIN 2 FOR THE PREPARATION OF PHARMACEUTICAL COMPOSITIONS FOR THE TREATMENT OF LEUKEMIA.
US5667986A (en) 1989-10-18 1997-09-16 Delta Biotechnology Limited Yeast promoter for expressing heterologous polypeptides
US5116944A (en) 1989-12-29 1992-05-26 Neorx Corporation Conjugates having improved characteristics for in vivo administration
US5545618A (en) 1990-01-24 1996-08-13 Buckley; Douglas I. GLP-1 analogs useful for diabetes treatment
US5208018A (en) 1990-03-19 1993-05-04 Brigham And Women's Hospital Treatment of cachexia with interleukin 2
FR2660863B1 (en) 1990-04-17 1994-01-21 Roussel Uclaf USE OF A POLYPEPTIDE HAVING THE ACTIVITY OF HUMAN INTERLEUKIN 2 FOR THE PREPARATION OF A PHARMACEUTICAL COMPOSITION FOR THE TREATMENT OF PRIMITIVE CANCERS OF THE POWDER.
GB9013017D0 (en) 1990-06-11 1990-08-01 Mars Uk Ltd Compounds
US5202239A (en) 1990-08-07 1993-04-13 Scios Nova Inc. Expression of recombinant polypeptides with improved purification
US5071872A (en) 1990-08-14 1991-12-10 The Ohio State University Research Foundation Method for improving interleukin-2 activity using aci-reductone compounds
FR2668368B1 (en) 1990-10-30 1995-03-10 Roussel Uclaf USE OF A POLYPEPTIDE HAVING THE ACTIVITY OF HUMAN INTERLEUKIN 2 FOR PREPARING A PHARMACEUTICAL COMPOSITION FOR THE TREATMENT OF MALIGNANT EPITHELIAL TUMORS.
US5272070A (en) 1991-03-08 1993-12-21 Board Of Regents, The University Of Texas System Method for the preparation of cell lines producing Man3 GlcNac 2 asparagine-linked gylcans and cell lines produced thereby
US5646012A (en) 1991-04-30 1997-07-08 Rhone-Poulenc Rorer S.A. Yeast promoter and use thereof
US5304473A (en) 1991-06-11 1994-04-19 Eli Lilly And Company A-C-B proinsulin, method of manufacturing and using same, and intermediates in insulin production
US5223408A (en) 1991-07-11 1993-06-29 Genentech, Inc. Method for making variant secreted proteins with altered properties
AU665599B2 (en) 1991-11-08 1996-01-11 Hemosol Inc. Hemoglobins as drug delivery agents
GB9200417D0 (en) 1992-01-09 1992-02-26 Bagshawe Kenneth D Cytotoxic drug therapy
FR2686620B1 (en) 1992-01-27 1995-06-23 Rhone Poulenc Rorer Sa HUMAN SERUM-ALBUMIN, PREPARATION AND USE.
FR2686900B1 (en) 1992-01-31 1995-07-21 Rhone Poulenc Rorer Sa NOVEL POLYPEPTIDES HAVING GRANULOCYTE COLONY STIMULATION ACTIVITY, THEIR PREPARATION AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM.
US5230886A (en) 1992-03-18 1993-07-27 Trustees Of Boston University Tumor cell suppression
US5460954A (en) 1992-04-01 1995-10-24 Cheil Foods & Chemicals, Inc. Production of human proinsulin using a novel vector system
US5229109A (en) 1992-04-14 1993-07-20 Board Of Regents, The University Of Texas System Low toxicity interleukin-2 analogues for use in immunotherapy
US5441734A (en) 1993-02-25 1995-08-15 Schering Corporation Metal-interferon-alpha crystals
US5521086A (en) 1993-09-16 1996-05-28 Cephalon, Inc. Secretion sequence for the production of a heterologous protein in yeast
US5459031A (en) 1993-11-05 1995-10-17 Amgen Inc. Methods for controlling sialic acid derivatives in recombinant glycoproteins
US5629286A (en) 1994-03-31 1997-05-13 Brewitt; Barbara Homeopathic dilutions of growth factors
US5646113A (en) 1994-04-07 1997-07-08 Genentech, Inc. Treatment of partial growth hormone insensitivity syndrome
US5639642A (en) 1994-06-16 1997-06-17 Novo Nordisk A/S Synthetic leader peptide sequences
US5574008A (en) 1994-08-30 1996-11-12 Eli Lilly And Company Biologically active fragments of glucagon-like insulinotropic peptide
US5512549A (en) 1994-10-18 1996-04-30 Eli Lilly And Company Glucagon-like insulinotropic peptide analogs, compositions, and methods of use
US5702717A (en) 1995-10-25 1997-12-30 Macromed, Inc. Thermosensitive biodegradable polymers based on poly(ether-ester)block copolymers
US20020048571A1 (en) * 1999-07-19 2002-04-25 Jeno Gyuris Chimeric polypeptides of serum albumin and uses related thereto
CA2405912A1 (en) * 2000-04-12 2001-10-18 Human Genome Sciences, Inc. Albumin fusion proteins
US20030143191A1 (en) * 2001-05-25 2003-07-31 Adam Bell Chemokine beta-1 fusion proteins
ES2545090T3 (en) * 2001-12-21 2015-09-08 Human Genome Sciences, Inc. Albumin and GCSF fusion proteins
AU2002364587A1 (en) * 2001-12-21 2003-07-30 Human Genome Sciences, Inc. Albumin fusion proteins
CA2485064A1 (en) * 2002-02-07 2003-08-14 Delta Biotechnology Limited Albumin-fused kunitz domain peptides

Also Published As

Publication number Publication date
EP1594530A2 (en) 2005-11-16
CA2513213A1 (en) 2005-01-13
WO2005003296A3 (en) 2005-04-21
US7521424B2 (en) 2009-04-21
US20100048472A1 (en) 2010-02-25
US20060014254A1 (en) 2006-01-19
WO2005003296A2 (en) 2005-01-13
EP1594530A4 (en) 2006-10-11

Similar Documents

Publication Publication Date Title
CA2513213C (en) Albumin fusion proteins
AU2005211725B2 (en) Albumin fusion proteins
AU2008319332B2 (en) Albumin fusion proteins
AU2007258609B2 (en) Albumin fusion proteins
EP1997829A1 (en) Albumin fusion proteins
US20100254944A1 (en) Albumin Fusion Proteins
US20080004206A1 (en) Albumin fusion proteins
CA2618476A1 (en) Albumin fusion proteins
CA2484556A1 (en) Albumin fusion proteins
CA2405550A1 (en) Albumin fusion proteins
CA2446739A1 (en) Chemokine beta-1 fusion proteins

Legal Events

Date Code Title Description
EEER Examination request
MKLA Lapsed

Effective date: 20180122